Update on management of pancreatic cancer: a literature review
Review Article

Update on management of pancreatic cancer: a literature review

Eric C. H. Lai ORCID logo, Aidan K. Y. Ung

Department of Surgery, Pamela Youde Nethersole Eastern Hospital, Hong Kong SAR, China

Contributions: (I) Conception and design: ECH Lai; (II) Administrative support: ECH Lai; (III) Provision of study materials or patients: ECH Lai; (IV) Collection and assembly of data: Both authors; (V) Data analysis and interpretation: Both authors; (VI) Manuscript writing: Both authors; (VII) Final approval of manuscript: Both authors.

Correspondence to: Dr. Eric C. H. Lai, MB ChB, MRCS(Ed), FRACS. Department of Surgery, Pamela Youde Nethersole Eastern Hospital, 3 Lok Man Road, Chai Wan, Hong Kong SAR, China. Email: elaichun@gmail.com.

Background and Objective: Pancreatic cancer is an aggressive malignancy with high mortality. At the time of diagnosis, majority of patients (80–90%) present with either locally advanced unresectable disease or metastatic disease. Even after curative resection, the recurrence rate remains quite high. This article aimed at reviewing the updated management of pancreatic cancer.

Methods: We identified literature by searching Medline and PubMed from January 2010 to June 2023 using the keywords.

Key Content and Findings: A multidisciplinary approach is essential to optimize the outcomes for both curable and advanced diseases. Management of pancreatic cancer divided into resectable, borderline resectable, locally advanced, and metastatic diseases. Surgery and adjuvant chemotherapy is a standard treatment approach for resectable pancreatic cancer. The recommended adjuvant chemotherapy regimen for patients with good functional status is modified FOLFIRINOX (5-fluorouracil, folinic acid, irinotecan, and oxaliplatin). The recommended adjuvant chemotherapy regimen for patients with suboptimal functional status is gemcitabine plus capecitabine or monotherapy gemcitabine. The optimal treatment strategy for borderline resectable pancreatic cancer is still uncertain. Traditionally, upfront surgery is the choice of treatment. There is increasing evidence showing benefits of neoadjuvant therapy in borderline resectable pancreatic cancer. However, the optimal neoadjuvant treatment regimen was not certain yet. Advancement of chemotherapy has a positive impact for the survival of advanced disease. For patients with good functional status, the recommended first-line systemic chemotherapy for unresectable locally advanced disease or metastatic disease is combination chemotherapy regimens such as FOLFIRINOX, gemcitabine plus nab-paclitaxel. For patients with suboptimal functional status, the recommended first-line systemic chemotherapy for unresectable locally advanced disease or metastatic disease is gemcitabine plus capecitabine or monotherapy gemcitabine. Recently, more researches showed promising results in the use of nanoliposomal irinotecan, targeted agents such as a poly [adenosine diphosphate (ADB)-ribose] polymerase inhibitor, tyrosine receptor kinase (TRK) inhibitors, and immune checkpoint-inhibitors.

Conclusions: Pancreatic cancer is a challenging disease for management. Radical surgery itself is not enough for prolong survival. The improvement of chemotherapy, target agents and immunotherapy with multidisciplinary approach will be the only solution for improvement of survival outcome and quality of life for patients with pancreatic cancer.

Keywords: Pancreatic cancer; borderline resectable pancreatic cancer; adjuvant chemotherapy; neoadjuvant chemotherapy; pancreatectomy


Submitted Sep 02, 2023. Accepted for publication Mar 18, 2024. Published online May 17, 2024.

doi: 10.21037/cco-23-94


Introduction

Approximately 85–95% of pancreatic cancers are pancreatic ductal adenocarcinomas originating from the exocrine part of the pancreas, and the term “pancreatic cancer” typically denotes this disease entity (1). It is an aggressive malignancy with increasing in incidence and with a high mortality. Globally, pancreatic cancer is currently the 7th highest cause of cancer-related death, and with the current trend, pancreatic cancer is projected to be the 2nd leading cause of cancer-related death by the year 2030 (2-4). Pancreatic cancer remains a disease with very poor prognosis (5-year survival rate, 2–10%) (5,6). In 2020, the number of mortalities from pancreatic cancer (n=466,003) was almost the same as the number of pancreatic cancer cases (n=495,773) (2). Radical surgery with adjuvant chemotherapy is the only chance of cure. However, approximately 80–90% of patients present with either locally advanced or metastatic disease at the time of diagnosis (6,7). A multidisciplinary approach is essential to optimize outcomes for both curable and advanced diseases.

This article aimed at reviewing the updated management of pancreatic cancer. We present this article in accordance with the Narrative Review reporting checklist (available at https://cco.amegroups.com/article/view/10.21037/cco-23-94/rc).


Methods

We identified literature by searching Medline and PubMed from 1st January 2010 to 30th June 2023 using the keywords “pancreatic cancer”, “borderline resectable pancreatic cancer”, “metastatic pancreatic cancer”, “systemic chemotherapy”, “adjuvant treatment”, and “neoadjuvant treatment”. Only English language literature was considered. Case reports were excluded from analysis. Additional papers were identified by a manual search of the references from the key articles. There were no exclusion criteria for published information to the topics (Table 1).

Table 1

The search strategy summary

Items Specification
Date of search 1st August 2023
Databases and other sources searched Medline and PubMed
Search terms used “pancreatic cancer”, “borderline resectable pancreatic cancer”, “metastatic pancreatic cancer”, “systemic chemotherapy”, “adjuvant treatment”, and “neoadjuvant treatment”
Timeframe 1st January 2010 to 30th June 2023
Inclusion and exclusion criteria Only English language literature was considered. Case reports were excluded from analysis
Selection process E.C.H.L. and A.K.Y.U. did the literature search and selection. It was conducted independently. Consensus was obtained by discussion

Epidemiology & risk factors

Globally, pancreatic cancer is the 12th most common cancer (7). The common age group with pancreatic cancer are 60–80 years of age. The highest incidence regions for pancreatic cancer include North America, Central and Eastern Europe. In last 10 years, increasing trends were observed in North America, Western Europe, and Australia (8,9). Although the incidence of pancreatic cancer varied in different country, its global burden has more than doubled over the last 25 years and the incidence of pancreatic cancer is increasing by 0.5–1.0% per year (10-12). Incidence and mortality rate in high human development index (HDI) countries are about 3–4-fold higher compared with low and medium HDI countries, and are slightly higher in male than in female gender (13). Pancreatic cancer is the 4th most common cause of cancer-related deaths in most developed countries (14,15). Risk factors for developing pancreatic cancer includes modifiable and non-modifiable factors (4,16-19). Pancreatic cancer associated with familial pancreatic cancer or hereditary syndromes accounts for about 10% of cases (16,17).

Modifiable risk factors of developing pancreatic cancer included:

  • Smoking.
  • Obesity.
  • Long-standing diabetes mellitus.
  • Heavy alcohol use.
  • Chronic pancreatitis.

Non-modifiable risk factors of developing pancreatic cancer included:

  • Hereditary breast and ovarian cancer (pathogenic alterations in BRCA1 and BRCA2).
  • Hereditary pancreatitis (pathogenic alterations in PRSS1).
  • Lynch syndrome [pathogenic alterations in mismatch repair (MMR) genes (MLH1, MSH2, MSH6, & PMS2) and/or EPCAM].
  • Peutz-Jeghers syndrome (pathogenic alterations in STK11/LKB1 gene).
  • Familial atypical multiple mole syndrome (pathogenic alterations in the cell cycle gene CDKN2A).
  • Li-Fraumeni syndrome (germline mutations in TP53 gene).
  • PALB2 mutations.
  • ATM mutations.
  • Familial pancreatic ductal adenocarcinoma (those families have at least two 1st degree relatives with pancreatic ductal adenocarcinoma, but do not have a known predisposing germline mutation).

