Open access peer-reviewed chapter

Pancreatic Tumorigenesis: Precursors, Genetic Risk Factors and Screening

Written By

Abdullah Esmail, Mohamed Badheeb and Maen Abdelrahim

Submitted: 21 February 2023 Reviewed: 15 March 2023 Published: 26 July 2023

DOI: 10.5772/intechopen.110887

From the Edited Volume

Pancreatic Cancer- Updates in Pathogenesis, Diagnosis and Therapies

Edited by Emad Hamdy Gad

Chapter metrics overview

79 Chapter Downloads

View Full Metrics

Abstract

Pancreatic cancer (PC) is a highly fatal malignancy with a unique tumor microenvironment that limits the effectiveness of chemotherapeutics. PC develops from genetic mutations, cellular injury, and environmental exposure, progressing from precursor lesions to malignant neoplasms. This silent disease presents non-specific symptoms, including abdominal pain and painless jaundice. Serological and imaging evaluation aids in the diagnosis, with imaging modality selection dependent on cholestasis presence. The meticulous evaluation of vascular involvement and distant metastasis determines the tumor’s resectability. Neoadjuvant therapy improves patient selection and limits micrometastases, while chemotherapy is the preferred treatment for unresectable cases. Early detection and personalized treatment are essential in improving PC’s clinical outcomes.

Keywords

  • pancreatic cancer
  • tumorigenesis
  • screening
  • neoadjuvant therapy
  • pancreatic molecular profiling
  • pancreatic tumor microenvironment (TME)

1. Introduction

Pancreatic cancer (PC) is used interchangeably to describe pancreatic ductal adenocarcinoma, the most common pancreatic malignancy and one of the most fatal cancers worldwide [1, 2]. To gain a better understanding of PC pathogenesis, it is crucial to comprehend the pancreatic tumor microenvironment (TME). The TME is uniquely characterized by a dense desmoplastic fibrotic stroma in which extracellular matrix proteins (e.g., collagens), along with tumor-derived immune cells (e.g., neutrophils, macrophages), host immune cells (e.g., T-cells), fibroblasts, and activated pancreatic stellate cells (PSCs), form a dense barrier that limits the efficacy of different chemotherapeutics. This renders PC a difficult-to-treat illness [3, 4, 5, 6]. Indeed, the tumorigenesis of PC involves genetic mutations, cellular injury, and environmental exposure that permit the transition into precursor lesions, which further progress into malignant neoplasms [7]. For instance, a constitutively active KRAS allows persistent downstream signaling with substantial cellular proliferation, resulting in ductal metaplasia [8, 9]. However, this process requires the acquisition of further genetic mutations, such as Angiopoietin-like 4, that permit the progression into pancreatic intraepithelial neoplasia (PanINs) [10, 11]. Additionally, mutated TP53, CDKN2A, and SMAD4 accelerate PC growth and progression [12, 13, 14, 15]. Moreover, various environmental factors are believed to contribute to PC tumorigenesis. Smoking has been shown to potentiate desmoplastic reactions by activating PSCs and the associated free radical injury [16]. Other contributing factors include obesity, primarily linked to its associated inflammatory status, which potentiates tumor progression [17, 18]. Furthermore, diabetes mellitus has been shown to over-activate PSCs, potentiating PC development [18]. Non-modifiable risk factors are also involved in PC development. Indeed, a higher incidence of PC was reported in patients of African American descent and patients with a family history of PC [19, 20]. Moreover, specific loci and familial cancer syndromes (e.g., hereditary non-polyposis colon cancer, familial atypical multiple mole melanoma syndromes) have been implicated in PC development [21]. Nonetheless, PC development is a multifactorial process, with various genetic and environmental factors contributing to its pathogenesis.

Advertisement

2. Clinical features of pancreatic cancer

The presentation of PC may vary based on the tumor location and stage. It is generally a silent disease, and if symptoms do occur, they tend to be non-specific, often leading to alternative diagnoses [22, 23]. Although “silent jaundice” is a classical symptom, abdominal pain is more frequently reported in 60–80% of cases [2425]. Tumors located in the head of the pancreas (70% of cases) tend to present with jaundice earlier in the course of the illness, while those in the body or tail present with jaundice later, indicating hepatic metastasis instead of biliary obstruction [26]. Other historical findings may include recent-onset diabetes, nausea or vomiting, anorexia, back pain, and weight loss.

In more advanced cases, pancreatic duct obstruction can result in symptoms of pancreatic failure, reported as post-prandial abdominal pain and steatorrhea. Fat malabsorption with associated vitamin deficiencies may also occur [24, 27, 28, 29]. Jaundice, hepatomegaly, and rarely epigastric mass may be noticed on examination [27]. Additionally, patients may experience recurrent venous stasis, resulting in splenomegaly with portal or splenic vein compression, ascites with inferior vena cava obstruction, and/or superficial thrombophlebitis (Trousseau’s syndrome), palpable gallbladder (Courvoisier’s sign), enlargement of the supraclavicular (Troisier’s sign), or periumbilical (Sister Mary Joseph’s node) lymph nodes may be observed [30, 31, 32]. Unfortunately, these findings are identified later in the course of the illness, indicating more advanced cases with poorer outcomes.

Clinicians should look for specific features of syndromes associated with PC, such as numerous atypical nevi in familial atypical multiple mole melanoma syndromes, mucocutaneous pigmentation in Peutz-Jeghers syndrome, and sebaceous tumors and cutaneous keratoacanthomas in patients with Lynch syndrome [33, 34, 35]. Syndromes associated with PC and their clinical features are summarized in Table 1.

SyndromeAssociated featuresIncreased risk of PCReferences
Hereditary non-polyposis colon cancerIncrease risk for endometrial, ovarian, gastric, colorectal, renal, gliomas, keratoacanthomas, and other malignancies8.6-fold[36]
Hereditary breast and ovarian cancer syndromeIncreased risk for breast (in males and females), ovarian, prostate, and skin (e.g., melanoma) malignancies3–7%[37]
Familial atypical multiple mole melanoma syndromeNumerous atypical nevi resembling early melanoma, and a family history of melanoma
Increase risk for lung, skin, larynx, and breast malignancies
13–22 folds[38]
Familial adenomatous polyposisIncreased risk for desmoid tumors, gastric/duodenal, hepatoblastoma, and thyroid malignancies4 folds[39]
Peutz-Jeghers syndromeIncreased risk for colorectal, gastric, breast, ovarian, cervical, and testicular malignancies15 folds[40]
Li-Fraumeni syndromeIncreased risk for bone and soft tissue sarcomas and breast, brain, and adrenocortical malignancies7 folds[41]

Table 1.

Brief summary of familial cancer syndromes associated with pancreatic cancer (PC).

Advertisement

3. Diagnosis of pancreatic cancer

The clinical presentation of PC is neither specific nor sensitive for establishing a diagnosis; therefore, suspected cases typically require serological and imaging testing. Liver function tests, including serum aminotransferase, bilirubin, and alkaline phosphatase, should be performed in all patients. The selection of subsequent testing primarily depends on the presence of jaundice or obstructive laboratory features (e.g., elevated direct bilirubin). In such cases, transabdominal ultrasound (TAUS) provides excellent sensitivity in detecting masses in the head of the pancreas and visualizing biliary tract patency or dilatation (Figure 1) [42, 43].

Figure 1.

Simplified algorithm for pancreatic cancer diagnosis. * Pancreatic protocol to assess the resectability of the tumor.

For anicteric patients who present with epigastric pain or other worrisome symptoms, such as weight loss, anorexia, or post-prandial flatulence, an abdominal computed tomography (CT) scan should be performed, which provides higher sensitivity in detecting lesions in the body and tail of the pancreas. In addition, a CT scan can be used initially, rather than TAUS, in cases of acute pancreatitis, as bowel gases may obscure the visualization of the biliary tract and the pancreas [4445]. If initial imaging is positive, further evaluation using a multi-phase contrast-enhanced, helical abdominal CT scan (i.e., pancreatic protocol) is the preferred option, accurate characterization of the pancreatic mass and resectability evaluation [46, 47, 48].

In cases where initial imaging (i.e., TAUS or CT scan) is negative, no further testing is required unless there is a strong suspicion that pancreatic cancer is the culprit of patient symptoms. In such cases, patients may undergo endoscopic retrograde cholangiopancreatography (ERCP), which allows for direct visualization of the biliary tract and pancreatic duct, tissue sampling for histopathological examination, and therapeutic decompression through stent insertion in selected cases. Alternatively, magnetic resonance cholangiopancreatography (MRCP) can be used in patients who are not qualified to undergo ERCP due to bowel obstruction or cases when ERCP fails to provide an informative visualization of the biliary tract [49, 50, 51]. When these modalities are negative, no further testing is required unless pancreatic cancer is strongly suspected. In such cases, endoscopic ultrasound (EUS) may be sought to to assess further the presence of any pathologies, which should be sampled through fine needle aspiration (FNA). More recently, contrast-enhances EUS appeared to be a more feasible approach for tissue sampling in such cases [43, 52, 53, 54].

Tumor, node, metastasis (TNM) system by the American Joint Committee on Cancer manual is a widely-accepted staging system that aids in the assignment of patients based on the resectability of PC. Additionally, it provides prognostic information based on the stage; for instance, patients in stage Ia had an overall 5-years survival of 39% compared to 11% in stage III [48, 49]. Nevertheless, a four-grouped classification system is used by many clinicians, which classifies PC based on resectability into; resectable, borderline resectability, locally advanced, and metastatic PC [50]. Regardless of the system used, the ultimate goal is to determine the suitable patient for curative resection.

Advertisement

4. Screening of pancreatic cancer

Early diagnosis of PC has been shown to improve overall survival. Nevertheless, the low incidence of PC discourages the implementation of nationwide screening modalities due to the high risk of false positive cases that may undergo unnecessary invasive testing. Furthermore, there are currently no guidelines regarding the optimal screening for PC [55, 56, 57, 58]. Therefore, patients should be selected cautiously and counseled regarding their risks, the benefits and harms of the test, and the probable outcomes of their testing.

Given the rarity of PC, a targeted screening approach may be the most suitable option. Initially, the identification of high-risk patients based on National Comprehensive Cancer Network (NCCN) recommendations [59] is primarily made on the basis of specific associated genetic mutations or syndromes to select the most appropriate age for screening initiation, as summarized in Table 2.

Patient groupAge of initiation
High-risk genetic mutation, any of the:
  • ATM, BRCA1, BRCA2, MLH1, MSH2, MSH6, PALB2 or TP53

With a first-degree relative diagnosed with PC
Whichever earlier:
  • At 50 years old or

  • 10 years prior to the first PDAC in the family

Peutz-Jeghers syndromeAt 30–35 years old
Hereditary pancreatitis
  • 40-years old or

  • 20 years following the onset of pancreatitis

CDKN2A mutation
  • 40-years old or

  • Within 10 years of the first PDAC in the family

Table 2.

Screening age recommendations for high-risk patient groups.

Various serological markers and liquid biopsies have been extensively studied; however, only Carbohydrate Antigen 19-9 (CA19-9) has gained approval from the Food and Drug Administration (FDA). Carcinoembryonic antigen (CEA), which is classically elevated in colorectal cancer, appears to have some diagnostic utility for detecting cancer but has lower specificity compared to CA19-9. The use of multiple biomarkers together provides higher cumulative sensitivity and specificity. For instance, CA19-9, CEA, CA125, and CA242 together had 90.4% and 93.8% sensitivity and specificity, respectively, substantially higher than any single marker [60, 61, 62]. More recently, liquid biopsies have gained tremendous interest as an alternative non-invasive method to detect PC. Mainly, circulating tumor DNA (ctDNA) and circulating tumor cells (CTCs) are among the most promising. However, they are not readily available in many healthcare settings and have variable diagnostic accuracy [63, 64, 65]. Table 3 summarizes different screening methods, their usefulness, and limitations.

TestSensitivitySpecificityAdvantageLimitations
Serology
CA19–980% [60]75% [60]Readily-available, FDA-approvedLow specificity, elevated in benign and non-PC cases, can be negative in up to 10% of Caucasians [66, 67]
CEA45% [68]89% [68]Low sensitivity, elevated in benign and non-PC cases [68]
CA12559% [69]78% [69]Not influenced by bilirubin levels, hence, its positive is the same in jaundiced and non-jaundiced individuals with PC [70]Elevated in benign and non-PC cases [71]
CA24266.2% [61]80.14% [72]May provide prognostic indications [73]Ineffective early screening as a high level indicates a huge tumor burden [73]
IgG472% [74]89% [74]Limited usefulness to differentiate hereditary pancreatitis from PC [75]
Glycoproteomics~90% [76]90% [77]Very high sensitivity and specificity can detect PC in its early stages
Lipodomic profiling>90% [78]>90% [78]Very high sensitivity and specificity may provide a prognostic indicator [78]
Liquid biopsy
CTCs and ctDNA25–100% [63, 64]95.4% [79]May serve as recurrence, invasion, and metastasis predictors [63]Variable sensitivity, and no standardized methodology to detect their recurrence [80]
cfDNA76% [81]59% [81]Can detect genetic mutations (e.g., KRAS), and may serve as a recurrence, predictor [82, 83]Low diagnostic accuracy, cannot detect the cancer cells origin [84]
Circulating miRNAs92.5% [85]90% [85]The detection rate is tumor-burden dependent [86]
Circulating exosomes75.4–100% [87]92.6–100% [87]High specificity can detect cancer DNA in its early stages [88]

Table 3.

Different pancreatic cancer screening methods, their usefulness, and limitations.

Little is known regarding the best approach to screening for PC. Nevertheless, a comprehensive evaluation with cautious patient selection and integrative serological and imaging testing may be the most appropriate approach.

Advertisement

5. Management of pancreatic cancer

The management of PC is multidisciplinary. The tumor resectability should be evaluated initially with a multi-phase contrast-enhanced, helical chest and abdominopelvic CT scans. The tumor is considered resectable when confined to the pancreas with no metastasis or vascular encasement, such as the superior mesenteric artery/vein, celiac trunk, or common hepatic artery. Conversely, the presence of hepatic, peritoneal, or extra-abdominal metastasis renders the tumor unresectable [89, 90]. Nevertheless, in selective cases, the NCCN considers PC to be borderline unresectable. Examples of such cases include head of pancreas cancer that directly contacts the inferior vena cava, hepatic artery with no extension to the bifurcation, or tail/body PC with a celiac axis of 180 degrees or less [59]. For resectable PC that involves the head of the pancreas, the Whipple procedure is performed, including pancreatic head, duodenum, proximal jejunum, common bile duct, gall bladder, and a portion of the stomach resection (i.e., pancreaticoduodenectomy) [91, 92]. In contrast, distal pancreatectomy is typically performed in PC of the body/tail, which occasionally may include splenectomy [93]. Biliary drainage has been classically performed pre-operatively in patients with obstructive jaundice. However, the clinical benefits of this approach are controversial; therefore, it should be reserved for patients with severe hyperbilirubinemia, protracted itching, or cholangitis [94, 95, 96].

Neoadjuvant therapy has been found to outperform initial surgical resection for PC in providing a more precise patient selection and possibly limiting micrometastases linked to PC recurrence even after surgical resection. In addition, lower margin-positive resections were observed with the use of neoadjuvant therapy [97, 98, 99]. However, there are currently no established guidelines regarding optimal chemotherapy. The FOLFIRINOX protocol and a combination of gemcitabine plus nab-paclitaxel have been used, but there is no sufficient evidence to support the superiority of each approach [100]. Thus, we recommend a multidisciplinary team evaluation that takes into account the patient’s preferences, institutional experience, and cost-effectiveness when selecting the chemotherapeutic agents.

Metastatic and locally advanced PC are generally considered unresectable, and chemotherapy is the preferred approach for such cases. Although there is no consensus available for the preferred regimen, FOLFIRINOX or Gemcitabine-based protocols may be used. Different clinical trials have demonstrated the efficacy of each approach. However, FOLFIRINOX has shown a longer overall survival compared to Gemcitabine [101, 102, 103, 104, 105]. Patients who fail one protocol may be considered for the other after assessing their performance status. Additionally, patients should be re-evaluated for possible resection following chemotherapy, as tumor downstaging may permit resection.

Advertisement

List of abbreviation

CA19-9carbohydrate antigen 19-9
CA125cancer antigen 125
CA242carbohydrate antigen 242
CEAcarcinoembryonic antigen
cfDNAcell-free DNA
CTcomputed tomography
ctDNAcirculating tumor DNA
CTCscirculating tumor cells
ERCPendoscopic retrograde cholangiopancreatography
EUSendoscopic ultrasound
FDAFood and Drug Administration
IgG4immunoglobulin G4
MRCPmagnetic resonance cholangiopancreatography
NCCNNational Comprehensive Cancer Network
PanINspancreatic intraepithelial neoplasia
PCpancreatic cancer
PSCspancreatic stellate cells
TAUStransabdominal ultrasound
TMEtumor microenvironment
TNMtumor, node, metastasis

References

  1. 1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA: A Cancer Journal for Clinicians. 2022;72(1):7-33
  2. 2. Rawla P, Sunkara T, Gaduputi V. Epidemiology of pancreatic cancer: Global trends, etiology and risk factors. World Journal of Oncology. 2019;10(1):10-27
  3. 3. DeClerck YA. Desmoplasia: A response or a niche? Cancer Discovery. 2012;2(9):772-774
  4. 4. Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003;4(6):437-450
  5. 5. Cannon A, Thompson C, Hall BR, Jain M, Kumar S, Batra SK. Desmoplasia in pancreatic ductal adenocarcinoma: Insight into pathological function and therapeutic potential. Genes & Cancer. 2018;9(3-4):78-86
  6. 6. Suklabaidya S, Dash P, Das B, Suresh V, Sasmal PK, Senapati S. Experimental models of pancreatic cancer desmoplasia. Laboratory Investigation; A Journal of Technical Methods and Pathology. 2018;98(1):27-40
  7. 7. Petersen GM. Familial pancreatic adenocarcinoma. Hematology/Oncology Clinics of North America. 2015;29(4):641-653
  8. 8. Colicelli J, Human RAS. Superfamily Proteins and Related GTPases. 2004;2004(250) re13-re
  9. 9. Hobbs GA, Der CJ, Rossman KL. RAS isoforms and mutations in cancer at a glance. Journal of Cell Science. 2016;129(7):1287-1292
  10. 10. Basturk O, Hong SM, Wood LD, Adsay NV, Albores-Saavedra J, Biankin AV, et al. A revised classification system and recommendations from the Baltimore consensus meeting for neoplastic precursor lesions in the pancreas. The American Journal of Surgical Pathology. 2015;39(12):1730-1741
  11. 11. Esposito I, Konukiewitz B, Schlitter AM, Klöppel G. Pathology of pancreatic ductal adenocarcinoma: Facts, challenges and future developments. World Journal of Gastroenterology. 2014;20(38):13833-13841
  12. 12. Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, et al. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes & Development. 2003;17(24):3112-3126
  13. 13. Jonckheere N, Vasseur R, Van Seuningen I. The cornerstone K-RAS mutation in pancreatic adenocarcinoma: From cell signaling network, target genes, biological processes to therapeutic targeting. Critical Reviews in Oncology/Hematology. 2017;111:7-19
  14. 14. Tang B, Li Y, Qi G, Yuan S, Wang Z, Yu S, et al. Clinicopathological significance of CDKN2A promoter hypermethylation frequency with pancreatic cancer. Scientific Reports. 2015;5:13563
  15. 15. Sherr CJ. The INK4a/ARF network in tumour suppression. Nature Reviews Molecular Cell Biology. 2001;2(10):731-737
  16. 16. Schaal C, Chellappan SP. Nicotine-mediated cell proliferation and tumor progression in smoking-related cancers. Molecular Cancer Research. 2014;12(1):14-23
  17. 17. Rawla P, Thandra KC, Sunkara T. Pancreatic cancer and obesity: Epidemiology, mechanism, and preventive strategies. Clinical Journal of Gastroenterology. 2019;12(4):285-291
  18. 18. Eibl G, Cruz-Monserrate Z, Korc M, Petrov MS, Goodarzi MO, Fisher WE, et al. Diabetes mellitus and obesity as risk factors for pancreatic cancer. Journal of the Academy of Nutrition and Dietetics. 2018;118(4):555-567
  19. 19. Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology. 2013;144(6):1252-1261
  20. 20. Arnold LD, Patel AV, Yan Y, Jacobs EJ, Thun MJ, Calle EE, et al. Are racial disparities in pancreatic cancer explained by smoking and overweight/obesity? Cancer Epidemiology, Biomarkers & Prevention. 2009;18(9):2397-2405
  21. 21. Badheeb M, Abdelrahim A, Esmail A, Umoru G, Abboud K, Al-Najjar E, et al. Pancreatic tumorigenesis: Precursors, genetic risk factors and screening. Current Oncology (Toronto, Ont). 2022;29(11):8693-8719
  22. 22. Macdonald S, Macleod U, Campbell NC, Weller D, Mitchell E. Systematic review of factors influencing patient and practitioner delay in diagnosis of upper gastrointestinal cancer. British Journal of Cancer. 2006;94(9):1272-1280
  23. 23. Walter FM, Mills K, Mendonça SC, Abel GA, Basu B, Carroll N, et al. Symptoms and patient factors associated with diagnostic intervals for pancreatic cancer (SYMPTOM pancreatic study): A prospective cohort study. The Lancet Gastroenterology & Hepatology. 2016;1(4):298-306
  24. 24. Schmidt-Hansen M, Berendse S, Hamilton W. Symptoms of pancreatic cancer in primary care: A systematic review. Pancreas. 2016;45(6):814-818
  25. 25. Furukawa H, Okada S, Saisho H, Ariyama J, Karasawa E, Nakaizumi A, et al. Clinicopathologic features of small pancreatic adenocarcinoma. A collective study. Cancer. 1996;78(5):986-990
  26. 26. Kalser MH, Barkin J, MacIntyre JM. Pancreatic cancer. Assessment of prognosis by clinical presentation. Cancer. 1985;56(2):397-402
  27. 27. Porta M, Fabregat X, Malats N, Guarner L, Carrato A, de Miguel A, et al. Exocrine pancreatic cancer: Symptoms at presentation and their relation to tumour site and stage. Clinical & Translational Oncology: Official Publication of the Federation of Spanish Oncology Societies and of the National Cancer Institute of Mexico. 2005;7(5):189-197
  28. 28. Holly EA, Chaliha I, Bracci PM, Gautam M. Signs and symptoms of pancreatic cancer: A population-based case-control study in the San Francisco Bay area. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2004;2(6):510-517
  29. 29. Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: A population-based study. Gastroenterology. 2005;129(2):504-511
  30. 30. DiMagno EP. Pancreatic cancer: Clinical presentation, pitfalls and early clues. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 1999;10(Suppl 4):140-142
  31. 31. Takhar AS, Palaniappan P, Dhingsa R, Lobo DN. Recent developments in diagnosis of pancreatic cancer. BMJ (Clinical Research ed). 2004;329(7467):668-673
  32. 32. Khorana AA, Fine RL. Pancreatic cancer and thromboembolic disease. The Lancet Oncology. 2004;5(11):655-663
  33. 33. Utsunomiya J, Gocho H, Miyanaga T, Hamaguchi E, Kashimure A. Peutz-Jeghers syndrome: Its natural course and management. The Johns Hopkins Medical Journal. 1975;136(2):71-82
  34. 34. Eckerle Mize D, Bishop M, Resse E, Sluzevich J. Familial atypical multiple mole melanoma syndrome. In: Riegert-Johnson DL, Boardman LA, Hefferon T, Roberts M, editors. Cancer Syndromes. Bethesda (MD): National Center for Biotechnology Information (US) Copyright © 2009, Douglas L Riegert-Johnson; 2009
  35. 35. Coquillard C, Boustany A, DeCoster RC, Vasconez HC. Muir-Torre syndrome presenting as a sebaceous carcinoma of the nasal ala. The American Surgeon. 2019;85(3):e115-e1e7
  36. 36. Hoskins JW, Ibrahim A, Emmanuel MA, Manmiller SM, Wu Y, O'Neill M, et al. Functional characterization of a chr13q22.1 pancreatic cancer risk locus reveals long-range interaction and allele-specific effects on DIS3 expression. Human Molecular Genetics. 2016;25(21):4726-4738
  37. 37. Petrucelli N, Daly MB, Pal T. BRCA1- and BRCA2-associated hereditary breast and ovarian cancer. In: Adam MP, Everman DB, Mirzaa GM, Pagon RA, Wallace SE, LJH B, et al., editors. GeneReviews(®). Seattle (WA): University of Washington, Seattle Copyright © 1993-2023, University of Washington, Seattle. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved; 1993
  38. 38. Riker AI, Hagmaier R. The Familial Atypical Multiple Mole Melanoma (FAMMM)-Pancreatic Carcinoma (PC) Syndrome. Inherited Cancer Syndromes: Springer New York; 2010. pp. 135-144
  39. 39. Kanth P, Grimmett J, Champine M, Burt R, Samadder NJ. Hereditary colorectal polyposis and cancer syndromes: A primer on diagnosis and management. The American Journal of Gastroenterology. 2017;112(10):1509-1525
  40. 40. Syngal S, Brand RE, Church JM, Giardiello FM, Hampel HL, Burt RW. ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes. 2015;110(2):223-262
  41. 41. Malkin D. Li-fraumeni syndrome. Genes & Cancer. 2011;2(4):475-484
  42. 42. Conrad C, Fernández-Del CC. Preoperative evaluation and management of the pancreatic head mass. Journal of Surgical Oncology. 2013;107(1):23-32
  43. 43. Kanno A, Masamune A, Hanada K, Maguchi H, Shimizu Y, Ueki T, et al. Multicenter study of early pancreatic cancer in Japan. Pancreatology: Official Journal of the International Association of Pancreatology (IAP) [et al.]. 2018;18(1):61-67
  44. 44. Pasanen PA, Eskelinen M, Partanen K, Pikkarainen P, Penttilä I, Alhava E. A prospective study of the value of imaging, serum markers and their combination in the diagnosis of pancreatic carcinoma in symptomatic patients. Anticancer Research. 1992;12(6b):2309-2314
  45. 45. Ashida R, Tanaka S, Yamanaka H, Okagaki S, Nakao K, Fukuda J, et al. The Role of Transabdominal Ultrasound in the Diagnosis of Early Stage Pancreatic Cancer: Review and Single-Center Experience. Diagnostics (Basel, Switzerland). 2018;9(1).
  46. 46. Brennan DD, Zamboni GA, Raptopoulos VD, Kruskal JB. Comprehensive preoperative assessment of pancreatic adenocarcinoma with 64-section volumetric CT. Radiographics: A Review Publication of the Radiological Society of North America, Inc. 2007;27(6):1653-1666
  47. 47. Fletcher JG, Wiersema MJ, Farrell MA, Fidler JL, Burgart LJ, Koyama T, et al. Pancreatic malignancy: Value of arterial, pancreatic, and hepatic phase imaging with multi-detector row CT. Radiology. 2003;229(1):81-90
  48. 48. Sahani DV, Bonaffini PA, Catalano OA, Guimaraes AR, Blake MA. State-of-the-art PET/CT of the pancreas: Current role and emerging indications. Radiographics: A Review Publication of the Radiological Society of North America, Inc. 2012;32(4):1133-1158 (discussion 58-60)
  49. 49. Niederau C, Grendell JH. Diagnosis of pancreatic carcinoma. Imaging techniques and tumor markers. Pancreas. 1992;7(1):66-86
  50. 50. Adamek HE, Albert J, Breer H, Weitz M, Schilling D, Riemann JF. Pancreatic cancer detection with magnetic resonance cholangiopancreatography and endoscopic retrograde cholangiopancreatography: A prospective controlled study. Lancet (London, England). 2000;356(9225):190-193
  51. 51. Lopez Hänninen E, Amthauer H, Hosten N, Ricke J, Böhmig M, Langrehr J, et al. Prospective evaluation of pancreatic tumors: Accuracy of MR imaging with MR cholangiopancreatography and MR angiography. Radiology. 2002;224(1):34-41
  52. 52. Dewitt J, Devereaux BM, Lehman GA, Sherman S, Imperiale TF. Comparison of endoscopic ultrasound and computed tomography for the preoperative evaluation of pancreatic cancer: A systematic review. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2006;4(6):717-725 quiz 664
  53. 53. Li Y, Jin H, Liao D, Qian B, Zhang Y, Xu M, et al. Contrast-enhanced harmonic endoscopic ultrasonography for the differential diagnosis of pancreatic masses: A systematic review and meta-analysis. Molecular and Clinical Oncology. 2019;11(4):425-433
  54. 54. Rahman MIO, Chan BPH, Far PM, Mbuagbaw L, Thabane L, Yaghoobi M. Endoscopic ultrasound versus computed tomography in determining the resectability of pancreatic cancer: A diagnostic test accuracy meta-analysis. Saudi Journal of Gastroenterology: Official Journal of the Saudi Gastroenterology Association. 2020;26(3):113-119
  55. 55. Canto MI, Goggins M, Hruban RH, Petersen GM, Giardiello FM, Yeo C, et al. Screening for early pancreatic neoplasia in high-risk individuals: A prospective controlled study. Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American Gastroenterological Association. 2006;4(6):766-781 quiz 665
  56. 56. Canto MI, Harinck F, Hruban RH, Offerhaus GJ, Poley J-W, Kamel I, et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. 2013;62(3):339-347
  57. 57. Hayashi A, Fan J, Chen R, Ho YJ, Makohon-Moore AP, Lecomte N, et al. A unifying paradigm for transcriptional heterogeneity and squamous features in pancreatic ductal adenocarcinoma. Nature Cancer. 2020;1(1):59-74
  58. 58. Pandharipande PV, Heberle C, Dowling EC, Kong CY, Tramontano A, Perzan KE, et al. Targeted screening of individuals at high risk for pancreatic cancer: Results of a simulation model. Radiology. 2015;275(1):177-187
  59. 59. NCC N. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®): Pancreatic Adenocarcinoma. 2019 version 1.2020
  60. 60. Xing H, Wang J, Wang Y, Tong M, Hu H, Huang C, et al. Diagnostic value of CA 19-9 and carcinoembryonic antigen for pancreatic cancer: A meta-analysis. Gastroenterology Research and Practice. 2018;2018:8704751
  61. 61. Pasanen PA, Eskelinen M, Partanen K, Pikkarainen P, Penttilä I, Alhava E. Clinical evaluation of a new serum tumour marker CA 242 in pancreatic carcinoma. British Journal of Cancer. 1992;65(5):731-734
  62. 62. Gu YL, Lan C, Pei H, Yang SN, Liu YF, Xiao LL. Applicative value of serum CA19-9, CEA, CA125 and CA242 in diagnosis and prognosis for patients with pancreatic cancer treated by concurrent chemoradiotherapy. Asian Pacific Journal of Cancer Prevention. 2015;16(15):6569-6573
  63. 63. Abdelrahim M, Esmail A, Xu J, Katz TA, Sharma S, Kalashnikova E, et al. Early relapse detection and monitoring disease status in patients with early-stage pancreatic adenocarcinoma using circulating tumor DNA. The Journal of Surgical Research. 2021;4:602-615
  64. 64. Xie ZB, Yao L, Jin C, Fu DL. Circulating tumor cells in pancreatic cancer patients: Efficacy in diagnosis and value in prognosis. Discovery Medicine. 2016;22(120):121-128
  65. 65. Herreros-Villanueva M, Bujanda L. Non-invasive biomarkers in pancreatic cancer diagnosis: What we need versus what we have. Annals of Translational Medicine. 2016;4(7):134
  66. 66. Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19-9 as a screening test for pancreatic cancer in an asymptomatic population. Journal of Gastroenterology and Hepatology. 2004;19(2):182-186
  67. 67. Ballehaninna UK, Chamberlain RS. The clinical utility of serum CA 19-9 in the diagnosis, prognosis and management of pancreatic adenocarcinoma: An evidence based appraisal. Journal of Gastrointestinal Oncology. 2012;3(2):105-119
  68. 68. Hao C, Zhang G, Zhang L. Serum CEA levels in 49 different types of cancer and noncancer diseases. Progress in Molecular Biology and Translational Science. 2019;162:213-227
  69. 69. Meng Q , Shi S, Liang C, Xiang J, Liang D, Zhang B, et al. Diagnostic accuracy of a CA125-based biomarker panel in patients with pancreatic cancer: A systematic review and meta-analysis. Journal of Cancer. 2017;8(17):3615-3622
  70. 70. Haglund C. Tumour marker antigen CA125 in pancreatic cancer: A comparison with CA19-9 and CEA. British Journal of Cancer. 1986;54(6):897-901
  71. 71. Yin BW, Lloyd KO. Molecular cloning of the CA125 ovarian cancer antigen: Identification as a new mucin, MUC16. The Journal of Biological Chemistry. 2001;276(29):27371-27375
  72. 72. Lei XF, Jia SZ, Ye J, Qiao YL, Zhao GM, Li XH, et al. Application values of detection of serum CA199, CA242 and CA50 in the diagnosis of pancreatic cancer. Journal of Biological Regulators and Homeostatic Agents. 2017;31(2):383-388
  73. 73. Dou H, Sun G, Zhang L. Chapter twelve—CA242 as a biomarker for pancreatic cancer and other diseases. In: Zhang L, editor. Progress in Molecular Biology and Translational Science. 162: Academic Press; 2019. pp. 229-239
  74. 74. Dai C, Cao Q , Jiang M, Sun MJ. Serum immunoglobulin G4 in discriminating autoimmune pancreatitis from pancreatic cancer: A diagnostic meta-analysis. Pancreas. 2018;47(3):280-284
  75. 75. Ghazale A, Chari ST, Smyrk TC, Levy MJ, Topazian MD, Takahashi N, et al. Value of serum IgG4 in the diagnosis of autoimmune pancreatitis and in distinguishing it from pancreatic cancer. Official Journal of the American College of Gastroenterology. 2007;102(8)
  76. 76. Aronsson L, Andersson R, Bauden M, Andersson B, Bygott T, Ansari D. High-density and targeted glycoproteomic profiling of serum proteins in pancreatic cancer and intraductal papillary mucinous neoplasm. Scandinavian Journal of Gastroenterology. 2018;53(12):1597-1603
  77. 77. Liu Y, Wang C, Wang R, Wu Y, Zhang L, Liu BF, et al. Isomer-specific profiling of N-glycans derived from human serum for potential biomarker discovery in pancreatic cancer. Journal of Proteomics. 2018;181:160-169
  78. 78. Wolrab D, Jirásko R, Cífková E, Höring M, Mei D, Chocholoušková M, et al. Lipidomic profiling of human serum enables detection of pancreatic cancer. Nature Communications. 2022;13(1):124
  79. 79. Tien YW, Kuo HC, Ho BI, Chang MC, Chang YT, Cheng MF, et al. A high circulating tumor cell count in portal vein predicts liver metastasis from periampullary or pancreatic cancer: A high portal venous CTC count predicts liver metastases. Medicine. 2016;95(16):e3407
  80. 80. Buscail E, Alix-Panabières C, Quincy P, Cauvin T, Chauvet A, Degrandi O, et al. High clinical value of liquid biopsy to detect circulating tumor cells and tumor exosomes in pancreatic ductal adenocarcinoma patients eligible for up-front surgery. Cancers. 2019;11(11):7-11
  81. 81. Brancaccio M, Natale F, Falco G, Angrisano T. Cell-free DNA methylation: The new frontiers of pancreatic cancer biomarkers’ discovery. 2020;11(1):14
  82. 82. Jaworski JJ, Morgan RD, Sivakumar S. Circulating cell-free tumour DNA for early detection of pancreatic cancer. Cancers. 2020;12(12):3-9
  83. 83. Sausen M, Phallen J, Adleff V, Jones S, Leary RJ, Barrett MT, et al. