Next Article in Journal
Expression of the Novel Cardiac Biomarkers sST2, GDF-15, suPAR, and H-FABP in HFpEF Patients Compared to ICM, DCM, and Controls
Next Article in Special Issue
Pattern of Recurrence and Patient Survival after Perioperative Chemotherapy with 5-FU, Leucovorin, Oxaliplatin and Docetaxel (FLOT) for Locally Advanced Esophagogastric Adenocarcinoma in Patients Treated Outside Clinical Trials
Previous Article in Journal
Prognostic Value of Neutrophil-To-Lymphocyte Ratio and Platelet-To-Lymphocyte Ratio for Renal Outcomes in Patients with Rapidly Progressive Glomerulonephritis
Previous Article in Special Issue
Impact of Neoadjuvant Chemotherapy on the Outcomes of Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy for Colorectal Peritoneal Metastases: A Multi-Institutional Retrospective Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Neoadjuvant Therapy for Resectable and Borderline Resectable Pancreatic Cancer: A Meta-Analysis of Randomized Controlled Trials

1
Division of Surgical Oncology, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
2
Radiation Oncology, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
3
Internal Medicine, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
4
Biomedical Informatics, The Ohio State Wexner Medical Center, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Submission received: 10 March 2020 / Revised: 7 April 2020 / Accepted: 13 April 2020 / Published: 15 April 2020

Abstract

:
The efficacy of neoadjuvant therapy (NT) versus surgery first (SF) for pancreatic ductal adenocarcinoma (PDAC) remains controversial. A random-effects meta-analysis of only prospective randomized controlled trials (RCTs) comparing NT versus SF for potentially resectable (PR) or borderline resectable (BR) PDAC was performed. Among six RCTs including 850 patients, 411 (48.3%) received NT and 439 (51.6%) SF. In all included trials, NT was gemcitabine-based: four using chemoradiation and two chemotherapy alone. Based on an intention-to-treat analysis, NT resulted in improved overall survival (OS) compared to SF (HR 0.73, 95% CI 0.61–0.86). This effect was independent of anatomic classification (PR: hazard ratio (HR) 0.73, 95% CI 0.59–0.91; BR: HR 0.51 95% CI 0.28–0.93) or NT type (chemoradiation: HR 0.77, 95% CI 0.61–0.98; chemotherapy alone: HR 0.68, 95% CI 0.54–0.87). Overall resection rate was similar (risk ratio (RR) 0.93, 95% CI 0.82–1.04, I2 = 39.0%) but NT increased the likelihood of a margin-negative (R0) resection (RR 1.51, 95% CI 1.18–1.93, I2 = 0%) and having negative lymph nodes (RR 2.07, 95% CI 1.47–2.91, I2 = 12.3%). In this meta-analysis of prospective RCTs, NT significantly improved OS in an intention-to-treat fashion, compared with SF for localized PDAC. Randomized controlled trials using contemporary multi-agent chemotherapy will be needed to confirm these findings and to define the optimal NT regimen.

1. Introduction

Despite recent improvements, pancreatic ductal adenocarcinoma (PDAC) remains a deadly cancer with an overall survival (OS) rate at 5 years of only 10% [1]. Even patients with localized cancers who undergo resection are still likely to experience recurrence [2]. Adjuvant chemotherapy following surgery improves OS rates [3,4,5]. However, a significant proportion of patients are unable to initiate adjuvant chemotherapy following pancreatectomy, frequently because of postoperative complications or rapid disease recurrence [6]; many more are unable to finish the planned course of adjuvant therapy [7]. Indeed, a strategy of surgery first (SF) followed by planned adjuvant therapy has not resulted in significant improvements in OS during the past several decades, even at high-volume, experienced institutions [8,9].
In contrast, the administration of chemotherapy and/or radiation therapy prior to surgery guarantees the delivery of systemic therapies. In retrospective analyses, neoadjuvant therapy (NT) has been associated with improved rates of margin-negative resection and a decreased incidence of lymph node metastases [10,11]. NT also offers several other theoretical benefits, including early treatment of presumed micro-metastatic disease, enhanced selection of patients with appropriate tumor biology for surgery, and the ability to histologically measure the response to therapy [12,13]. Evidence of improved survival has also been suggested based on data from cancer databases [10], meta-analyses of retrospective studies [14], and Markov decision models [15]. In turn, NT has become the preferred approach for borderline resectable (BR) PDAC, while guidelines support the use of either SF or NT for potentially resectable (PR) PDAC [16,17,18].
Despite the theoretical and empirical advantages of NT, its use in the United States has remained relatively low [19,20], potentially driven by the lack of level I evidence for its efficacy. Until recently, only two small randomized controlled trials (RCTs) had been performed comparing SF to NT, and both were terminated early due to poor accrual [21,22]. Previous systematic reviews and meta-analyses that have purported a survival benefit with NT included non-randomized prospective and retrospective studies, which are limited due to their inherent selection biases [14,23,24,25,26]. As several larger RCTs have recently been completed, albeit with older neoadjuvant regimens, the purpose of the current study was to perform a meta-analysis limited to only RCTs evaluating SF vs. NT for PDAC.