Presentations

The presenting symptoms of patients with pancreatic cancer are often subtle and nonspecific. Majority of pancreatic cancers (almost 70%) originated at the pancreatic head. Pancreatic head cancer often present with progressive obstructive jaundice, decreased appetite, tiredness, weight loss, and pancreatic exocrine insufficiency. Typically, the jaundice is painless in nature.

On the other hand, pancreatic cancers originated from the body and tail of pancreas often present with nonspecific symptoms, such as epigastric pain, back pain, weight loss, decreased appetite and tiredness (6,20). Due to its anatomical location, the symptoms take longer to develop not until the pancreatic tumor grow to a certain size or metastasize.

Pain with radiation to back is a poor prognostic symptom as there is a possibility of tumor involvement of adjacent nerve plexus. Sometimes, new-onset of diabetes mellitus or worsening of pre-existing diabetes mellitus may be a sign of pancreatic cancer and warrants further investigation for underlying cause also (21).

Therefore, no matter the location of the main tumor, if preliminary investigations cannot sort out the cause of persistent abdominal pain, either computed tomography (CT) scan or magnetic resonance imaging (MRI) is needed to rule out the possibility of pancreatic cancer.


Diagnostic techniques and imaging

A high-quality fine cut pancreatic protocol CT scan including thorax, abdomen and pelvis is the primary imaging modality for: (I) diagnosis; (II) staging; (III) assess resectability of pancreatic cancer, including vascular anatomy, relationship of tumor and major vessels; and (IV) assess any complications due to pancreatic cancer including biliary obstruction, gastric outlet obstruction, and nerve plexus involvement. The circumferential tumor-vessel involvement by measuring the degree of contact between the pancreatic tumor and adjacent blood vessels [i.e., the portal vein (PV) and superior mesenteric vein (SMV) as well as the celiac artery, common hepatic artery (CHA), gastroduodenal artery (GDA) and superior mesenteric arteries (SMAs)] is categorized as either: (I) uninvolved; (II) abutted (abutment implies ≤180° of circumferential tumor-vessel involvement); or (III) encased (encasement implies >180° of circumferential tumor-vessel involvement) (Figures 1-3). MRI and CT scan in assessing resectability and vascular relationship for pancreatic cancer are comparable (22-24). However, MRI has advantages in better detection of small, non-contour-deforming tumors, in characterizing indeterminate pancreatic findings in CT scan and in diagnosis of liver metastases (25,26).

Figure 1 Pancreatic tumor encasing PV-SMV. PV, portal vein; SMV, superior mesenteric vein.
Figure 2 Pancreatic tumor abutting SMV & SMA. SMA, superior mesenteric artery; SMV, superior mesenteric vein.
Figure 3 T4 pancreatic tumor encasing PV & abutting CHA/CA. PV, portal vein; CHA, common hepatic artery; CA, celiac artery.

Visualization of an obstructed biliary tree can be performed by endoscopic retrograde cholangiopancreatography (ERCP) or magnetic resonance cholangiopancreatography (MRCP). ERCP has the additional role in getting brush cytology of biliary stricture and insertion of biliary stent for biliary decompression.

18F-fluorodeoxyglucose (FDG) positron emission tomography (PET)/CT and endoscopic ultrasonography (EUS) are not considered a routine staging tool currently. They are supplementary investigations to CT scan or MRI (27). EUS has a role in obtaining a tissue diagnosis and getting tissue for molecular studies. Both fine-needle aspiration (EUS-FNA) and EUS guided fine-needle core biopsy (EUS-FNB) are safe and yield high diagnostic value (28,29). Even though the high diagnostic yield of EUS-FNA, it has potential limitations due to the cytological aspirations, such as inability to examine histologic architecture, and a small yield of tissue specimen for further immunohistochemical staining. EUS-FNB has the advantage of overcoming these potential limitations.

Serum carbohydrate antigen (CA) 19-9 (a cell surface glycoprotein complex), is a well-established biomarker for pancreatic cancer, and this tumor marker CA19-9 is routinely utilized during the diagnostic work-up of pancreatic cancer. CA19-9 is useful in monitoring treatment response and detecting recurrence. As a biomarker, its sensitivity is approximately 80%. It is well-known that Lewis and Secretor status can affect CA19-9 biosynthesis. CA19-9 has limitations for its clinical use for diagnosis and screening, including false positive elevation in patients with benign biliary, pancreatic and gastrointestinal disease, false negative results in patients with Lewis (a−b−) genotype, and its poor positive predictive value (72.3%) (30,31). These limitations make CA19-9 not a good cancer-specific biomarker. Recently, there is a promising development in the field of early detection of pancreatic cancer using diagnostic biomarkers, such as the detection of microRNA (miRNA) in serum and circulating tumour cells (32,33). However, this development is still in an initial phase.


Staging system

The staging system of pancreatic cancer is based on the American Joint Committee on Cancer (AJCC) Staging Manual (8th edition) (34). Stages T1 to T3 are defined only by the size of tumor (T1: ≤2 cm; T2: >2 cm–4 cm; T3: >4 cm). T4 is defined when tumor involves large blood vessels (celiac axis, CHA, and/or SMA) [T1 further divided into T1a (≤0.5 cm), T1b (>0.5–<1 cm), and T1c (1–2 cm) to encompass those small invasive carcinomas increasingly detected in association with cystic neoplasms of pancreas].

The N classification is divided into N0, N1 (1–3) and N2 (>3).

AJCC staging groupings are defined as follow:

  • Stage IA: T1N0M0;
  • Stage IB: T2N0M0;
  • Stage IIA: T3N0M0;
  • Stage IIB: T1-3N1M0;
  • Stage III: T1-3N2M0 or T4, any N, M0;
  • Stage IV: any T, any N, M1.

Management

Pancreatic cancer basically divides into resectable disease, borderline resectable disease, unresectable locally advanced disease, and metastatic disease. Resectability for localized disease bases on the extent of adjacent arterial and venous involvement by tumor. For resectable pancreatic cancer, radical resection of pancreatic cancer with adjuvant systemic chemotherapy currently remains the standard treatment approach and is the only chance for long-term survival. A borderline resectable pancreatic cancer (5–10%) is a tumor with limited vascular involvement that is technically resectable but with a high chance of positive resection margins. Many groups have proposed various definitions of borderline resectable pancreatic cancers (35-41) (Table 2). The optimal treatment strategy for borderline resectable pancreatic cancer is still uncertain. Traditionally, “upfront surgery” is the main treatment of choice. Identifying borderline resectable disease is important because a vascular resection +/− reconstruction is more likely needed at the time of pancreatoduodenectomy, and instead of a traditional “upfront surgery” approach, these patients may benefit from neoadjuvant treatment to increase the chance of R0 resection and decrease the surgical morbidity. However, the role and advantages of neoadjuvant treatment in resectable pancreatic cancer and borderline resectable pancreatic cancer remains unclear. There is increasing evidence showing benefits of neoadjuvant therapy in borderline resectable pancreatic cancer. For unresectable locally advanced pancreatic cancer, and metastatic disease, systemic chemotherapy is the choice of treatment. Surgical resection can be considered for those down-staged from unresectable locally advanced disease to resectable disease.

Table 2

Anatomic criteria for defining borderline resectable pancreatic cancer

Vessels NCCN (35,36) MDACC (37,38) ACTO (39) AHPBA/SSAT/SSO (40) JPS classification 7th edition (41)
Artery Head or uncinate process • Abutment of SMA ≤180° • Any TVI with the CHA with a normal artery proximal and distal to the TVI amenable to reconstruction • GDA: encasement up to the HA with either short segment encasement or direct abutment of the HA, without extension to the CA • SMA/CA: tumour contact/invasion ≤180° without showing stenosis or deformity
• SMA: solid tumor contact of <180° • Abutment or encasement (>180°) of short segment of HA • A TVI with the SMA measuring <180° of the circumference of the vessel wall • SMA: tumour abutment <180° of the circumference of the vessel wall • CHA: tumour contact/invasion without showing tumour contact/invasion of the PHA and/or CA
• CHA: solid tumor contact without extension to CA/HA bifurcation allowing for safe and complete resection and reconstruction • In case of contact/invasion to both PV and peripancreatic arteries, it was graded as BR-A
• Presence of variant arterial anatomy (RHA, CHA) and the presence of tumor contact as it may affect surgical planning
Body or tail
• CA: solid tumor contact of <180°
• CA: solid tumor contact of ≥180° without involvement of the aorta and with intact and uninvolved GDA
Vein • SMV/PV: solid tumor contact of ≥180°, contact of <180° with contour irregularity of the vein or thrombosis of the vein but with suitable vessel proximal and distal to the site of involvement allowing for safe and complete resection and vein reconstruction • Short-segment occlusion of SMV, PV, SMV-PV confluence amenable to resection & reconstruction • A TVI with the SMV/PV measuring ≥180° of the circumference of either vein’s wall, or short-segment occlusion of either vein with a normal vein above and below the obstruction amenable to reconstruction • SMV/PV: venous involvement demonstrating tumour abutment with or without impingement and narrowing of the lumen • SMV/PV: tumor contact/invasion of ≥180/occlusion, not exceeding the inferior border of the duodenum
• IVC: solid tumor contact • SMV/PV: encasement but without encasement of the nearby arteries • SMA, CA, CHA: no tumor contact/invasion
• SMV/PV: short segment venous occlusion resulting from either tumour thrombus or encasement but with suitable vessel proximal and distal to the area of vessel involvement, allowing for safe resection and reconstruction

NCCN, National Comprehensive Cancer Network; MDACC, The University of Texas MD Anderson Cancer Center; ACTO, Alliance for Clinical Trials in Oncology; AHPBA, American Hepato-Pancreato-Biliary Association; SSAT, Society for Surgery of the Alimentary Tract; SSO, Society for Surgical Oncology; JPS, Japan Pancreas Society; SMA, superior mesenteric artery; TVI, tumor-vessel interface; CHA, common hepatic artery; GDA, gastroduodenal artery; HA, hepatic artery; CA, celiac artery; PHA, proper hepatic artery; PV, portal vein; BR-A, borderline resectable with artery invasion; RHA, right hepatic artery; SMV, superior mesenteric vein; IVC, inferior vena cava.


Surgery

For resectable pancreatic cancer, radical resection of pancreatic cancer with adjuvant systemic chemotherapy remains the current standard treatment approach and is the only chance for long-term survival, even there is a high rate of recurrence (42). However, only small proportion of patients (10–20%) with pancreatic cancer are found to be resectable during the diagnosis. Tumors localised to pancreatic head or uncinate process can be treated with pancreaticoduodenectomy. Pancreaticoduodenectomy is the standard of surgical approach for pancreatic head cancer. Pancreaticoduodenectomy with vein resection and reconstruction is needed, in case of locally advanced tumor involving PV/SMV with adequate inflow and outflow veins for reconstruction, and the tumor does not involve SMA/CHA. Arterial resection with pancreatectomy remains controversial due to (I) increased surgical morbidity and mortality; and (II) unclear oncologic and survival benefit (43-45). Nevertheless, pancreatectomy with arterial resection continues to be performed at expert centers with acceptable reported outcomes in order to extend the indications and benefits of surgery to more patients. Importantly, combined venous/arterial resections should only be performed for extremely selected cases at expert centers. Its benefits and risks should be well balanced. Pancreatic cancer over the body and tail are treated with a radical distal pancreatectomy, often combined with splenectomy. Advances of pancreatectomy have been made over the past decade in the safety and efficacy of surgery and have resulted in surgical mortality of <3% and 5-year survival approaching 30% after resection with adjuvant systemic chemotherapy. The optimal number of lymph nodes for analysis and accurate tumor staging following standard pancreaticoduodenectomy and distal pancreatectomy were reported to be ≥15, and ≥20, respectively (46,47). Pancreaticoduodenectomy with extended lymphadenectomy of pancreatic head cancer does not offer any survival advantage over standard lymphadenectomy, but it increased surgical morbidity instead (48-50). A negative resection margin (R0) is one of the critical factors determining the oncological outcomes (51,52). Minimally invasive pancreaticoduodenectomy and distal pancreatectomy are being performed increasingly to treat pancreatic pathologies, including both robotic and laparoscopic approaches. Its indications have steadily broadened to encompass other conditions including pancreatic cancer, and it has gained increased acceptance and popularity. The advantages of minimally invasive approach include less blood loss and shorter hospital stay with comparable surgical mortality and morbidity to open approach (53-58). More and more studies showed similar oncological outcomes between minimally invasive and open approach when the techniques are applied in appropriate patients (53-58). The other potential advantage of minimally invasive approach may be earlier recovery with avoiding delay in adjuvant treatment. Larger sized and well-organized studies are needed to evaluate the surgical and oncological outcomes. However, the technique of minimally invasive approach of pancreatectomy is challenging and it is important to be performed by credentialed surgeons at experienced centers.


Adjuvant therapy after surgery for resectable pancreatic cancer

The European Study Group for Pancreatic Cancer-1 (ESPAC-1) (Neoptolemos et al.) trial was the 1st multicenter, randomized large-scale study to establish the use and benefit of adjuvant chemotherapy in resected pancreatic cancer (59,60). ESPAC-1 trial showed no survival benefit for adjuvant chemoradiotherapy but revealed a potential benefit for adjuvant fluorouracil plus folinic acid. ESPAC-3 trial (Neoptolemos et al.) compared adjuvant gemcitabine (n=537) with fluorouracil plus folinic acid (n=551) in patients with resected pancreatic cancer (61). There were no significant differences in overall survival, progression-free survival, global quality-of-life scores between both groups. However, there were significantly fewer treatment-related serious adverse events in gemcitabine group. Ninety-seven treatment-related serious adverse events in 77 patients (14%) in fluorouracil plus folinic acid group were reported while 52 treatment-related serious adverse events in 40 patients (7.5%) in gemcitabine group were reported. CONKO-001 (Charité Onkologie 001) compared adjuvant gemcitabine (n=179) with no adjuvant treatment (control) (n=175) in patients with resectable pancreatic cancer (62). After a median follow-up of 53 months, disease recurrences were happened in 133 patients (74%) in gemcitabine group and 161 patients (92%) in control group. Gemcitabine group had significantly better disease-free survival than control group (median, 13.4 vs. 6.9 months). Estimated 3-year/5-year disease-free survival rate was 23.5%/16.5% in gemcitabine group, and 7.5%/5.5% in control group, respectively. There was no difference in overall survival between gemcitabine group and control group (median survival, 22.1 vs. 20.2 months; estimated 3-year survival rate, 34% vs. 20.5%; 5-year survival rate, 22.5% vs. 11.5%). Grade 3/4 adverse events rarely occurred. There was no difference in quality of life. In their follow-up report, at a median follow-up of 136 months, gemcitabine group had significantly longer disease-free survival than control group (median, 13.4 vs. 6.7 months) (63). Adjuvant gemcitabine treatment was shown to have better overall survival than control group (5-year overall survival rate, 20.7% vs. 10.4%; 10-year overall survival rate, 12.2% vs. 7.7%). A multicentre randomized trial ESPAC-4 (Neoptolemos et al.) compared the uses of adjuvant gemcitabine plus capecitabine (n=364) with monotherapy gemcitabine (n=366) in patients with resected pancreatic cancer (64). After a median follow-up of 43.2 months, gemcitabine plus capecitabine group had significantly better overall survival than gemcitabine group (median, 28.0 vs. 25.5 months). Estimated 5-year overall survival rate was 28.8% for gemcitabine plus capecitabine group and 16.3% for gemcitabine group. Six hundred and eight grade 3/4 adverse events were reported by 63% of the patients in gemcitabine plus capecitabine group while 481 grade 3/4 adverse events were reported by 53.6% of the patients in gemcitabine group. Adjuvant gemcitabine plus capecitabine group was found to have a 21% reduction of mortality following recurrence compared with gemcitabine group (65). A multicentre randomized trial by Canadian Cancer Trials Group and the Unicancer-GI–PRODIGE Group (Conroy et al.) compared adjuvant modified-FOLFIRINOX (fluorouracil, oxaliplatin, irinotecan, leucovorin) (n=247) with monotherapy gemcitabine (n=246) in patients with resected pancreatic cancer (66). At a median follow-up of 33.6 months, modified-FOLFIRINOX group had significantly better disease-free survival than gemcitabine group (median, 21.6 vs. 12.8 months). The 3-year disease-free survival rate was 39.7% and 21.4%, respectively. Modified-FOLFIRINOX group also had a significantly better overall survival (median, 54.4 vs. 35.0 months; 3-year overall survival rate, 63.4% vs. 48.6%). Grade 3/4 adverse events happened in 75.9% of the patients in modified-FOLFIRINOX group and in 52.9% of the patients in gemcitabine group. Mortality due to adverse event of interstitial pneumonitis was reported in one patient in gemcitabine group. Later, the follow report of 5-year indicated that adjuvant treatment with modified FOLFIRINOX had significantly better survival than gemcitabine in patients with resected pancreatic cancer (67). At a median follow-up of 69.7 months, there were 367 disease-free survival events. Modified-FOLFIRINOX group had significantly better disease-free survival than gemcitabine group (median, 21.4 vs. 12.8 months; 5-year disease-free survival rate, 26.1% vs. 19.0%). The overall survival was also significantly better in modified-FOLFIRINOX group (median, 53.5 vs. 35.5 months; 5-year overall survival rate, 43.2% vs. 31.4%). Modified-FOLFIRINOX group had significantly better metastasis-free survival than gemcitabine group (median, 29.4 vs. 17.7 months) and had significantly better cancer-specific survival (median, 54.7 vs. 36.3 months).

The current evidence is based on a limited number of well-designed randomized controlled trials for each treatment regimen. The current recommended adjuvant chemotherapy for resected pancreatic cancer is either modified FOLFIRINOX for patients with good well-being and functional status, or gemcitabine plus capecitabine or monotherapy with gemcitabine for patients with suboptimal functional status.


Neoadjuvant therapy for resectable pancreatic cancer and borderline resectable pancreatic cancer

The potential advantages of neoadjuvant therapy before surgery include: (I) early treatment of micrometastasis; (II) decreased lymph node involvement; (III) decreased the difficulties of surgery after downsizing of the tumor; (IV) improved R0 resection rates; and (V) selection of patients with favorable tumor biology. In addition, a significant proportion of patients after pancreatectomy cannot receive adjuvant therapy as planned due to surgical complications and prolong hospital stay (68,69). The benefit of neoadjuvant therapy will give chance of these patients for perioperative systemic treatment. The potential drawbacks of neoadjuvant therapy is the disease progression to an unresectable stage during the period of receiving neoadjuvant treatment, and the patient will lose the chance of curative resection. Neoadjuvant therapy has been tested in resectable pancreatic cancer and borderline resectable pancreatic cancer. Due to the great variations of the case selection, bias in the studies, variations in treatment regimens, and without comparison with upfront surgery groups, comparison of these studies with a valid conclusion are very difficult.

SWOG/NCI S1505 (Sohal et al.) randomized patients with resectable pancreatic cancer to neoadjuvant FOLFIRINOX group (n=55) and gemcitabine with nab-paclitaxel group (n=47). SWOG/NCI S1505 showed 2-year overall survival rate of 47% and 48%, respectively (70). Among the 102 patients, preoperative chemotherapy was completed in 84% and 85% of patients, respectively; 73% and 70% of patients underwent resection, and 49% and 40% completed all treatment, respectively. SWOG/NCI S1505 did not show an improved overall survival with perioperative chemotherapy, compared with historical data from adjuvant treatment studies in resectable pancreatic cancer. PANACHE01-PRODIGE48 evaluated the safety and efficacy of two neoadjuvant regimens of modified FOLFIRINOX (n=70) or FOLFOX (fluorouracil, oxaliplatin, leucovorin) (n=50) relative to the current standard treatment (upfront surgery and adjuvant chemotherapy) (n=26) in patients with resectable pancreatic cancer. There was no significant differences in 1-year survival rates (modified FOLFIRINOX—84.1%, FOLFOX—71.8%, and upfront surgery—80.8%) (71). There was a 10% increase in 1-year event-free survival rate (51.4% vs. 41.7%) and a 3-month improvement in event-free survival (median, 12.4 vs. 9.2 months) between neoadjuvant modified FOLFIRINOX group and upfront surgery group. NEONAX (Seufferlein et al.) studied the outcome of patients with resectable pancreatic cancer receiving gemcitabine plus nab-paclitaxel, either perioperatively (two pre-operative and four post-operative cycles) or adjuvant (six cycles) (72). The primary endpoint of disease-free survival rate (intention-to-treat population) of 55% at 18 months was not reached in both groups (33.3% vs. 41.4%). The secondary endpoint of median overall survival (intention-to-treat population) numerically favored the neoadjuvant group (25.5 vs. 16.7 months). There was a difference in chemotherapy exposure with 90% in perioperative chemotherapy group completing pre-operative chemotherapy and 58% of patients starting adjuvant chemotherapy in adjuvant chemotherapy group.

In the RCT PREOPANC (Versteijne et al.) trial, 246 patients with resectable pancreatic cancer and borderline resectable pancreatic cancer were randomized to neoadjuvant chemoradiotherapy (n=119) (gemcitabine for 3 courses, the 2nd combined with 15×2.4 Gy radiotherapy, followed by surgery and adjuvant gemcitabine for 4 courses) and upfront surgery (n=127) (upfront surgery with adjuvant gemcitabine for 6 courses) (73,74). Both groups have similar resection rate (61% vs. 72%). Neoadjuvant chemoradiotherapy did not increase the incidence of surgical morbidity or mortality (75). Neoadjuvant group had a significantly better R0 resection rate (72% vs. 43%). In neoadjuvant group, 55 (81%) of the 68 patients started adjuvant chemotherapy and 34 patients (62%) finished all cycles. In upfront surgery group, 65 (79%) of the 82 patients started adjuvant chemotherapy and 35 patients (54%) completed all cycles. By intention to treat analysis, 46% of the 119 patients in the neoadjuvant chemoradiotherapy group started adjuvant chemotherapy, while 51% of the 127 patients in the upfront surgery group started adjuvant chemotherapy. The proportion of patients with serious adverse events was similar (52% vs. 41%). Neoadjuvant group had a significant better disease-free survival and locoregional failure-free interval. Neoadjuvant group also had a significant lower rates of pathologic lymph nodes, venous invasion and perineural invasion. After a median follow-up of 59 months, mortality was happened in 93 (78%) in neoadjuvant group and 117 (92%) in upfront surgery group. By intention to treat analysis, the median overall survival had no significant difference (15.7 vs. 14.3 months). Neoadjuvant group and upfront surgery group had an estimated 3-/5-year survival rates of 27.7%/20.5%, and 16.5%/6.5%, respectively. The limitation of this study was the use of adjuvant gemcitabine monotherapy, which is not considered as standard treatment regimen nowadays.

In phase 2 RCT ESPAC5 (Ghaneh et al.), 90 patients with borderline resectable pancreatic cancer were randomly assigned to immediate surgery (n=33), and three different types of short-course neoadjuvant therapy—gemcitabine plus capecitabine (n=20), FOLFIRINOX (n=20), and capecitabine-based chemoradiation (n=17) (76). Four patients were excluded from intention-to-treat analysis. No significant difference in resection rates (68% vs. 55%) and R0 resection rates (14% vs. 23%) between immediate surgery group and neoadjuvant therapy group. Surgical complication rate and 30-day mortality was 43% and 0% of patients who underwent surgery, respectively. Six (13%) of 46 had partial response among the 46 (84%) of 55 patients having neoadjuvant therapy available for restaging. At median follow-up of 12.2 months, there was significant difference in 1-year overall survival rate (39% vs. 78% vs. 84% vs. 60%) for immediate surgery, gemcitabine plus capecitabine, FOLFIRINOX, and capecitabine-based chemoradiotherapy groups. There was significant difference in 1-year disease-free survival from surgery (33% vs. 59%) for immediate surgery and combined neoadjuvant therapies. Two patients in immediate surgery group and 1 patient in FOLFIRINOX group had local recurrence. Nineteen (24%) of 78 patients (immediate surgery group, n=2 and combined neoadjuvant therapy groups, n=17) had a grade ≥3 adverse event. The most common adverse events were neutropenia, infection, and hyperglycaemia. ESPAC5 supported the use of short-course neoadjuvant chemotherapy (either gemcitabine plus capecitabine or FOLFIRINOX) for patients with borderline resectable pancreatic cancer.

Based on the current evidence, there is increasing evidence showing benefits of neoadjuvant therapy in borderline resectable pancreatic cancer. The optimal neoadjuvant treatment regimen was not certain yet. The use of neoadjuvant therapy in resectable pancreatic cancer has been restricted to clinical trials, as the data are limited and no clear benefits have yet been shown in resectable pancreatic cancer.


Systemic therapy for metastatic pancreatic cancer

At the time of diagnosis, approximately 50% of patients with pancreatic cancer present with distant metastases. Systemic therapy remains the primary treatment modality aiming at palliating symptoms and prolonging survival.

Von Hoff et al. randomized patients (Karnofsky performance-status score of ≥70) with metastatic pancreatic cancer to gemcitabine plus nab-paclitaxel (n=431) or monotherapy gemcitabine (n=430) (77). The median overall survival was significantly better in the gemcitabine plus nab-paclitaxel group (8.5 vs. 6.7 months). The 1-year survival rate (35% vs. 22%) and 2-year survival rate (9% vs. 4%) were significantly better in gemcitabine plus nab-paclitaxel group than gemcitabine group. Gemcitabine plus nab-paclitaxel group also had significantly better median progression-free survival (5.5 vs. 3.7 months), and response rate (23% vs. 7%). However, rates of grade 3/4 adverse events of peripheral neuropathy (17% vs. 1%), fatigue (17% vs. 7%) and neutropenia (38% vs. 27%) were increased.

Conroy et al. randomized patients (Eastern Cooperative Oncology Group performance status score of 0 or 1) with metastatic pancreatic cancer to FOLFIRINOX (n=171) or monotherapy gemcitabine (n=171) (78). As compared with gemcitabine, FOLFIRINOX group had a significant better overall survival (median, 11.1 vs. 6.8 months), progression-free survival (median, 6.4 vs. 3.3 months), and objective response rate (31.6% vs. 9.4%). Grade 3/4 adverse events of neutropenia, neutropenic fever, thrombocytopenia, sensory neuropathy and diarrhea were significantly higher in FOLFIRINOX group, whereas grade 3/4 adverse event of elevated alanine aminotransferase levels was significantly higher in gemcitabine group.

Adverse events of FOLFIRINOX and gemcitabine-plus-nab-paclitaxel often lead to dose-reductions and limit their long-term use. Maintenance therapies aim to prolong progression-free and overall survival without compromising quality of life. Mutations in breast- and ovarian-cancer-predisposing genes (BRCA1 and, especially, BRCA2) are also associated with increased risk of pancreatic cancer, and are present in 5–9% of patients with pancreatic cancer. The protein products of BRCA1 or BRCA2 genes are responsible in DNA damage response and repair of double-strand DNA breaks through homologous recombination. Golan et al. randomized metastatic pancreatic cancer patients with mutations in BRCA1 or BRCA2 genes that had not progressed during 1st-line platinum-based chemotherapy to receive maintenance poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor—olaparib (n=92) or placebo (n=62) (79). The olaparib group had a significantly better progression-free survival than control group (median, 7.4 vs. 3.8 months). An interim analysis of median overall survival showed no difference between the 2 groups (18.9 vs. 18.1 months). The incidence of grade 3/4 adverse events in olaparib group and control group was 40% and 23%, respectively, and 5% and 2% of the patients were discontinued from the trial intervention due to adverse events, respectively.

Nanoliposomal irinotecan (nal-IRI) is a novel formulation of irinotecan, in which encapsulating drug molecules within long-circulating liposome-based nanoparticles with resulting favorable pharmacokinetic and biodistribution characteristics with a higher intra-tumoural levels of both irinotecan and SN-38 (an active metabolite of irinotecan) than traditional irinotecan. NAPOLI-1 Study Group (Wang-Gillam et al.) randomized patients with metastatic pancreatic cancer previously treated with gemcitabine-based therapy to either nal-IRI plus 5-fluorouracil and leucovorin (5-FU/LV) (n=117), monotherapy nal-IRI (n=151), or 5-FU/LV (n=149) (80,81). After 313 events, nal-IRI + 5-FU/LV group had a significantly better median overall survival better than 5-FU/LV group (6.1 vs. 4.2 months), while monotherapy nal-IRI group and 5-FU/LV group had similar median overall survival (4.9 vs. 4.2 months). The most frequently occurred grade 3/4 adverse events in nal-IRI + 5-FU/LV group were neutropenia (27%), fatigue (14%), diarrhoea (13%) and vomiting (11%). The survival benefits of nal-IRI + 5-FU/LV vs. 5-FU/LV were maintained over an extended follow-up. Estimated 1-year overall survival rates of nal-IRI + 5-FU/LV group and 5-FU/LV group were 26% and 16%, respectively.

The recommended first line chemotherapy for patients with metastatic pancreatic cancer is either FOLFIRINOX and gemcitabine plus nab-paclitaxel for patients having good functional status, or monotherapy gemcitabine for patients having suboptimal functional status. The evidence is based on a limited number of well-designed randomized controlled trials for each treatment regimen. Although modified FOLFIRINOX regimens have not been compared with traditional FOLFIRINOX in prospective randomized trials, it appears to be less toxic and data from retrospective studies showed similar outcomes (82,83). Nal-IRI + 5-FU/LV is a second line option of treatment for patients with metastatic pancreatic cancer previously treated with gemcitabine-based therapy. Recently, olaparib has been demonstrated to improve progression-free survival when used as a maintenance treatment in the subgroup of metastatic pancreatic cancer patients with BRCA1 or BRCA2 genes mutations had not progressed for at least 16 weeks during 1st-line platinum-based chemotherapy. The decision to continue treatment with chemotherapy or change to maintenance therapy with olaparib should be based on a discussion between the patient and the oncologist, and the consideration of the response to chemotherapy and the tolerance and adverse events associated with chemotherapy.

Recently, the Food and Drug Administration (FDA) approved the use of pembrolizumab (an anti-programmed-death receptor 1, anti-PD-1) for patients with any microsatellite instability (MSI) high tumors, including pancreatic cancer. Phase 2 study KEYNOTE-158 recruited 233 patients with microsatellite instability-high (MSI-H)/DNA mismatch repair (dMMR) noncolorectal cancer (84). Among the study group, 22 patients (9.4%) had pancreatic cancer. Overall, 151 patients (64.8%) and 34 (14.6%) had treatment-related adverse events and grade 3 to 5 treatment-related adverse events, respectively. Eighteen patients (7.7%) had serious adverse events, and 22 (9.4%) discontinued trial intervention due to treatment-related adverse events. One patient (4.5%) and 3 patients (13.6%) with pancreatic cancer had complete response and partial response, respectively. Median progression-free survival, overall survival and duration of response were 2.1, 4 and 13.4 months, respectively. Follow up study showed that ≥3-year overall survival rate and ≥3-year progression free survival rate were 22.7% and 0%, respectively (85). Evidence about pembrolizumab usage for pancreatic cancer is still limited. More immunotherapy data in in pancreatic cancer treatment is pending.

At present, tyrosine receptor kinase (TRK) inhibitors targeting neurotrophic tyrosine receptor kinase (NTRK) gene fusions are among the first “tumor agnostic” drugs approved for pan-cancer use. Recently, FDA also approved the use of larotrectinib or entrectinib for patients with any tumors harboring a NTRK gene fusion in a second-line setting, including pancreatic cancer, in 2018 and 2019, respectively (86-89). However, the data for pancreatic cancer is still very limited.

In Drilon et al., a total of 55 patients (aged 4 months–76 years old) harboring 17 unique TRK fusion-positive tumor types were recruited and treated (86). The overall response rate according to independent review and investigator assessment was 75% and 80%, respectively (87). At 1 year, 71% and 55% of the responses were ongoing and maintained progression-free, respectively. The median duration of response and progression-free survival had not been reached. At a median follow-up of 9.4 months, 86% of the patients with a response were continuing treatment or had undergone operation with curative intent. Adverse events were mainly grade 1, and no grade 3/4 adverse event that was considered by the investigators to be related to larotrectinib occurred in >5% of patients. No patient discontinued larotrectinib owing to treatment-related adverse events. In a pooled analysis of three phase 1/2 clinical trials, 159 patients with TRK fusion-positive cancers were recruited and received larotrectinib. The objective response rate according to investigator assessment was 79% (complete responses—16%). In a safety population of 260 patients treated regardless of TRK fusion status, the most common grade 3/4 larotrectinib-related adverse events were increased alanine aminotransferase level (3%), decreased neutrophil count (2%) and anaemia (2%). The most common larotrectinib-related serious adverse events were increased aspartate aminotransferase level (<1%), increased alanine aminotransferase level (<1%), and nausea (<1%). No treatment-related deaths occurred.

In a pooled analysis of three phase 1/2 clinical studies, a durable overall response rate of 57%, including a partial response rate of 50% and complete response rate of 7%, among 54 entrectinib-treated patients with 10 different tumor types and 19 different histologies harboring an NTRK fusion recruited after a median follow-up of 12.9 months (88,89). Median duration of response was 10 months. The most common serious treatment-related adverse events were nervous system disorders [3 of 68 patients (4%) in the NTRK fusion-positive safety population and in 10 of 355 patients (3%) in the overall safety-evaluable population]. The most common treatment-related grade 3/4 adverse events in both safety populations were increased weight [7 of 68 patients (10%) and 18 of 355 patients (5%)], and anaemia [8 (12%) and 16 (5%)]. No treatment-related mortality happened.


Palliation of complications of advanced pancreatic cancer

For patients with unresectable advanced pancreatic cancers, biliary obstruction, duodenal obstruction, and pain are the possible complications that needed to be managed. An appropriate modality of treatment should be chosen with consideration of the patient’s physical and functional status, the expected life expectancy, expertise availability, disease status and anatomical configuration. Psychosocial support to the patients and family is also important for patients with advanced disease.


Limitations

The main limitation of this review is that it is a narrative review rather than a systematic review. Currently, the treatment regimens and end-point measurements were too heterogenous to perform a systematic review. The number of studies for each treatment was small also. In future, if there is a greater number of randomized trials and well-designed nonrandomized comparative studies, systematic review with meta-analysis about pancreatic cancer treatment will be more useful.


Conclusions

Pancreatic cancer is a challenging disease for management. Radical surgery itself is not enough for prolong survival. The improvement of chemotherapy, target agents and immunotherapy with multidisciplinary approach for pancreatic cancer patients will be the only solution for improvement of their quality of life and survival outcomes.

The adoption of adjuvant treatment for operable disease and systemic treatment for inoperable disease is based on the published randomized trials. The adoption of neoadjuvant treatment and new treatment modalities such as target agents and immunotherapy is still limited by small number of studies with favorable outcomes. In the coming time, more well-designed large-scale studies are needed to give more evidence for the literature.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://cco.amegroups.com/article/view/10.21037/cco-23-94/rc

Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-23-94/prf

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-23-94/coif). E.C.H.L. serves as an unpaid editorial board member of Chinese Clinical Oncology from December 2022 to November 2024. The other author has 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. Schawkat K, Manning MA, Glickman JN, et al. Pancreatic Ductal Adenocarcinoma and Its Variants: Pearls and Perils. Radiographics 2020;40:1219-39. [Crossref] [PubMed]
  2. Seufferlein T, Kestler A. Exocrine pancreatic cancer - what is new in the update of the S3 guideline? Dtsch Med Wochenschr 2023;148:737-43. [PubMed]
  3. Schober M, Javed MA, Beyer G, et al. New Advances in the Treatment of Metastatic Pancreatic Cancer. Digestion 2015;92:175-84. [Crossref] [PubMed]
  4. Rustgi SD, Hilfrank KJ, Kastrinos F. Familial Predisposition and Genetic Risk Factors Associated with Pancreatic Cancer. Gastrointest Endosc Clin N Am 2022;32:1-12. [Crossref] [PubMed]
  5. McGuigan A, Kelly P, Turkington RC, et al. Pancreatic cancer: A review of clinical diagnosis, epidemiology, treatment and outcomes. World J Gastroenterol 2018;24:4846-61. [Crossref] [PubMed]
  6. Park W, Chawla A, O'Reilly EM. Pancreatic Cancer: A Review. JAMA 2021;326:851-62. [Crossref] [PubMed]
  7. Shinde RS, Bhandare M, Chaudhari V, et al. Cutting-edge strategies for borderline resectable pancreatic cancer. Ann Gastroenterol Surg 2019;3:368-72. [Crossref] [PubMed]
  8. Luo G, Zhang Y, Guo P, et al. Global Patterns and Trends in Pancreatic Cancer Incidence: Age, Period, and Birth Cohort Analysis. Pancreas 2019;48:199-208. [Crossref] [PubMed]
  9. Huang J, Lok V, Ngai CH, et al. Worldwide Burden of, Risk Factors for, and Trends in Pancreatic Cancer. Gastroenterology 2021;160:744-54. [Crossref] [PubMed]
  10. Hu JX, Zhao CF, Chen WB, et al. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J Gastroenterol 2021;27:4298-321. [Crossref] [PubMed]
  11. Afghani E, Klein AP. Pancreatic Adenocarcinoma: Trends in Epidemiology, Risk Factors, and Outcomes. Hematol Oncol Clin North Am 2022;36:879-95. [Crossref] [PubMed]
  12. Bhoopathi P, Mannangatti P, Das SK, et al. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023;159:285-341. [Crossref] [PubMed]
  13. Goodarzi E, Dehkordi AH, Beiranvand R, et al. Epidemiology of the Incidence and Mortality of Pancreas Cancer and its Relationship with the Human Development Index (HDI) in the World: An Ecological Study in 2018. Curr Pharm Des 2020;26:5163-73. [Crossref] [PubMed]
  14. Klein AP. Pancreatic cancer epidemiology: understanding the role of lifestyle and inherited risk factors. Nat Rev Gastroenterol Hepatol 2021;18:493-502. [Crossref] [PubMed]
  15. Maisonneuve P. Epidemiology and burden of pancreatic cancer. Presse Med 2019;48:e113-23. [Crossref] [PubMed]
  16. Ohmoto A, Yachida S, Morizane C. Genomic Features and Clinical Management of Patients with Hereditary Pancreatic Cancer Syndromes and Familial Pancreatic Cancer. Int J Mol Sci 2019;20:561. [Crossref] [PubMed]
  17. Llach J, Carballal S, Moreira L. Familial Pancreatic Cancer: Current Perspectives. Cancer Manag Res 2020;12:743-58. [Crossref] [PubMed]
  18. Cai J, Chen H, Lu M, et al. Advances in the epidemiology of pancreatic cancer: Trends, risk factors, screening, and prognosis. Cancer Lett 2021;520:1-11. [Crossref] [PubMed]
  19. Olakowski M, Bułdak Ł. Modifiable and Non-Modifiable Risk Factors for the Development of Non-Hereditary Pancreatic Cancer. Medicina (Kaunas) 2022;58:978. [Crossref] [PubMed]
  20. Mizrahi JD, Surana R, Valle JW, et al. Pancreatic cancer. Lancet 2020;395:2008-20. [Crossref] [PubMed]
  21. Khadka R, Tian W, Hao X, et al. Risk factor, early diagnosis and overall survival on outcome of association between pancreatic cancer and diabetes mellitus: Changes and advances, a review. Int J Surg 2018;52:342-6. [Crossref] [PubMed]
  22. Kulkarni NM, Hough DM, Tolat PP, et al. Pancreatic adenocarcinoma: cross-sectional imaging techniques. Abdom Radiol (NY) 2018;43:253-63. [Crossref] [PubMed]
  23. Noda Y, Kawai N, Kaga T, et al. Vascular involvement and resectability of pancreatic ductal adenocarcinoma on contrast-enhanced MRI: comparison with pancreatic protocol CT. Abdom Radiol (NY) 2022;47:2835-44. [Crossref] [PubMed]
  24. Jajodia A, Wang A, Alabousi M, et al. MRI vs. CT for pancreatic adenocarcinoma vascular invasion: comparative diagnostic test accuracy systematic review and meta-analysis. Eur Radiol 2023;33:6883-91. [Crossref] [PubMed]
  25. Jha P, Yeh BM, Zagoria R, et al. The Role of MR Imaging in Pancreatic Cancer. Magn Reson Imaging Clin N Am 2018;26:363-73. [Crossref] [PubMed]
  26. Alabousi M, McInnes MD, Salameh JP, et al. MRI vs. CT for the Detection of Liver Metastases in Patients With Pancreatic Carcinoma: A Comparative Diagnostic Test Accuracy Systematic Review and Meta-Analysis. J Magn Reson Imaging 2021;53:38-48. [Crossref] [PubMed]
  27. Ghidini M, Vuozzo M, Galassi B, et al. The Role of Positron Emission Tomography/Computed Tomography (PET/CT) for Staging and Disease Response Assessment in Localized and Locally Advanced Pancreatic Cancer. Cancers (Basel) 2021;13:4155. [Crossref] [PubMed]
  28. Kitano M, Yoshida T, Itonaga M, et al. Impact of endoscopic ultrasonography on diagnosis of pancreatic cancer. J Gastroenterol 2019;54:19-32. [Crossref] [PubMed]
  29. Yousaf MN, Chaudhary FS, Ehsan A, et al. Endoscopic ultrasound (EUS) and the management of pancreatic cancer. BMJ Open Gastroenterol 2020;7:e000408. [Crossref] [PubMed]
  30. 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]
  31. Scarà S, Bottoni P, Scatena R. CA 19-9: Biochemical and Clinical Aspects. Adv Exp Med Biol 2015;867:247-60. [Crossref] [PubMed]
  32. Yang J, Xu R, Wang C, et al. Early screening and diagnosis strategies of pancreatic cancer: a comprehensive review. Cancer Commun (Lond) 2021;41:1257-74. [Crossref] [PubMed]
  33. Daoud AZ, Mulholland EJ, Cole G, et al. MicroRNAs in Pancreatic Cancer: biomarkers, prognostic, and therapeutic modulators. BMC Cancer 2019;19:1130. [Crossref] [PubMed]
  34. Chun YS, Pawlik TM, Vauthey JN. 8th Edition of the AJCC Cancer Staging Manual: Pancreas and Hepatobiliary Cancers. Ann Surg Oncol 2018;25:845-7.
  35. Tempero MA, Malafa MP, Chiorean EG, et al. Pancreatic Adenocarcinoma, Version 1.2019. J Natl Compr Canc Netw 2019;17:202-10. [Crossref] [PubMed]
  36. Kulkarni NM, Soloff EV, Tolat PP, et al. White paper on pancreatic ductal adenocarcinoma from society of abdominal radiology's disease-focused panel for pancreatic ductal adenocarcinoma: Part I, AJCC staging system, NCCN guidelines, and borderline resectable disease. Abdom Radiol (NY) 2020;45:716-28. [Crossref] [PubMed]
  37. Varadhachary GR, Tamm EP, Abbruzzese JL, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol 2006;13:1035-46. [Crossref] [PubMed]
  38. Katz MH, Pisters PW, Evans DB, et al. Borderline resectable pancreatic cancer: the importance of this emerging stage of disease. J Am Coll Surg 2008;206:833-46; discussion 846-8. [Crossref] [PubMed]
  39. Katz MH, Marsh R, Herman JM, et al. Borderline resectable pancreatic cancer: need for standardization and methods for optimal clinical trial design. Ann Surg Oncol 2013;20:2787-95. [Crossref] [PubMed]
  40. Callery MP, Chang KJ, Fishman EK, et al. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol 2009;16:1727-33. [Crossref] [PubMed]
  41. Japan Pancreas Society. Classification of pancreatic carcinoma. 4th English edition. Tokyo: Kanehara & Co., Ltd.; 2017. Available online: http://www.suizou.org/pdf/Classification_of_Pancreatic_Carcinoma_4th_Engl_ed.pdf
  42. Lau WY, Lai EC. Development and controversies of adjuvant therapy for pancreatic cancer. Hepatobiliary Pancreat Dis Int 2008;7:121-5. [PubMed]
  43. Karunakaran M, Barreto SG. Surgery for pancreatic cancer: current controversies and challenges. Future Oncol 2021;17:5135-62. [Crossref] [PubMed]
  44. Zettervall SL, Ju T, Holzmacher JL, et al. Arterial, but Not Venous, Reconstruction Increases 30-Day Morbidity and Mortality in Pancreaticoduodenectomy. J Gastrointest Surg 2020;24:578-84. [Crossref] [PubMed]
  45. Kimura K, Amano R, Tauchi J, et al. Pancreaticoduodenectomy with celiac artery resection (PD-CAR) for unresctable locally advanced pancreatic ductal adenocarcinoma. Langenbecks Arch Surg 2023;408:174. [Crossref] [PubMed]
  46. Slidell MB, Chang DC, Cameron JL, et al. Impact of total lymph node count and lymph node ratio on staging and survival after pancreatectomy for pancreatic adenocarcinoma: a large, population-based analysis. Ann Surg Oncol 2008;15:165-74. [Crossref] [PubMed]
  47. Malleo G, Maggino L, Ferrone CR, et al. Number of Examined Lymph Nodes and Nodal Status Assessment in Distal Pancreatectomy for Body/Tail Ductal Adenocarcinoma. Ann Surg 2019;270:1138-46. [Crossref] [PubMed]
  48. Jang JY, Kang MJ, Heo JS, et al. A prospective randomized controlled study comparing outcomes of standard resection and extended resection, including dissection of the nerve plexus and various lymph nodes, in patients with pancreatic head cancer. Ann Surg 2014;259:656-64. [Crossref] [PubMed]
  49. Orci LA, Meyer J, Combescure C, et al. A meta-analysis of extended versus standard lymphadenectomy in patients undergoing pancreatoduodenectomy for pancreatic adenocarcinoma. HPB (Oxford) 2015;17:565-72. [Crossref] [PubMed]
  50. Wang W, He Y, Wu L, et al. Efficacy of extended versus standard lymphadenectomy in pancreatoduodenectomy for pancreatic head adenocarcinoma. An update meta-analysis. Pancreatology 2019;19:1074-80. [Crossref] [PubMed]
  51. Howard TJ, Krug JE, Yu J, et al. A margin-negative R0 resection accomplished with minimal postoperative complications is the surgeon's contribution to long-term survival in pancreatic cancer. J Gastrointest Surg 2006;10:1338-45; discussion 1345-6. [Crossref] [PubMed]
  52. Kato K, Yamada S, Sugimoto H, et al. Prognostic factors for survival after extended pancreatectomy for pancreatic head cancer: influence of resection margin status on survival. Pancreas 2009;38:605-12. [Crossref] [PubMed]
  53. Qin R, Kendrick ML, Wolfgang CL, et al. International expert consensus on laparoscopic pancreaticoduodenectomy. Hepatobiliary Surg Nutr 2020;9:464-83. [Crossref] [PubMed]
  54. Magge D, Zureikat A, Hogg M, et al. Minimally Invasive Approaches to Pancreatic Surgery. Surg Oncol Clin N Am 2016;25:273-86. [Crossref] [PubMed]
  55. Broucek JR, Sanford D, Stauffer JA, et al. Minimally Invasive Approaches to Pancreatic Cancer. Surg Oncol Clin N Am 2019;28:255-72. [Crossref] [PubMed]
  56. Lai EC, Yang GP, Tang CN. Robot-assisted laparoscopic pancreaticoduodenectomy versus open pancreaticoduodenectomy--a comparative study. Int J Surg 2012;10:475-9. [Crossref] [PubMed]
  57. Lai EC, Tang CN. Current status of robot-assisted laparoscopic pancreaticoduodenectomy and distal pancreatectomy: a comprehensive review. Asian J Endosc Surg 2013;6:158-64. [Crossref] [PubMed]
  58. Lai EC, Tang CN. Robotic distal pancreatectomy versus conventional laparoscopic distal pancreatectomy: a comparative study for short-term outcomes. Front Med 2015;9:356-60. [Crossref] [PubMed]
  59. 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]
  60. 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]
  61. 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]
  62. Oettle H, Post S, Neuhaus P, et al. Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA 2007;297:267-77. [Crossref] [PubMed]
  63. 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]
  64. 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]
  65. Jones RP, Psarelli EE, Jackson R, et al. Patterns of Recurrence After Resection of Pancreatic Ductal Adenocarcinoma: A Secondary Analysis of the ESPAC-4 Randomized Adjuvant Chemotherapy Trial. JAMA Surg 2019;154:1038-48. [Crossref] [PubMed]
  66. 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]
  67. Conroy T, Castan F, Lopez A, et al. Five-Year Outcomes of FOLFIRINOX vs Gemcitabine as Adjuvant Therapy for Pancreatic Cancer: A Randomized Clinical Trial. JAMA Oncol 2022;8:1571-8. [Crossref] [PubMed]
  68. Yang SQ, Zou RQ, Dai YS, et al. Comparison of the upfront surgery and neoadjuvant therapy in resectable and borderline resectable pancreatic cancer: an updated systematic review and meta-analysis. Updates Surg 2024;76:1-15. [Crossref] [PubMed]
  69. Kwaśniewska D, Fudalej M, Nurzyński P, et al. How A Patient with Resectable or Borderline Resectable Pancreatic Cancer should Be Treated-A Comprehensive Review. Cancers (Basel) 2023;15:4275. [Crossref] [PubMed]
  70. 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. [Crossref] [PubMed]
  71. 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]
  72. Seufferlein T, Uhl W, Kornmann M, et al. Perioperative or only adjuvant gemcitabine plus nab-paclitaxel for resectable pancreatic cancer (NEONAX)-a randomized phase II trial of the AIO pancreatic cancer group. Ann Oncol 2023;34:91-100. [Crossref] [PubMed]
  73. Versteijne E, Suker M, Groothuis K, 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 2020;38:1763-73. [Crossref] [PubMed]
  74. 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]
  75. van Dongen JC, Suker M, Versteijne E, et al. Surgical Complications in a Multicenter Randomized Trial Comparing Preoperative Chemoradiotherapy and Immediate Surgery in Patients With Resectable and Borderline Resectable Pancreatic Cancer (PREOPANC Trial). Ann Surg 2022;275:979-84. [Crossref] [PubMed]
  76. Ghaneh P, Palmer D, Cicconi S, et al. Immediate surgery compared with short-course neoadjuvant gemcitabine plus capecitabine, FOLFIRINOX, or chemoradiotherapy in patients with borderline resectable pancreatic cancer (ESPAC5): a four-arm, multicentre, randomised, phase 2 trial. Lancet Gastroenterol Hepatol 2023;8:157-68. [Crossref] [PubMed]
  77. Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691-703. [Crossref] [PubMed]
  78. Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817-25. [Crossref] [PubMed]
  79. Golan T, Hammel P, Reni M, et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N Engl J Med 2019;381:317-27. [Crossref] [PubMed]
  80. Wang-Gillam A, Li CP, Bodoky G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet 2016;387:545-57. [Crossref] [PubMed]
  81. Wang-Gillam A, Hubner RA, Siveke JT, et al. NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. Eur J Cancer 2019;108:78-87. [Crossref] [PubMed]
  82. Stein SM, James ES, Deng Y, et al. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br J Cancer 2016;114:737-43. [Crossref] [PubMed]
  83. Nakazawa J, Tsuruta N, Shimokawa M, et al. Multicenter Retrospective Analysis of Original versus Modified FOLFIRINOX in Metastatic Pancreatic Cancer: Results of the NAPOLEON Study. Oncology 2023;101:22-31. [Crossref] [PubMed]
  84. Marabelle A, Le DT, Ascierto PA, et al. Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol 2020;38:1-10. [Crossref] [PubMed]
  85. Maio M, Ascierto PA, Manzyuk L, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol 2022;33:929-38. [Crossref] [PubMed]
  86. Drilon A, Laetsch TW, Kummar S, et al. Efficacy of Larotrectinib in TRK Fusion-Positive Cancers in Adults and Children. N Engl J Med 2018;378:731-9. [Crossref] [PubMed]
  87. Hong DS, DuBois SG, Kummar S, et al. Larotrectinib in patients with TRK fusion-positive solid tumours: a pooled analysis of three phase 1/2 clinical trials. Lancet Oncol 2020;21:531-40. [Crossref] [PubMed]
  88. Doebele RC, Drilon A, Paz-Ares L, et al. Entrectinib in patients with advanced or metastatic NTRK fusion-positive solid tumours: integrated analysis of three phase 1-2 trials. Lancet Oncol 2020;21:271-82. [Crossref] [PubMed]
  89. Marcus L, Donoghue M, Aungst S, et al. FDA Approval Summary: Entrectinib for the Treatment of NTRK gene Fusion Solid Tumors. Clin Cancer Res 2021;27:928-32. [Crossref] [PubMed]
Cite this article as: Lai ECH, Ung AKY. Update on management of pancreatic cancer: a literature review. Chin Clin Oncol 2024;13(3):41. doi: 10.21037/cco-23-94

Download Citation