Role of neoadjuvant therapy in resectable pancreatic carcinoma: a review article
Review Article

Role of neoadjuvant therapy in resectable pancreatic carcinoma: a review article

Sandeep Purohit ORCID logo, Parveen Ahlawat, Sarthak Tandon, Akash Raghavan Bellige, Munish Gairola

Department of Radiation Oncology, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India

Contributions: (I) Conception and design: S Purohit; (II) Administrative support: None; (III) Provision of study materials or patients: S Purohit, P Ahlawat, S Tandon; (IV) Collection and assembly of data: S Purohit, P Ahlawat, S Tandon; (V) Data analysis and interpretation: S Purohit, P Ahlawat, S Tandon; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Sandeep Purohit, MD. Department of Radiation Oncology, Rajiv Gandhi Cancer Institute and Research Centre, Rohini, Sector – 5, New Delhi, India. Email: purohit_sandeep@yahoo.com.

Abstract: Pancreatic carcinoma is a disease commonly diagnosed at an advanced stage and is associated with a poor five-year survival rate of around 9%. The treatment for pancreatic carcinoma is based on imaging that helps in staging and predicting its resectability. Surgery followed by adjuvant chemotherapy is the current treatment for resectable pancreatic cancer. The benefit of this treatment sequence for patients with resectable disease remains elusive as upto 80% of patients develop recurrence after surgery, primarily metastatic. The purpose of neoadjuvant therapy in the form of chemotherapy or radiation therapy is to downsize the tumor volume, reduce tumor extension from nearby structures, and reduce the chances of early metastasis. The main concern with neoadjuvant therapy is the risk of disease progression during chemotherapy and failure to undergo the intended curative surgery. Although the benefit of neoadjuvant therapy has been validated in non-metastatic unresectable or borderline resectable pancreatic cancer, its role in resectable disease is still lacking robust data. We have reviewed all recent literature on resectable pancreatic carcinoma including meta-analyses and randomized controlled trials to summarize the role of the neoadjuvant treatment in these patients. We have also discussed obstacles associated with neoadjuvant therapy in resectable pancreatic cancer patients and possible solutions to overcome these challenges.

Keywords: Pancreatic carcinoma; neoadjuvant therapy (NAT); resectable; chemotherapy; chemo-radiation


Received: 10 October 2023; Accepted: 05 March 2024; Published online: 20 May 2024.

doi: 10.21037/apc-23-14


Introduction

Pancreatic carcinoma is a common gastrointestinal malignancy that accounts for nearly 3% of all malignancies (1). The incidence of pancreatic carcinoma is rising and is currently the seventh leading cause of cancer-related death worldwide (2). It is more common in men than women (3). Around 90% of patients with pancreatic carcinoma are adenocarcinoma. Other subtypes include acinar carcinoma, pancreatic blastoma, or neuroendocrine tumor (4). Diagnosis and intervention in the early stages of pancreatic cancer are difficult due to non-specific symptoms such as nausea, bloating, fatigue, and loss of appetite (5). Endoscopy is performed to give pathological evidence and relieve obstruction in a jaundiced person by biliary stenting. A triple-phase computed tomography (CT) scan is required to evaluate pancreatic tumor resectability. National Comprehensive Cancer Network (NCCN) recognizes resectable pancreatic carcinoma (RPC) as tumors that do not show any contact with arteries (celiac axis, superior mesenteric artery, or common hepatic artery) or contact with ≤180° of the venous circumference (the superior mesenteric vein or portal vein) without any contour irregularity (6).

At diagnosis, around 10–20% are found to be RPC (7). It has been seen that 20–25% of RPC patients who had an exploratory laparotomy or laparoscopy cannot undergo the desired tumor resection (8,9). It could be due to inter-observer variation in determining resectability based on the CT scan. Around 50% of these RPC patients have a positive margin on resection, as seen in many randomized trials. It was also observed that the diffusion sequences in magnetic resonance image (MRI) of the abdomen in RPC helped find hidden liver metastases or omental deposits, sparing needless surgeries in 12% of cases (10). A positron emission tomography (PET) CT scan helps rule out metastasis in RPC patients with high-risk features such as high carbohydrate antigen 19.9 (CA 19.9), large tumor size, regional lymphadenopathy, excessive weight loss, and pain (11).

The prognosis of RPC can be dismal. This is due to radiographically occult infiltration of nearby vascular structures and premature metastasis in the early stages of the disease (12-14). Also, the effect of margin-positive RPC patients can worsen the prognosis. The addition of adjuvant chemotherapy has improved five-year survival in pancreatic cancer from 6% to 43% (15-20). The role of neoadjuvant therapy (NAT) in RPC is one of the recent topics in pancreatic cancer after trials have shown better pathological and survival outcomes in unresectable and borderline resectable pancreatic cancer (BRPC). Many randomized studies such as PRODIGE 24, ESPAC 4, and RTOG 9704 have shown worse prognosis in R1 resection RPC. The NCCN 2022 guidelines recommend NAT in high-risk RPC based on low-level evidence that shows a survival benefit. Well-structured and executed large studies are required to emphasize the advantage of NAT in RPC over upfront surgery.


Rationale for NAT

Pancreatic cancer is considered a systemic disease owing to its early metastatic potential (21). Like other systemic diseases, the outcome benefits might be seen with NAT in RPC due to a few theoretical advantages. These include:

  • Eradication of occult micro-metastasis and reducing distant relapses.
  • Selection of chemo-sensitive patients to increase chances of resection.
  • Identification of highly aggressive tumors that might not benefit from surgery.
  • Increase tolerance to chemotherapy by giving a chemotherapy-free interval during surgery.
  • To mitigate high-risk factors for improved treatment outcomes.

Limitations of NAT

The main limitation is the potential risk of metastasis or progression in an RPC patient while on NAT (22). Such tumors might respond to different types of chemotherapy or targeted therapy. These aggressive tumors would likely not benefit from upfront surgery due to the high chance of occult micro-metastasis. The other limitation is the toxicity of NAT which might result in opting out or delaying surgery (22). In the SWOG s1505 study, 25–30% of neoadjuvant chemotherapy (NACT) patients could not undergo surgery due to disease progression and its toxicity (23). However, compared to NACT, only 66% of patients can undergo adjuvant chemotherapy after upfront surgery as per the literature (24). The possibility of overtreatment in the patients treated with NAT is less as few patients are cured after completion of treatment.


Meta-analysis of NAT in RPC

We performed an online search for meta-analyses comparing NAT and upfront surgery in RPC, including intention-to-treat and per-protocol analysis. We found seven meta-analyses (shown in Table 1) that analyzed the role of NAT in RPC and showed an increased pathological free (R0) resection rate, better treatment compliance, and fewer lymph nodes. All the studies compared the effectiveness of NAT in the form of chemotherapy and chemo-radiation. Two out of these studies showed improved survival of RPC in their per-protocol analysis, including the neoadjuvant chemo-radiation (NACT-RT) arm in the study by Pan et al. (25) and the NACT arm in the study by Lee et al. (26). Another study by Ye et al. showed a survival benefit in its sub-group analysis with the NACT arm (27) while Ghanem et al. showed a survival benefit with NACT and NACT-RT in the NAT arm (28).

Table 1

Meta-analysis comparing NAT versus upfront surgery in RPC

Meta-analysis Overall survival, HR (95% CI) Advantage Disadvantage
Pan et al. (25) Per-protocol analysis for RPC: 0.75 (0.63–0.89), P<0.001 in favor of NACT-RT 1. Mostly retrospective studies and non-randomized controlled trials
Intention-to-treat analysis for RPC: 1.02 (0.85–1.22), P=0.818 2. Heterogeneity in chemotherapy regimen and radiation doses
3. Included trials with chemotherapy alone and chemo-radiation treatment as well
4. RPC and BRPC included
Lee et al. (26) Per-protocol analysis: 0.72 (0.68–0.76), P<0.001 in favor of NACT 1. No heterogeneity between sub-groups 1. All retrospective studies
Intention-to-treat analysis: 0.96 (0.82–1.12), P=0.601 2. RPC only 2. Old chemotherapy regimen
Ye et al. (27) Overall: 0.86 (0.73–1.03), P=0.10 1. Intention-to-treat analysis 1. Mostly retrospective studies
Subgroup analysis with gemcitabine-based NACT: 0.75 (0.57–0.99), P=0.04 in favor of NACT 2. Few trials included newer chemotherapy regimens
3. RPC only
Ghanem
et al. (28)
Overall: 0.75 (0.58–0.98), P=0.033 1. Intention-to-treat analysis 1. Heterogeneity of neoadjuvant regimen
2. RPC only 2. Included trials with chemotherapy alone and chemo-radiation treatment as well
Ren et al. (29) Overall: 0.91 (0.79–1.05), P=0.561 1. Intention-to-treat analysis 1. Mostly retrospective studies
2. RPC only
van Dam et al. (30) Overall, for RPC: 0.77 (0.53–1.12), P=0.18 1. Intention-to-treat analysis 1. Heterogeneity of neoadjuvant regimen
2. All randomized controlled trials included 2. Use of gemcitabine-based chemotherapy in the adjuvant setting
3. RPC and BRPC included
Uson Junior et al. (31) Overall: 0.76 (0.52–1.11), P=0.150 1. All phase II/III prospective studies 1. Only one trial evaluated the new regimen (gemcitabine + nab-paclitaxel)
2. Intention-to-treat analysis
3. RPC only

NAT, neoadjuvant therapy; RPC, resectable pancreatic carcinoma; HR, hazard ratio; CI, confidence interval; NACT-RT, neoadjuvant chemo-radiation; BRPC, borderline resectable pancreatic cancer; NACT, neoadjuvant chemotherapy.

The issues with all the meta-analyses were that most of them included retrospective studies and old chemotherapy regimens, few interpreted results on per-protocol analysis while others included both RPC and BRPC. One of the recent meta-analyses by Uson Junior et al. did not show any survival benefit of NAT in RPC, however, most of the studies included in it used gemcitabine-based chemotherapy when gemcitabine/nab-paclitaxel combination was the choice of chemotherapy in the adjuvant treatment of pancreatic cancer (31). Since 2019, the presence of specific molecular markers in pancreatic cancer has shown an excellent response to targeted therapy. The data on prognostic biomarkers or genetic mutations have been lacking in recent meta-analyses.

Due to heterogeneity in patient selection with outdated chemotherapy regimens, different radiation doses, and the retrospective nature of the studies, it is necessary to assess outcomes based on randomized controlled trials (RCTs) to rule out selection bias, with the current standard of treatment along with prognostic and predictive markers to select patients that could benefit from NAT.


RCTs of NACT in RPC

One of the earlier studies, the NEOPAC randomized phase III study, was done to compare NACT with upfront surgery but it was stopped prematurely due to low accrual of patients (32).

Five phase II/III RCTs (shown in Table 2) have shown the outcome of NACT compared to upfront surgery in RPC.

Table 2

RCTs comparing NACT versus upfront surgery

Trial/phase No. of patients Treatment arms Median OS (months) P value
Arm A Arm B/C
NEPAFOX/II (33) 40 Arm A: FOLFIRINOX followed by surgery, then adjuvant FOLFIRINOX 10 25.7 0.03*
Arm B: surgery followed by adjuvant gemcitabine
PACT-15/II (34) 88 Arm A: PEXG followed by surgery 38.2 20.4 (arm B) Not done
Arm B: surgery followed by adjuvant gemcitabine 26.4 (arm C)
Arm C: surgery followed by adjuvant PEXG
NEONAX/II (35) 59 Arm A: gemcitabine + nab-paclitaxel followed by surgery, then adjuvant gemcitabine + nab-paclitaxel 25.5 16.7 Not done
Arm B: surgery followed by gemcitabine and nab-paclitaxel
PREP-02 JSAP-05/III (36) 362 Arm A: gemcitabine + S1 followed by surgery, then adjuvant S1 36.7 26.6 0.02*
Arm B: surgery followed by adjuvant S1
NORPACT-1/II (37) 140 Arm A: FOLFIRINOX (4 cycles) followed by surgery, then adjuvant FOLFIRINOX (8 cycles) 25.1 38.5 0.096
Arm B: surgery followed by adjuvant FOLFIRINOX (12 cycles)

*, statistically significant. RCTs, randomized controlled trials; NACT, neoadjuvant chemotherapy; OS, overall survival. PEXG: cisplatin, epirubicin, capecitabine, gemcitabine; S1: tegafur/gimeracil/octerasil; FOLFIRINOX: folinic acid, fluorouracil, irinotecan, oxaliplatin.

The NEPAFOX phase II study included NACT with FOLFIRINOX versus upfront surgery followed by adjuvant gemcitabine and it showed worse survival outcomes in the NACT arm. Due to low patient numbers, the analyses for primary and secondary endpoints were not robust and only descriptive. Another limitation was that the treatment arms were not balanced in the chemotherapy regimen and included BRPC patients as well (33).

Two trials, PACT-15 and NEONAX showed better numerical median survival in the NACT arm as compared to the upfront surgery arm. While the primary endpoint of the PACT-15 study was event-free survival analysis (34), the NEONAX showed that the gemcitabine/nab-paclitaxel combination was well tolerated in the neoadjuvant setting. In the NEONAX trial, there was a discrepancy of chemotherapy exposure in both the arms with 90% of patients in the neoadjuvant arm completing pre-operative chemotherapy and only 42% of patients starting chemotherapy in the adjuvant arm (35).

The largest of these studies, the Japanese PREP-02 phase III trial of neoadjuvant gemcitabine and oral fluoropyrimidine S-1 versus adjuvant chemotherapy S-1, randomized 362 RPC patients and showed a significant advantage of NACT over surgery followed by adjuvant chemotherapy [median overall survival (OS) of 36.7 versus 26.6 months; P=0.015] (36). It was seen that the patients in the neoadjuvant arm received two more cycles than the patients treated in the upfront surgery group. BRPC patients with portal vein involvement were also included in this study. The non-availability of S-1 in Europe and the United States limited its research and application in other parts of the world.

NORPACT-1 study presented in ASCO 2023 compared neoadjuvant FOLFIRINOX with upfront surgery in RPC and showed a numerical survival benefit of upfront surgery (37). The results were argumentative since 83% of the patients in the neoadjuvant arm started treatment, out of which 50% completed NACT. About 66% of patients in the upfront surgery arm received adjuvant chemotherapy. Though there was a favorable surgical outcome in the neoadjuvant arm, it did not result in improved survival, as seen in most of the previous trials.

Many studies were done to find the optimal choice of chemotherapy for pancreatic cancer. Gemcitabine/nab-paclitaxel and mFOLFIRINOX have shown better OS in both the metastatic first-line and adjuvant settings (38). SWOG s1505 study was done to compare the efficacy of these two chemotherapy regimens in neoadjuvant settings of RPC. The study showed that both regimens were safe, equally efficacious, and showed high resectability rates, without comparing it to upfront surgery (23). While the NEONAX study showed better survival outcomes in the NACT arm with gemcitabine/nab-paclitaxel, there are no trials showing benefit with neoadjuvant FOLFIRINOX compared to upfront surgery. Three randomized phase III trials ALLIANCE A021806, PRODIGE 48, and PREPOANC 3 are ongoing to evaluate the role of neoadjuvant versus adjuvant FOLFIRINOX in patients with RPC in terms of disease-free survival and OS (39-41).


RCTs of NACT-RT in RPC

Three phase III RCTs (shown in Table 3) have investigated the efficacy of NACT-RT over immediate surgery in RPC.

Table 3

RCTs comparing NACT-RT versus upfront surgery in RPC

Trial/phase No. of patients Treatment arms Median OS (months) P value
Arm A Arm B
Casadei et al./III (42) 38 (stopped due to poor accrual) Arm A: gemcitabine + RT 54 Gy/30# followed by surgery, then adjuvant gemcitabine 22.4 19.5 0.174
Arm B: Surgery followed by adjuvant gemcitabine
Golcher et al./II (43) 66 (stopped due to poor accrual) Arm A: gemcitabine/cisplatin + RT 55.8 Gy/30# followed by surgery, then adjuvant gemcitabine 17.4 14.4 0.96
Arm B: surgery followed by adjuvant gemcitabine
PREOPANC/III (44) 246 Arm A: gemcitabine + RT 36 Gy/15# followed by surgery, then adjuvant gemcitabine 15.7 14.3 0.23
Arm B: surgery followed by adjuvant gemcitabine

RCTs, randomized controlled trials; NACT-RT, neoadjuvant chemo-radiation; RPC, resectable pancreatic carcinoma; OS, overall survival; RT, radiation therapy.

Two trials done by Casadei and Golcher intended to evaluate the use of preoperative CRT for patients with RPC. Both trials were closed early due to poor patient accrual with no significant differences observed in OS (42,43).

The PREOPANC was the first randomized phase III trial that reached full accrual in evaluating the efficacy of NACT-RT in RPC. NACT-RT was associated with better disease-free survival and locoregional failure-free period, with significantly lower rates of pathologic lymph nodes, perineural invasion, and venous invasion. In the final analysis, neoadjuvant treatment improved OS in the overall cohort; however, in the RPC group, the results were not statistically significant (P=0.23). The benefit of neoadjuvant treatment in the trial was mainly driven by borderline pancreatic cancer (P=0.045) (44). After the PRODIGE phase 3 study showed better OS with FOLFIRINOX than gemcitabine monotherapy in advanced pancreatic cancer, it was hypothesized that FOLFIRINOX might supersede the efficacy of gemcitabine-based chemoradiotherapy in the neoadjuvant setting. PREOPANC-2 study presented in ESMO 2023 compared neoadjuvant FOLFIRINOX versus neoadjuvant gemcitabine-based chemoradiotherapy in patients with early pancreatic cancer. However, the median OS was 21.9% in the FOLFIRINOX arm versus 21.3% in the gemcitabine-based NACT-RT arm [hazard ratio (HR) 0.87; 95% confidence interval (CI): 0.68–1.12; P=0.28]. Also, the resection rates (77% versus 75%, respectively; P=0.7) and serious adverse rates (49% versus 43%, respectively; P=0.26) were also similar between the treatment arms.


Potential role of bio-markers and molecular testing in RPC

Various biomarkers and genetic mutations (Table 4) are used to predict the treatment of pancreatic cancer. These markers can help select RPC patients with high chances of early metastasis and can benefit from NAT. A recent study performing next-generation sequencing (NGS) done on resected pancreatic cancer specimens found pathogenic variants in 94% of samples, 18% of which were potentially actionable (45).

Table 4

Biomarkers and genetic mutations in pancreatic cancer

Biomarker/genetic mutation Incidence Diagnostic Prognostic Predictive
CA 19.9 80–85% + + +
CEA 30–60% + +
MMR/MSI 1–2% + +
GATA6 90–98% +
ctDNA 85–90% + +
KRAS 90–95% + + +
TP53 70–75% + +
SMAD4 15–20% + +
CDKN2A 35–40% + + +
BRCA1/2 4–5% + +

The most common biomarker is CA 19.9 which has a sensitivity of 79% and specificity of 82% in symptomatic patients (46). CA 19.9 is not tumor-type specific and can be raised in other malignancies or benign diseases (47). Hence, other biomarkers such as CEA have been used in combination with CA 19.9 to increase the positive predictive value and know the prognosis of pancreatic cancer (48).

CA 19.9 has been used in pancreatic cancer to track the treatment response, predict resectability, and monitor the recurrence. Preoperative elevation of CA 19.9 in patients with RPC has been associated with decreased OS (49). A study by Humphris et al. analyzed RPC patients who underwent resection and found that a pre-operative CA 19.9 >120 U/mL had significantly longer disease-free survival than patients with a CA 19.9 ≤120 U/mL (36 versus 17 months, respectively). Post-operative CA 19.9 >37 U/mL was also associated with poor prognosis and shorter OS (50).

RPC with elevated CA 19.9 levels at diagnosis are biologically borderline resectable regardless of anatomic resectability, and neoadjuvant systemic therapy is advised (51). A recent prospective randomized study by Chiu et al. has shown CA 19.9 as a predictor of survival and distant failure in RPC based on response to adjuvant gemcitabine (52). Another study by Al Abbas et al. has shown serum CA 19.9 response as a useful marker after NAT and resection in predicting patient survival in pancreatic cancer. It has shown that serum CA 19.9 response ≥85% remained a strong independent predictor of survival (HR: 0.47; P=0.007) on multivariate analysis (53).

Some mutations have shown a correlation with treatment response in metastatic pancreatic cancer.

Tumors with deficient mismatch repair gene (dMMR) have been seen to benefit from programmed death-ligand 1 (PD-L1) inhibitors (54). Riazy et al. showed the addition of adjuvant chemotherapy in RPC patients significantly increased survival in those with proficient mismatch repair gene (pMMR) than dMMR patients. This data indicated the potential for dMMR as a useful predictive chemotherapy biomarker. NAT may be more appropriate in pMMR tumors than upfront surgery (55).

Patients with BRCA1/2 mutations have shown a higher response to platinum-based therapy (56) while tumors with low GATA6 expression show poor response to mFOLFIRINOX (57). In a retrospective review of 61 RPC and BRPC patients with known BRCA1/2 mutations, neoadjuvant FOLFIRINOX showed significantly higher rates of pathological complete response in the patients with a germline BRCA1/2 mutations (44%) compared to the BRCA wildtype (10%) (58). The application of BRCA1/2 testing is, however, currently limited to those with a positive family history or metastatic disease. GATA6 has been shown to inhibit metastasis by inhibiting de-differentiation of epithelial to mesenchymal transition (EMT). In the ESPAC 3 study, Martinelli et al. demonstrated that GATA6 expression strongly predicted survival in patients treated with adjuvant 5-FU, with a median survival of 27 months in patients with high expression against 14 months with low expression. This survival difference in GATA expression levels was not detected in patients treated with adjuvant gemcitabine (59).

KRAS inhibitors have shown promising results in early studies for patients with this mutation. A certain KRASG12C mutation seen in 1–2% of pancreatic adenocarcinoma patients has recently been identified as a druggable target. However, their application in assessing treatment response in RPC is still a long way off. A prospective phase II trial enrolling patients with resectable and BRPC and selecting NAT (fluoropyrimidine-based or gemcitabine-based) based on molecular profiling has reported high resection rates (60). Another phase II PIONEER-Panc study is investigating novel therapeutic strategies in early-stage pancreatic cancer.

In a retrospective study by Chen et al., the predictive role of CDKN2A in RPC patients receiving chemoradiation with gemcitabine showed that the presence of a CDKN2A mutation did not predict treatment response (61).

There is an emerging role of circulating tumor DNA (ctDNA) in early detection, treatment monitoring, and its prediction in pancreatic cancer. Genomic sequencing has shown the presence of mutations across four genes (KRAS, TP53, SMAD4, CDKN2A) in more than 90% of patients (62). Somatic variations are seen in around 5% of pancreatic cancer, reflecting extensive inter-tumoural genetic heterogeneity. Due to this, there is low concordance between plasma ctDNA detection rates in pancreatic cancer (63). It is around 10% in early pancreatic cancer which restricts its sensitivity for profiling. This suggests that the use of liquid biopsy testing requires greater sensitivity and specificity than targeting ctDNA alone. Molecules derived from tumors can be used to enhance their analytical validity and clinical utility. Such methods require rigorous testing in large-scale prospective studies to determine the use of liquid biopsy profiling in pancreatic cancer. Therefore, we need RCTs to incorporate these biomarkers to identify a subset of patients who can benefit from a specific NAT in RPC.


Conclusions

This review article aims to provide an update on the recent state of NAT in RPC. Studies have shown better surgical rates and compliance with NAT for RPC, however, very few have translated into better survival outcomes. Although NAT has shown better survival outcomes in BRPC, its limited benefit in RPC might be due to certain unknown confounding factors. Based on recent evidence, there is a need for the identification of biologically aggressive pancreatic cancer through reliable biomarkers, and personalization of therapy through molecular characterization and targeting. Molecular and genetic markers might help in identifying this subset of RPC patients who would benefit from NAT, compared to upfront surgery.


Acknowledgments

Funding: None.


Footnote

Peer Review File: Available at https://apc.amegroups.com/article/view/10.21037/apc-23-14/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://apc.amegroups.com/article/view/10.21037/apc-23-14/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Huxley R, Ansary-Moghaddam A, Berrington de González A, et al. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer 2005;92:2076-83. [Crossref] [PubMed]
  2. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  3. Bray F, Ferlay J, Soerjomataram I, et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2018;68:394-424. [Crossref] [PubMed]
  4. Park W, Chawla A, O'Reilly EM. Pancreatic Cancer: A Review. JAMA 2021;326:851-62. [Crossref] [PubMed]
  5. Krech RL, Walsh D. Symptoms of pancreatic cancer. J Pain Symptom Manage 1991;6:360-7. [Crossref] [PubMed]
  6. National Comprehensive Cancer Network. Clinical Practice Guidelines in Oncology: Pancreatic Adenocarcinoma. Version 1 2022.
  7. Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. N Engl J Med 2014;371:2140-1. [Crossref] [PubMed]
  8. Allen VB, Gurusamy KS, Takwoingi Y, et al. 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 2016;7:CD009323. [Crossref] [PubMed]
  9. Ta R, O'Connor DB, Sulistijo A, et al. The Role of Staging Laparoscopy in Resectable and Borderline Resectable Pancreatic Cancer: A Systematic Review and Meta-Analysis. Dig Surg 2019;36:251-60. [Crossref] [PubMed]
  10. Marion-Audibert AM, Vullierme MP, Ronot M, et al. Routine MRI With DWI Sequences to Detect Liver Metastases in Patients With Potentially Resectable Pancreatic Ductal Carcinoma and Normal Liver CT: A Prospective Multicenter Study. AJR Am J Roentgenol 2018;211:W217. [Crossref] [PubMed]
  11. Tempero MA, Malafa MP, Al-Hawary M, et al. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2021;19:439-57. [Crossref] [PubMed]
  12. Suker M, Beumer BR, Sadot E, et al. FOLFIRINOX for locally advanced pancreatic cancer: a systematic review and patient-level meta-analysis. Lancet Oncol 2016;17:801-10. [Crossref] [PubMed]
  13. Peddi PF, Lubner S, McWilliams R, et al. Multi-institutional experience with FOLFIRINOX in pancreatic adenocarcinoma. JOP 2012;13:497-501. [PubMed]
  14. de W Marsh R. Primary systemic therapy in resectable pancreatic ductal adenocarcinoma using mFOLFIRINOX: A pilot study. J Surg Oncol 2018;117:354-62. [Crossref] [PubMed]
  15. Neoptolemos JP, Dunn JA, Stocken DD, et al. Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet 2001;358:1576-85. [Crossref] [PubMed]
  16. Neoptolemos JP, Stocken DD, Friess H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med 2004;350:1200-10. [Crossref] [PubMed]
  17. Neoptolemos JP, Stocken DD, Bassi C, et al. Adjuvant chemotherapy with fluorouracil plus folinic acid vs gemcitabine following pancreatic cancer resection: a randomized controlled trial. JAMA 2010;304:1073-81. [Crossref] [PubMed]
  18. Oettle H, Neuhaus P, Hochhaus A, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA 2013;310:1473-81. [Crossref] [PubMed]
  19. Neoptolemos JP, Palmer DH, Ghaneh P, 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;389:1011-24. [Crossref] [PubMed]
  20. Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N Engl J Med 2018;379:2395-406. [Crossref] [PubMed]
  21. Sohal DP, Walsh RM, Ramanathan RK, et al. Pancreatic adenocarcinoma: treating a systemic disease with systemic therapy. J Natl Cancer Inst 2014;106:dju011. [Crossref] [PubMed]
  22. Shubert CR, Bergquist JR, Groeschl RT, et al. Overall survival is increased among stage III pancreatic adenocarcinoma patients receiving neoadjuvant chemotherapy compared to surgery first and adjuvant chemotherapy: An intention to treat analysis of the National Cancer Database. Surgery 2016;160:1080-96. [Crossref] [PubMed]
  23. Sohal DPS, Duong M, Ahmad SA, et al. Efficacy of Perioperative Chemotherapy for Resectable Pancreatic Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2021;7:421-7. Erratum in: JAMA Oncol 2021 Sep 23. [Crossref] [PubMed]
  24. Sata N, Kurashina K, Nagai H, et al. The effect of adjuvant and neoadjuvant chemo(radio)therapy on survival in 1,679 resected pancreatic carcinoma cases in Japan: report of the national survey in the 34th annual meeting of Japanese Society of Pancreatic Surgery. J Hepatobiliary Pancreat Surg 2009;16:485-92. [Crossref] [PubMed]
  25. Pan L, Fang J, Tong C, et al. 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. [Crossref] [PubMed]
  26. Lee YS, Lee JC, Yang SY, et al. Neoadjuvant therapy versus upfront surgery in resectable pancreatic cancer according to intention-to-treat and per-protocol analysis: A systematic review and meta-analysis. Sci Rep 2019;9:15662. [Crossref] [PubMed]
  27. Ye M, Zhang Q, Chen Y, et al. Neoadjuvant chemotherapy for primary resectable pancreatic cancer: a systematic review and meta-analysis. HPB (Oxford) 2020;22:821-32. [Crossref] [PubMed]
  28. Ghanem I, Lora D, Herradón N, et al. Neoadjuvant chemotherapy with or without radiotherapy versus upfront surgery for resectable pancreatic adenocarcinoma: a meta-analysis of randomized clinical trials. ESMO Open 2022;7:100485. [Crossref] [PubMed]
  29. Ren X, Wei X, Ding Y, et al. Comparison of neoadjuvant therapy and upfront surgery in resectable pancreatic cancer: a meta-analysis and systematic review. Onco Targets Ther 2019;12:733-44. [Crossref] [PubMed]
  30. van Dam JL, Janssen QP, Besselink MG, et al. Neoadjuvant therapy or upfront surgery for resectable and borderline resectable pancreatic cancer: A meta-analysis of randomised controlled trials. Eur J Cancer 2022;160:140-9. [Crossref] [PubMed]
  31. Uson PLS Junior, Dias E. Does neoadjuvant treatment in resectable pancreatic cancer improve overall survival? A systematic review and meta-analysis of randomized controlled trials. ESMO Open 2023;8:100771. [Crossref] [PubMed]
  32. Heinrich S, Pestalozzi B, Lesurtel M, et al. Adjuvant gemcitabine versus NEOadjuvant gemcitabine/oxaliplatin plus adjuvant gemcitabine in resectable pancreatic cancer: a randomized multicenter phase III study (NEOPAC study). BMC Cancer 2011;11:346. [Crossref] [PubMed]
  33. Al-Batran SE, Reichart A, Bankstahl US, 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 2021;39:406. [Crossref]
  34. Reni M, Balzano G, Zanon S, 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;3:413-23. [Crossref] [PubMed]
  35. Seufferlein T, Uhl W, Algül H, et al. LBA56 Perioperative or only adjuvant nab-paclitaxel plus gemcitabine for resectable pancreatic cancer: results of the NEONAX trial, a randomized phase II AIO study. Ann Oncol 2021;32:S1333. [Crossref]
  36. Motoi F, Kosuge T, Ueno 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/JSAP05). Jpn J Clin Oncol 2019;49:190-4. [Crossref] [PubMed]
  37. Labori KJ, Lassen K, Hoem D, et al. 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. [Crossref] [PubMed]
  38. Riedl JM, Posch F, Horvath L, et al. Gemcitabine/nab-Paclitaxel versus FOLFIRINOX for palliative first-line treatment of advanced pancreatic cancer: A propensity score analysis. Eur J Cancer 2021;151:3-13. [Crossref] [PubMed]
  39. Chawla A, Shi Q, Ko AH, et al. Alliance A021806: A phase III trial evaluating perioperative versus adjuvant therapy for resectable pancreatic cancer. J Clin Oncol 2023 41:TPS4204.
  40. Schwarz L, Vernerey D, Bachet JB, et al. 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. [Crossref] [PubMed]
  41. van Dam JL, Verkolf EMM, Dekker EN, et al. Perioperative or adjuvant mFOLFIRINOX for resectable pancreatic cancer (PREOPANC-3): study protocol for a multicenter randomized controlled trial. BMC Cancer 2023;23:728. [Crossref] [PubMed]
  42. Casadei R, Di Marco M, Ricci C, et al. 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 2015;19:1802-12. [Crossref] [PubMed]
  43. Golcher H, Brunner TB, Witzigmann H, 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 2015;191:7-16. [Crossref] [PubMed]
  44. Versteijne E, van Dam JL, Suker M, et al. Neoadjuvant Chemoradiotherapy Versus Upfront Surgery for Resectable and Borderline Resectable Pancreatic Cancer: Long-Term Results of the Dutch Randomized PREOPANC Trial. J Clin Oncol 2022;40:1220-30. [Crossref] [PubMed]
  45. Krepline AN, Bliss L, Geurts J, et al. Role of Molecular Profiling of Pancreatic Cancer After Neoadjuvant Therapy: Does it Change Practice? J Gastrointest Surg 2020;24:235-42. [Crossref] [PubMed]
  46. Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19-9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol 2007;33:266-70. [Crossref] [PubMed]
  47. Poruk KE, Gay DZ, Brown K, et al. The clinical utility of CA 19-9 in pancreatic adenocarcinoma: diagnostic and prognostic updates. Curr Mol Med 2013;13:340-51. [PubMed]
  48. van Manen L, Groen JV, Putter H, et al. Elevated CEA and CA19-9 serum levels independently predict advanced pancreatic cancer at diagnosis. Biomarkers 2020;25:186-93. [Crossref] [PubMed]
  49. Palmquist C, Dehlendorff C, Calatayud D, et al. Prediction of Unresectability and Prognosis in Patients Undergoing Surgery on Suspicion of Pancreatic Cancer Using Carbohydrate Antigen 19-9, Interleukin 6, and YKL-40. Pancreas 2020;49:53-61. [Crossref] [PubMed]
  50. Humphris JL, Chang DK, Johns AL, et al. The prognostic and predictive value of serum CA19.9 in pancreatic cancer. Ann Oncol 2012;23:1713-22. [Crossref] [PubMed]
  51. Bergquist JR, Puig CA, Shubert CR, et al. Carbohydrate Antigen 19-9 Elevation in Anatomically Resectable, Early Stage Pancreatic Cancer Is Independently Associated with Decreased Overall Survival and an Indication for Neoadjuvant Therapy: A National Cancer Database Study. J Am Coll Surg 2016;223:52-65. [Crossref] [PubMed]
  52. Chiu YF, Liu TW, Shan YS, et al. Carbohydrate Antigen 19-9 Response to Initial Adjuvant Chemotherapy Predicts Survival and Failure Pattern of Resected Pancreatic Adenocarcinoma but Not Which Patients Are Suited for Additional Adjuvant Chemoradiation Therapy: From a Prospective Randomized Study. Int J Radiat Oncol Biol Phys 2023;117:74-86. [Crossref] [PubMed]
  53. Al Abbas AI, Zenati M, Reiser CJ, et al. Serum CA19-9 Response to Neoadjuvant Therapy Predicts Tumor Size Reduction and Survival in Pancreatic Adenocarcinoma. Ann Surg Oncol 2020;27:2007-14. Erratum in: Ann Surg Oncol 2020 Feb 7. [Crossref] [PubMed]
  54. Constantin A, Iovănescu V, Cazacu IM, et al. Evaluation of MMR Status and PD-L1 Expression Using Specimens Obtained by EUS-FNB in Patients with Pancreatic Ductal Adenocarcinoma (PDAC). Diagnostics (Basel) 2022;12:294. [Crossref] [PubMed]
  55. Riazy M, Kalloger SE, Sheffield BS, et al. Mismatch repair status may predict response to adjuvant chemotherapy in resectable pancreatic ductal adenocarcinoma. Mod Pathol 2015;28:1383-9. [Crossref] [PubMed]
  56. Uson PLS Jr, Samadder NJ, Riegert-Johnson D, et al. Clinical Impact of Pathogenic Germline Variants in Pancreatic Cancer: Results From a Multicenter, Prospective, Universal Genetic Testing Study. Clin Transl Gastroenterol 2021;12:e00414. [Crossref] [PubMed]
  57. O'Kane GM, Grünwald BT, Jang GH, et al. GATA6 Expression Distinguishes Classical and Basal-like Subtypes in Advanced Pancreatic Cancer. Clin Cancer Res 2020;26:4901-10. [Crossref] [PubMed]
  58. Golan T, Barenboim A, Lahat G, et al. Increased Rate of Complete Pathologic Response After Neoadjuvant FOLFIRINOX for BRCA Mutation Carriers with Borderline Resectable Pancreatic Cancer. Ann Surg Oncol 2020;27:3963-70. [Crossref] [PubMed]
  59. Martinelli P, Carrillo-de Santa Pau E, Cox T, et al. GATA6 regulates EMT and tumour dissemination, and is a marker of response to adjuvant chemotherapy in pancreatic cancer. Gut 2017;66:1665-76. [Crossref] [PubMed]
  60. Tsai S, Christians KK, George B, et al. A Phase II Clinical Trial of Molecular Profiled Neoadjuvant Therapy for Localized Pancreatic Ductal Adenocarcinoma. Ann Surg 2018;268:610-9. [Crossref] [PubMed]
  61. Chen J, Li D, Killary AM, et al. Polymorphisms of p16, p27, p73, and MDM2 modulate response and survival of pancreatic cancer patients treated with preoperative chemoradiation. Ann Surg Oncol 2009;16:431-9. [Crossref] [PubMed]
  62. Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016;531:47-52. [Crossref] [PubMed]
  63. Heitzer E, Haque IS, Roberts CES, et al. Current and future perspectives of liquid biopsies in genomics-driven oncology. Nat Rev Genet 2019;20:71-88. [Crossref] [PubMed]
doi: 10.21037/apc-23-14
Cite this article as: Purohit S, Ahlawat P, Tandon S, Bellige AR, Gairola M. Role of neoadjuvant therapy in resectable pancreatic carcinoma: a review article. Ann Pancreat Cancer 2024;7:4.

Download Citation