2. Materials and Methods

2.1. Literature Search Strategy

The meta-analysis was performed according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [27,28]. A systematic review of the Pubmed, Cinahl, Medline, Embase, and CENTRAL databases was performed using the following Medical Subject Heading (MeSH) search terms: pancreatic cancer, neoadjuvant therapy, and randomized controlled trial. Multiple combinations of search terms were used. The final search was completed on 31 January 2020 and updated on 25 March 2020; the search strategy is reported in Table S1. Screening was performed using Covidence (Melbourne, Australia).
Studies including patients with PR or BR PDAC who were randomized to either SF or neoadjuvant chemotherapy and/or radiation therapy, and reported OS as a primary or secondary outcome, were included. Studies involving locally advanced (LA) PDAC were excluded. Retrospective and non-randomized prospective studies, and those without English-language abstracts, were excluded while published conference abstracts were not.
All titles, abstracts, and full texts were reviewed for inclusion according to the above criteria. Data extraction from the included studies was performed by two authors, and disagreements were resolved with discussion until consensus was achieved. Data on number of patients, resection rate, margin-negative (R0) resection rate, lymph node positivity rate, grade ≥3 complications, OS duration or hazard ratio, and type of adjuvant therapy were recorded for both treatment arms; type of NT, grade ≥3 serious adverse events (SAE) during NT, and completion rate were recorded for patients undergoing NT. All studies were assessed for risk of bias according to the Cochrane ROB-2 tool from the Cochrane Collaboration [29]. The quality of evidence for each outcome was estimated according to the Grading of Recommendations Assessment, Development and Evaluation (GRADE) process [30].

2.2. Statistical Analysis

The primary outcome was OS on an intention-to-treat basis, while secondary outcomes were overall resection rate (intention-to-treat) and R0 resection rate (among patients who underwent resection). Hazard ratios (HRs) and risk ratios (RRs) were used as effect size for each respective outcome. For the primary outcome, HRs were extracted from articles in which these data were provided. In instances where HRs were not reported, HRs and 95% confidence intervals (95% CIs) were estimated using indirect methods, based on observed events, total events, log-rank or Cox proportional model p-values and published Kaplan–Meier curves [31]. For the secondary outcome, RR and 95% CI were estimated based on event rates reported in 2 × 2 table formats.
RRs and HRs (95% CI) were log-transformed, and standard errors were computed based on log-transformed confidence interval bounds. Pooled logRR and logHR were then calculated using random-effects models via the restricted maximum likelihood estimator (REML). REML is approximately unbiased and efficient [32]. Median OS was estimated for each group via the following methodology [33]: log-transformed survival times (logMST) from each study were pooled in each treatment group, using weights derived from variances resulting from both sample size in each group and study precision of HRs. The standard error of the combined logMST was then calculated as the square root of the sum of the inverse variance weights.
Sensitivity analysis based on the random-effects model was done to examine effects of setting (PR vs. BR) and NT type (chemotherapy vs. chemoradiation (CRT)) on the primary outcomes by analyzing the relevant trials. HR < 1 for OS or RR > 1 for the secondary outcomes where the 95% CI does not contain 1 would favor NT. The amount of true heterogeneity in logRR or logHR was assessed using the I2 statistic. For ease of interpretation, estimated average logRR and logHR and 95% CIs were transformed back to RR and HR scales through exponentiation and results displayed using forest plots. All meta-analyses were done using R package (metafor).

3. Results

Among 1586 articles identified, 24 were selected for full-text review, and 6 RCTs comparing SF to NT for PDAC were included (Figure 1) [21,22,34,35,36,37]. The characteristics of the included trials are reported in Table 1. A total of 850 patients were included: 411 (48.3%) received NT and 439 (51.6%) SF. Of the six trials, four included patients with PR disease, one with BR tumors, and one included PR and BR tumors. Similarly, four trials used neoadjuvant CRT while two used systemic chemotherapy alone. All NT was gemcitabine-based; none utilized mFOLFIRINOX or gemcitabine/nab-paclitaxel. All included trials also recommended adjuvant therapy; five regimens were gemcitabine-based and one used S-1. Results of the methodological assessment are reported in Figure S1; none was assessed as having a high risk of bias.
The primary and secondary outcomes from the six included trials are summarized in Table 2. Based on an intention-to-treat analysis, the pooled HR for OS of NT compared to SF was 0.73 (95% CI 0.61–0.86, I2 = 0%) (Figure 2). Similarly, the pooled median OS across all studies was greater among patients receiving NT than SF (25.4 months (95% CI 22.4–28.7) vs. 19.4 (95% CI 17.2–21.8), p < 0.001). In pre-determined subset analyses, the pooled HR remained significantly in favor of NT independent of anatomic classification (PR: HR 0.73, 95% CI 0.59–0.91; BR: HR 0.51 95% CI 0.28–0.93) or neoadjuvant treatment type (CRT: HR 0.77, 95% CI 0.61–0.98; chemotherapy alone: HR 0.68, 95% CI 0.54–0.87).
Five trials reported adverse event rates during NT with SAE rates ranging from 11.1% to 72.0%. On an intention-to-treat basis, there was no significant difference in overall resection rates among the two groups (RR 0.93, 95% CI 0.82–1.04, I2 = 39.0%) (Figure 3A). On the other hand, among patients who underwent resection, NT increased the likelihood of an R0 resection (RR 1.51, 95% CI 1.18–1.93, I2 = 0%) (Figure 3B) and increased the pathologic lymph node negative rate (RR 2.07, 95% CI 1.47–2.91, I2 = 12.3%) (Figure 3C). Three studies reported grade ≥3 morbidity rates following surgery with rates ranging from 11.1% to 31.56% among patients who received NT and from 16.7% to 65.2% among those who underwent SF. The overall quality of evidence according to GRADE was felt to be moderate (Table S2).

4. Discussion

In this meta-analysis of only prospective RCTs, the OS of patients with resectable or borderline resectable PDAC who received NT was nearly 30% greater than that of patients who underwent immediate surgery using an intention-to-treat design. These findings remained significant among both patients with PR and BR disease and were independent of the type of NT. The current study also confirmed improvements in R0 resection rate and lymph node positivity with the use of NT and no difference in overall resection rate. Taken together, these results represent the strongest evidence to date for NT in the management of localized PDAC.
Immediate surgical resection followed by adjuvant chemotherapy has long been the standard treatment for PDAC, and recent trials suggest improving OS for those patients who are able to undergo resection and receive adjuvant therapy [4,5,38]. However, pancreatectomy is a complex, potentially morbid operation, and a significant proportion of patients are unable to receive adjuvant therapy either due to postoperative complications or poor performance status [6,7]. In addition, many patients develop rapid disease progression in the immediate postoperative period, suggesting that surgery was not only not beneficial for these patients, but potentially detrimental in delaying needed systemic therapy [39]. Administering non-surgical therapy prior to pancreatectomy would not only ensure its delivery and improve patient selection for surgery, but has also been theorized to improve complete microscopic tumor resection and locoregional control [12].
Early RCTs evaluating the role of NT for PDAC were unsuccessful. Both Golcher et al. and Casadei et al. intended to evaluate the use of preoperative CRT for patients with PR PDAC, but both trials were closed early due to poor patient accrual with no significant differences observed in OS [21,22]. Jang et al. performed a multicenter Korean RCT comparing neoadjuvant CRT to surgery first for BR PDAC. Based on the findings of improved OS and R0 resection rate, the trial was terminated early [37]. Similarly, Reni et al. performed a three-arm RCT comparing immediate surgery and two different adjuvant chemotherapy regimens to neoadjuvant chemotherapy followed by surgery and adjuvant therapy for patients with resectable PDAC. Based on the observed improved OS, the planned phase III component was not continued [34]. These first four RCTs highlight not only the challenges in designing, conducting, accruing, and completing trials in PDAC, but also the strong desire for novel treatment options with stronger evidence for this aggressive cancer. More recently, the PREOPANC trial randomized patients with PR and BR PDAC to either neoadjuvant gemcitabine-based CRT or SF; while the primary outcome OS was non-significant (p < 0.1), several of the secondary endpoints and predefined subgroup analyses were encouraging [36]. Finally, Prep-02/JSAP-05 was a large multicenter Japanese RCT comparing neoadjuvant gemcitabine and S-1 with SF in patients with resectable PDAC. Although presented in conference form only, NT led to significantly improved OS compared to SF (36.7 vs. 26.6 months; p=0.02) [35].
While recent RCTs and the current meta-analysis provide strong evidence to support the use of NT, many questions remain, particularly regarding the optimal components of therapy. Early trials primarily focused on preoperative CRT [40,41,42]; however, as the chemotherapy options for PDAC have improved, increasingly chemotherapy alone is being used prior to surgery, especially for patients with PR disease [43]. Nevertheless, the best chemotherapy regimen has not been established, and it is unlikely that the chemotherapy regimens used in the PACT-15 (Cisplatin, Epirubicin, Gemcitabine, Capecitabine) or Prep-02/JSAP-05 (gemcitabine, S-1) will be widely adopted [34,35]. Given that most patients currently receive either gemcitabine/nab-paclitaxel or mFOLFIRINOX, the ongoing NEONAX [44], NEPAFOX [45], NORPACT-1 [46], PANACHE01-PRODIGE48 [47], PREOPANC-2 [48], ESPAC-5 [49], and SWOG S1505 [50] trials as well as the upcoming Alliance A021806 trial, which incorporate one of these regimens, should improve our understanding of the preferred preoperative regimen.
In the era of more effective systemic chemotherapy, the role of CRT in the management of PDAC remains poorly understood. The four included RCTs and the pooled analysis in the current study support the use of neoadjuvant CRT compared to SF for patients with PR and BR PDAC. Compared to chemotherapy alone, retrospective studies have suggested that CRT is associated with improved margin-negative resection rates and locoregional control [51,52]. However, few prospective trials have compared chemotherapy alone to CRT, and thus the ongoing PREOPANC-2 and ESPAC-5 will be informative [48,49]. An important question remains regarding the role of neoadjuvant radiation therapy following induction systemic chemotherapy, particularly for patients with BR and LA disease [53]. The ongoing Alliance A021501 trial, comparing mFOLFORINOX with or without stereotactic body radiation therapy, may provide new insights [54].
As the prior literature on the efficacy of NT has largely been based on retrospective studies, many investigators have posited that any benefits observed when comparing NT and SF must occur due to the patient selection afforded by NT. However, the results of the current intention-to-treat meta-analysis (which include those patients who were unable to undergo curative-intent surgery) suggest that these findings may represent true treatment benefits. Despite the importance of these findings, several limitations should be acknowledged. First, the study included both PR and BR cancers, which have different anatomic characteristics and were not uniformly defined in the included trials. Second, the NT treatment types were heterogeneous and none included contemporary Western chemotherapy regimens more commonly used today. Third, one included trial has only been published in conference form, and our methodology was unable to use individual patient-level data [55]. Fourth, none of the randomized controlled trials included in this meta-analysis were blinded to the receipt of NT, potentially leading to a bias in the decision to proceed with resection in the neoadjuvant cohorts; since LN negative and R0 resection rates were only calculated among those who undergo resection, this could potentially confound these secondary outcomes. Finally, while minimal statistical heterogeneity was observed in the current meta-analysis, this could be an underestimate given the small number of included studies [56]; random-effects modeling was used regardless to account for possible heterogeneity.
In conclusion, in the first meta-analysis of NT versus SF for localized PDAC using only prospective RCTs, NT significantly improved OS in an intention-to-treat fashion. These results suggest that NT should be the preferred treatment strategy for patients with BR PDAC and can be considered for patients with PR PDAC. However, the included studies were largely based on outdated neoadjuvant regimens. Randomized controlled trials using contemporary multi-agent chemotherapy will be needed to confirm these findings and to define the optimal NT regimen.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/2077-0383/9/4/1129/s1. Figure S1: Risk of bias assessment according to Cochrane Collaboration ROB-2 tool, Table S1: Search terms used in systematic literature review, Table S2: Results of Cochrane Collaboration risk of bias assessment.

Author Contributions

Conceptualization, All Authors; Methodology, J.M.C., V.H., H.S.; Formal Analysis, J.M.C., V.H.; Writing—Original Draft Preparation, J.M.C.; Writing—Review and Editing, J.M.C., V.H., T.M.P., A.T., M.D., T.W., L.A., J.F.P.B., H.S.; Supervision, J.M.C., H.S.; Project Administration, J.M.C.; Funding Acquisition, J.M.C., H.S. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

The authors thank Sarah Hyman for assistance with literature review and Guy Brock for statistical advice.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Groot, V.P.; Rezaee, N.; Wu, W.; Cameron, J.L.; Fishman, E.K.; Hruban, R.H.; Weiss, M.J.; Zheng, L.; Wolfgang, C.L.; He, J. Patterns, Timing, and Predictors of Recurrence Following Pancreatectomy for Pancreatic Ductal Adenocarcinoma. Ann. Surg. 2017, 267, 936–945. [Google Scholar] [CrossRef] [PubMed]
  3. Neoptolemos, J.P.; Stocken, D.D.; Friess, H.; Bassi, C.; Dunn, J.A.; Hickey, H.; Beger, H.; Fernandez-Cruz, L.; Dervenis, C.; Lacaine, F.; et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N. Engl. J. Med. 2004, 350, 1200–1210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Oettle, H.; Post, S.; Neuhaus, P.; Gellert, K.; Langrehr, J.; Ridwelski, K.; Schramm, H.; Fahlke, J.; Zuelke, C.; Burkart, C.; et al. Adjuvant chemotherapy with gemcitabine vs. observation in patients undergoing curative-intent resection of pancreatic cancer: A randomized controlled trial. JAMA 2007, 297, 267–277. [Google Scholar] [CrossRef]
  5. Conroy, T.; Hammel, P.; Hebbar, M.; Ben Abdelghani, M.; Wei, A.C.; Raoul, J.-L.; Choné, L.; Francois, E.; Artru, P.; Biagi, J.J.; et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N. Engl. J. Med. 2018, 379, 2395–2406. [Google Scholar] [CrossRef]
  6. Bilimoria, K.Y.; Bentrem, D.J.; Ko, C.Y.; Tomlinson, J.S.; Stewart, A.K.; Winchester, D.P.; Talamonti, M.S. Multimodality therapy for pancreatic cancer in the U.S.: Utilization, outcomes, and the effect of hospital volume. Cancer 2007, 110, 1227–1234. [Google Scholar] [CrossRef]
  7. Altman, A.M.; Wirth, K.; Marmor, S.; Lou, E.; Chang, K.; Hui, J.Y.C.; Tuttle, T.M.; Jensen, E.H.; Denbo, J.W. Completion of Adjuvant Chemotherapy After Upfront Surgical Resection for Pancreatic Cancer Is Uncommon Yet Associated With Improved Survival. Ann. Surg. Oncol. 2019, 26, 4108–4116. [Google Scholar] [CrossRef]
  8. He, J.; Ahuja, N.; Makary, M.A.; Cameron, J.L.; Eckhauser, F.E.; Choti, M.A.; Hruban, R.H.; Pawlik, T.M.; Wolfgang, C.L. 2564 resected periampullary adenocarcinomas at a single institution: Trends over three decades. HPB 2014, 16, 83–90. [Google Scholar] [CrossRef] [Green Version]
  9. Winter, J.M.; Brennan, M.F.; Tang, L.H.; D’Angelica, M.I.; Dematteo, R.P.; Fong, Y.; Klimstra, D.S.; Jarnagin, W.R.; Allen, P.J. Survival after resection of pancreatic adenocarcinoma: Results from a single institution over three decades. Ann. Surg. Oncol. 2012, 19, 169–175. [Google Scholar] [CrossRef] [PubMed]
  10. Mokdad, A.A.; Minter, R.M.; Zhu, H.; Augustine, M.M.; Porembka, M.R.; Wang, S.C.; Yopp, A.C.; Mansour, J.C.; Choti, M.A.; Polanco, P.M. Neoadjuvant Therapy Followed by Resection Versus Upfront Resection for Resectable Pancreatic Cancer: A Propensity Score Matched Analysis. J. Clin. Oncol. 2016, 35, 515–522. [Google Scholar] [CrossRef] [PubMed]
  11. Sugimoto, M.; Takahashi, N.; Farnell, M.B.; Smyrk, T.C.; Truty, M.J.; Nagorney, D.M.; Smoot, R.L.; Chari, S.T.; Carter, R.E.; Kendrick, M.L. Survival benefit of neoadjuvant therapy in patients with non-metastatic pancreatic ductal adenocarcinoma: A propensity matching and intention-to-treat analysis. J. Surg. Oncol. 2019, 120, 976–984. [Google Scholar] [CrossRef] [PubMed]
  12. Cloyd, J.M.; Katz, M.H.G.; Prakash, L.; Varadhachary, G.R.; Wolff, R.A.; Shroff, R.T.; Javle, M.; Fogelman, D.; Overman, M.; Crane, C.H.; et al. Preoperative Therapy and Pancreatoduodenectomy for Pancreatic Ductal Adenocarcinoma: A 25-Year Single-Institution Experience. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract 2017, 21, 164–174. [Google Scholar] [CrossRef] [PubMed]
  13. Cloyd, J.M.; Wang, H.; Egger, M.E.; Tzeng, C.-W.D.; Prakash, L.R.; Maitra, A.; Varadhachary, G.R.; Shroff, R.; Javle, M.; Fogelman, D.; et al. Association of Clinical Factors With a Major Pathologic Response Following Preoperative Therapy for Pancreatic Ductal Adenocarcinoma. JAMA Surg. 2017, 152, 1048–1056. [Google Scholar] [CrossRef] [PubMed]
  14. Versteijne, E.; Vogel, J.A.; Besselink, M.G.; Busch, O.R.C.; Wilmink, J.W.; Daams, J.G.; van Eijck, C.H.J.; Groot Koerkamp, B.; Rasch, C.R.N.; van Tienhoven, G.; et al. Meta-analysis comparing upfront surgery with neoadjuvant treatment in patients with resectable or borderline resectable pancreatic cancer. Br. J. Surg. 2018, 105, 946–958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. De Geus, S.W.L.; Evans, D.B.; Bliss, L.A.; Eskander, M.F.; Smith, J.K.; Wolff, R.A.; Miksad, R.A.; Weinstein, M.C.; Tseng, J.F. Neoadjuvant therapy versus upfront surgical strategies in resectable pancreatic cancer: A Markov decision analysis. Eur. J. Surg. Oncol. J. Eur. Soc. Surg. Oncol. Br. Assoc. Surg. Oncol. 2016, 42, 1552–1560. [Google Scholar] [CrossRef]
  16. Khorana, A.A.; Mangu, P.B.; Berlin, J.; Engebretson, A.; Hong, T.S.; Maitra, A.; Mohile, S.G.; Mumber, M.; Schulick, R.; Shapiro, M.; et al. Potentially Curable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 34, 2541–2556. [Google Scholar] [CrossRef]
  17. Tempero, M.A.; Malafa, M.P.; Chiorean, E.G.; Czito, B.; Scaife, C.; Narang, A.K.; Fountzilas, C.; Wolpin, B.M.; Al-Hawary, M.; Asbun, H.; et al. Pancreatic Adenocarcinoma, Version 1.2019. J. Natl. Compr. Cancer Netw. JNCCN 2019, 17, 202–210. [Google Scholar] [CrossRef] [Green Version]
  18. Abrams, R.A.; Lowy, A.M.; O’Reilly, E.M.; Wolff, R.A.; Picozzi, V.J.; Pisters, P.W.T. Combined modality treatment of resectable and borderline resectable pancreas cancer: Expert consensus statement. Ann. Surg. Oncol. 2009, 16, 1751–1756. [Google Scholar] [CrossRef]
  19. Youngwirth, L.M.; Nussbaum, D.P.; Thomas, S.; Adam, M.A.; Blazer, D.G.; Roman, S.A.; Sosa, J.A. Nationwide trends and outcomes associated with neoadjuvant therapy in pancreatic cancer: An analysis of 18,243 patients. J. Surg. Oncol. 2017, 116, 127–132. [Google Scholar] [CrossRef]
  20. Hashmi, A.; Kozick, Z.; Fluck, M.; Hunsinger, M.A.; Wild, J.; Arora, T.K.; Shabahang, M.M.; Blansfield, J.A. Neoadjuvant versus Adjuvant Chemotherapy for Resectable Pancreatic Adenocarcinoma: A National Cancer Database Analysis. Am. Surg. 2018, 84, 1439–1445. [Google Scholar]
  21. Casadei, R.; Di Marco, M.; Ricci, C.; Santini, D.; Serra, C.; Calculli, L.; D’Ambra, M.; Guido, A.; Morselli-Labate, A.M.; Minni, F. Neoadjuvant Chemoradiotherapy and Surgery Versus Surgery Alone in Resectable Pancreatic Cancer: A Single-Center Prospective, Randomized, Controlled Trial Which Failed to Achieve Accrual Targets. J. Gastrointest. Surg. Off. J. Soc. Surg. Aliment. Tract. 2015, 19, 1802–1812. [Google Scholar] [CrossRef] [PubMed]
  22. Golcher, H.; Brunner, T.B.; Witzigmann, H.; Marti, L.; Bechstein, W.-O.; Bruns, C.; Jungnickel, H.; Schreiber, S.; Grabenbauer, G.G.; Meyer, T.; et al. Neoadjuvant chemoradiation therapy with gemcitabine/cisplatin and surgery versus immediate surgery in resectable pancreatic cancer: Results of the first prospective randomized phase II trial. Strahlenther. Onkol. Organ Dtsch. Röntgenges. Al. 2015, 191, 7–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Bradley, A.; Van Der Meer, R. Upfront Surgery versus Neoadjuvant Therapy for Resectable Pancreatic Cancer: Systematic Review and Bayesian Network Meta-analysis. Sci. Rep. 2019, 9, 4354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Unno, M.; Hata, T.; Motoi, F. Long-term outcome following neoadjuvant therapy for resectable and borderline resectable pancreatic cancer compared to upfront surgery: A meta-analysis of comparative studies by intention-to-treat analysis. Surg. Today 2019, 49, 295–299. [Google Scholar] [CrossRef] [PubMed]
  25. Ye, M.; Zhang, Q.; Chen, Y.; Fu, Q.; Li, X.; Bai, X.; Liang, T. Neoadjuvant chemotherapy for primary resectable pancreatic cancer: A systematic review and meta-analysis. HPB 2020. [Google Scholar] [CrossRef] [Green Version]
  26. Pan, L.; Fang, J.; Tong, C.; Chen, M.; Zhang, B.; Juengpanich, S.; Wang, Y.; Cai, X. Survival benefits of neoadjuvant chemo(radio)therapy versus surgery first in patients with resectable or borderline resectable pancreatic cancer: A systematic review and meta-analysis. World J. Surg. Oncol. 2019, 18, 1. [Google Scholar] [CrossRef]
  27. Liberati, A.; Altman, D.G.; Tetzlaff, J.; Mulrow, C.; Gøtzsche, P.C.; Ioannidis, J.P.A.; Clarke, M.; Devereaux, P.J.; Kleijnen, J.; Moher, D. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: Explanation and elaboration. PLoS Med. 2009, 6, e1000100. [Google Scholar] [CrossRef]
  28. Beller, E.M.; Glasziou, P.P.; Altman, D.G.; Hopewell, S.; Bastian, H.; Chalmers, I.; Gøtzsche, P.C.; Lasserson, T.; Tovey, D. PRISMA for Abstracts Group PRISMA for Abstracts: Reporting systematic reviews in journal and conference abstracts. PLoS Med. 2013, 10, e1001419. [Google Scholar] [CrossRef] [Green Version]
  29. Sterne, J.A.C.; Savović, J.; Page, M.J.; Elbers, R.G.; Blencowe, N.S.; Boutron, I.; Cates, C.J.; Cheng, H.-Y.; Corbett, M.S.; Eldridge, S.M.; et al. RoB 2: A revised tool for assessing risk of bias in randomised trials. BMJ 2019, 366, l4898. [Google Scholar] [CrossRef] [Green Version]
  30. Guyatt, G.; Oxman, A.D.; Akl, E.A.; Kunz, R.; Vist, G.; Brozek, J.; Norris, S.; Falck-Ytter, Y.; Glasziou, P.; DeBeer, H.; et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J. Clin. Epidemiol. 2011, 64, 383–394. [Google Scholar] [CrossRef]
  31. Tierney, J.F.; Stewart, L.A.; Ghersi, D.; Burdett, S.; Sydes, M.R. Practical methods for incorporating summary time-to-event data into meta-analysis. Trials 2007, 8, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Viechtbauer, W. Conducting Meta-Analyses in R with the metafor Package. J. Stat. Softw. 2010, 36. [Google Scholar] [CrossRef] [Green Version]
  33. Zang, J.; Xu, J.; Xiang, C.; Zou, S.; He, J. Statistical modeling and verification for the synthesis of median survival time in multilevel meta-analysis of survival data. J. Epidemiol. Res. 2015, 1, 25. [Google Scholar] [CrossRef]
  34. Reni, M.; Balzano, G.; Zanon, S.; Zerbi, A.; Rimassa, L.; Castoldi, R.; Pinelli, D.; Mosconi, S.; Doglioni, C.; Chiaravalli, M.; et al. Safety and efficacy of preoperative or postoperative chemotherapy for resectable pancreatic adenocarcinoma (PACT-15): A randomised, open-label, phase 2–3 trial. Lancet Gastroenterol. Hepatol. 2018, 3, 413–423. [Google Scholar] [CrossRef]
  35. Unno, M.; Motoi, F.; Matsuyama, Y.; Satoi, S.; Matsumoto, I.; Aosasa, S.; Shirakawa, H.; Wada, K.; Fujii, T.; Yoshitomi, H.; et al. Randomized phase II/III trial of neoadjuvant chemotherapy with gemcitabine and S-1 versus upfront surgery for resectable pancreatic cancer (Prep-02/JSAP-05). J. Clin. Oncol. 2019, 37, 189. [Google Scholar] [CrossRef]
  36. Versteijne, E.; Suker, M.; Groothuis, K.; Akkermans-Vogelaar, J.M.; Besselink, M.G.; Bonsing, B.A.; Buijsen, J.; Busch, O.R.; Creemers, G.-J.M.; van Dam, R.M.; et al. Preoperative Chemoradiotherapy Versus Immediate Surgery for Resectable and Borderline Resectable Pancreatic Cancer: Results of the Dutch Randomized Phase III PREOPANC Trial. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2020. [Google Scholar] [CrossRef]
  37. Jang, J.-Y.; Han, Y.; Lee, H.; Kim, S.-W.; Kwon, W.; Lee, K.-H.; Oh, D.-Y.; Chie, E.K.; Lee, J.M.; Heo, J.S.; et al. Oncological Benefits of Neoadjuvant Chemoradiation With Gemcitabine Versus Upfront Surgery in Patients With Borderline Resectable Pancreatic Cancer: A Prospective, Randomized, Open-label, Multicenter Phase 2/3 Trial. Ann. Surg. 2018, 268, 215–222. [Google Scholar] [CrossRef]
  38. Neoptolemos, J.P.; Palmer, D.H.; Ghaneh, P.; Psarelli, E.E.; Valle, J.W.; Halloran, C.M.; Faluyi, O.; O’Reilly, D.A.; Cunningham, D.; Wadsley, J.; et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): A multicentre, open-label, randomised, phase 3 trial. Lancet 2017, 389, 1011–1024. [Google Scholar] [CrossRef]
  39. Groot, V.P.; Gemenetzis, G.; Blair, A.B.; Rivero-Soto, R.J.; Yu, J.; Javed, A.A.; Burkhart, R.A.; Rinkes, I.H.M.B.; Molenaar, I.Q.; Cameron, J.L.; et al. Defining and Predicting Early Recurrence in 957 Patients With Resected Pancreatic Ductal Adenocarcinoma. Ann. Surg. 2019, 269, 1154–1162. [Google Scholar] [CrossRef]
  40. Evans, D.B.; Rich, T.A.; Byrd, D.R.; Cleary, K.R.; Connelly, J.H.; Levin, B.; Charnsangavej, C.; Fenoglio, C.J.; Ames, F.C. Preoperative chemoradiation and pancreaticoduodenectomy for adenocarcinoma of the pancreas. Arch. Surg. Chic. III 1960 1992, 127, 1335–1339. [Google Scholar] [CrossRef]
  41. Evans, D.B.; Varadhachary, G.R.; Crane, C.H.; Sun, C.C.; Lee, J.E.; Pisters, P.W.T.; Vauthey, J.-N.; Wang, H.; Cleary, K.R.; Staerkel, G.A.; et al. Preoperative gemcitabine-based chemoradiation for patients with resectable adenocarcinoma of the pancreatic head. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2008, 26, 3496–3502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Pisters, P.W.T.; Wolff, R.A.; Janjan, N.A.; Cleary, K.R.; Charnsangavej, C.; Crane, C.N.; Lenzi, R.; Vauthey, J.N.; Lee, J.E.; Abbruzzese, J.L.; et al. Preoperative paclitaxel and concurrent rapid-fractionation radiation for resectable pancreatic adenocarcinoma: Toxicities, histologic response rates, and event-free outcome. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2002, 20, 2537–2544. [Google Scholar] [CrossRef] [PubMed]
  43. Cloyd, J.; Shen, C.; Santry, H.; Bridges, J.; Dillhoff, M.; Ejaz, A.; Pawlik, T.; Tsung, A. Disparities in the Use of Neoadjuvant Therapy for Resectable Pancreatic Ductal Adenocarcinoma. J. Natl. Compr. Canc. Netw. 2020, 18, 1–8. [Google Scholar]
  44. Ettrich, T.J.; Berger, A.W.; Perkhofer, L.; Daum, S.; König, A.; Dickhut, A.; Wittel, U.; Wille, K.; Geissler, M.; Algül, H.; et al. Neoadjuvant plus adjuvant or only adjuvant nab-paclitaxel plus gemcitabine for resectable pancreatic cancer—The NEONAX trial (AIO-PAK-0313), a prospective, randomized, controlled, phase II study of the AIO pancreatic cancer group. BMC Cancer 2018, 18, 1298. [Google Scholar] [CrossRef]
  45. Hozaeel, W.; Pauligk, C.; Homann, N.; Luley, K.; Kraus, T.W.; Trojan, J.; Bechstein, W.O.; Grimm, K.; Heise, B.; Schmiegel, W.; et al. Randomized multicenter phase II/III study with adjuvant gemcitabine versus neoadjuvant/adjuvant FOLFIRINOX in resectable pancreatic cancer: The NEPAFOX trial. J. Clin. Oncol. 2015, 33, TPS4152. [Google Scholar] [CrossRef]
  46. Labori, K.J.; Lassen, K.; Hoem, D.; Grønbech, J.E.; Søreide, J.A.; Mortensen, K.; Smaaland, R.; Sorbye, H.; Verbeke, C.; Dueland, S. Neoadjuvant chemotherapy versus surgery first for resectable pancreatic cancer (Norwegian Pancreatic Cancer Trial—1 (NorPACT-1))—Study protocol for a national multicentre randomized controlled trial. BMC Surg. 2017, 17, 94. [Google Scholar] [CrossRef]
  47. Schwarz, L.; Vernerey, D.; Bachet, J.-B.; Tuech, J.-J.; Portales, F.; Michel, P.; Cunha, A.S. Resectable pancreatic adenocarcinoma neo-adjuvant FOLF(IRIN)OX-based chemotherapy—A multicenter, non-comparative, randomized, phase II trial (PANACHE01-PRODIGE48 study). BMC Cancer 2018, 18, 762. [Google Scholar] [CrossRef]
  48. Dutch Pancreatic Cancer Group PREOPANC-2—DPCG. Available online: http://www.dpcg.nl/projecten/preopanc-2.html (accessed on 18 February 2020).
  49. European Study Group for Pancreatic Cancer ESPAC-5F: European Study Group for Pancreatic Cancer—Trial 5F. Available online: http://www.isrctn.com/ISRCTN89500674 (accessed on 18 February 2020).
  50. Sohal, D.; McDonough, S.; Ahmad, S.A.; Gandhi, N.; Beg, M.S.; Wang-Gillam, A.; Wade, J.L.; Guthrie, K.A.; Lowy, A.M.; Philip, P.A.; et al. SWOG S1505: Initial findings on eligibility and neoadjuvant chemotherapy experience with mfolfirinox versus gemcitabine/nab-paclitaxel for resectable pancreatic adenocarcinoma. J. Clin. Oncol. 2019, 37, 4137. [Google Scholar] [CrossRef]
  51. Cloyd, J.M.; Chen, H.-C.; Wang, X.; Tzeng, C.-W.D.; Kim, M.P.; Aloia, T.A.; Vauthey, J.-N.; Lee, J.E.; Katz, M.H.G. Chemotherapy Versus Chemoradiation as Preoperative Therapy for Resectable Pancreatic Ductal Adenocarcinoma: A Propensity Score Adjusted Analysis. Pancreas 2019, 48, 216–222. [Google Scholar] [CrossRef]
  52. Cloyd, J.M.; Crane, C.H.; Koay, E.J.; Das, P.; Krishnan, S.; Prakash, L.; Snyder, R.A.; Varadhachary, G.R.; Wolff, R.A.; Javle, M.; et al. Impact of hypofractionated and standard fractionated chemoradiation before pancreatoduodenectomy for pancreatic ductal adenocarcinoma. Cancer 2016, 122, 2671–2679. [Google Scholar] [CrossRef] [Green Version]
  53. Katz, M.H.G.; Shi, Q.; Ahmad, S.A.; Herman, J.M.; Marsh Rde, W.; Collisson, E.; Schwartz, L.; Frankel, W.; Martin, R.; Conway, W.; et al. Preoperative Modified FOLFIRINOX Treatment Followed by Capecitabine-Based Chemoradiation for Borderline Resectable Pancreatic Cancer: Alliance for Clinical Trials in Oncology Trial A021101. JAMA Surg. 2016, 151, e161137. [Google Scholar] [PubMed] [Green Version]
  54. Katz, M.H.G.; Ou, F.-S.; Herman, J.M.; Ahmad, S.A.; Wolpin, B.; Marsh, R.; Behr, S.; Shi, Q.; Chuong, M.; Schwartz, L.H.; et al. Alliance for clinical trials in oncology (ALLIANCE) trial A021501: Preoperative extended chemotherapy vs. chemotherapy plus hypofractionated radiation therapy for borderline resectable adenocarcinoma of the head of the pancreas. BMC Cancer 2017, 17, 505. [Google Scholar] [CrossRef] [PubMed]
  55. Pignon, J.P.; Hill, C. Meta-analyses of randomised clinical trials in oncology. Lancet Oncol. 2001, 2, 475–482. [Google Scholar] [CrossRef]
  56. Kontopantelis, E.; Springate, D.A.; Reeves, D. A re-analysis of the Cochrane Library data: The dangers of unobserved heterogeneity in meta-analyses. PLoS ONE 2013, 8, e69930. [Google Scholar] [CrossRef]
Figure 1. Flow diagram of study selection using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.
Figure 1. Flow diagram of study selection using Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines.
Jcm 09 01129 g001
Figure 2. Forest plot of pooled hazard ratio for overall survival among patients with pancreatic cancer randomized to neoadjuvant therapy versus surgery first.
Figure 2. Forest plot of pooled hazard ratio for overall survival among patients with pancreatic cancer randomized to neoadjuvant therapy versus surgery first.
Jcm 09 01129 g002
Figure 3. Forest plots of pooled relative risk ratios for (A) overall resection rates, (B) R0 resection rates, and (C) lymph node negative rates among patients with pancreatic cancer randomized to neoadjuvant therapy versus surgery first.
Figure 3. Forest plots of pooled relative risk ratios for (A) overall resection rates, (B) R0 resection rates, and (C) lymph node negative rates among patients with pancreatic cancer randomized to neoadjuvant therapy versus surgery first.
Jcm 09 01129 g003
Table 1. Study characteristics for randomized controlled trials included in the meta-analysis.
Table 1. Study characteristics for randomized controlled trials included in the meta-analysis.
AuthorInstitutionOriginSettingPR/BR DefinitionSample SizeNeoadjuvant TherapyRegimenAdjuvant Therapy
GolcherMulti-GermanyPR≤180 “peripancreatic vessels”66CRTGemcitabine/Cisplatin; 56GyGemcitabine
CasadeiSingle-ItalyPR<180 SMV/PV; No contact to CA/HA/SMA 38CRTGemcitabine; 54GyGemcitabine
JangMulti-KoreaBR2012 NCCN criteria50CRTGemcitabine; 54GyCRT, Gemcitabine
ReniMulti-ItalyPRNo invasion of SMA/SMV/PV/CA/HA88ChemoCisplatin, Epirubicin, Gemcitabine, CapecitabineCisplatin, Epirubicin, Gemcitabine, Capecitabine or Gemcitabine
VersteijneMulti-NetherlandsPR/BRPR: <90 SMV/PV; no CA/HA/SMA contact
BR: <90 CA/HA/SMA; 90–270 PV/SMV without occlusion
246CRTGemcitabine; 36GyGemcitabine
UnnoMulti-JapanPRNo CA/HA/SMA abutment362ChemoGemcitabine, S-1S1
PR, potentially resectable; BR, borderline resectable; CRT, chemoradiation; SMV, superior mesenteric vein; PV, portal vein; CA, celiac axis; HA, hepatic artery; SMA, superior mesenteric artery; NCCN, National Comprehensive Cancer Network.
Table 2. Outcomes of randomized controlled trials included in the meta-analysis.
Table 2. Outcomes of randomized controlled trials included in the meta-analysis.
AuthorSAE Rate (%)Resected Rate (%)R0 Resection Rate (%)pN0 Rate (%)Grade ≥3 Postoperative Morbidity (%)Overall Survival (mo)
NTNTSFNTSFNTSFNTSFNTSF
Golcher 45.557.669.752.647.868.443.531.5665.217.414.4
Casadei 22.261.175.063.633.345.513.3N/AN/A22.419.5
Jang 11.163.078.382.433.370.616.723.516.721.012.0
Reni 34.484.487.563.032.748.126.511.120.438.220.4–26.4
VersteijneN/A60.572.470.840.266.721.768.1 *50.0 *16.014.3
Unno72.076.972.2N/AN/AN/AN/AN/AN/A36.726.6
SAE, serious adverse events; NT, neoadjuvant therapy; SF, surgery first; N/A, not applicable; * Any complication.

Share and Cite

MDPI and ACS Style

Cloyd, J.M.; Heh, V.; Pawlik, T.M.; Ejaz, A.; Dillhoff, M.; Tsung, A.; Williams, T.; Abushahin, L.; Bridges, J.F.P.; Santry, H. Neoadjuvant Therapy for Resectable and Borderline Resectable Pancreatic Cancer: A Meta-Analysis of Randomized Controlled Trials. J. Clin. Med. 2020, 9, 1129. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm9041129

AMA Style

Cloyd JM, Heh V, Pawlik TM, Ejaz A, Dillhoff M, Tsung A, Williams T, Abushahin L, Bridges JFP, Santry H. Neoadjuvant Therapy for Resectable and Borderline Resectable Pancreatic Cancer: A Meta-Analysis of Randomized Controlled Trials. Journal of Clinical Medicine. 2020; 9(4):1129. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm9041129

Chicago/Turabian Style

Cloyd, Jordan M., Victor Heh, Timothy M. Pawlik, Aslam Ejaz, Mary Dillhoff, Allan Tsung, Terence Williams, Laith Abushahin, John F. P. Bridges, and Heena Santry. 2020. "Neoadjuvant Therapy for Resectable and Borderline Resectable Pancreatic Cancer: A Meta-Analysis of Randomized Controlled Trials" Journal of Clinical Medicine 9, no. 4: 1129. https://0-doi-org.brum.beds.ac.uk/10.3390/jcm9041129

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop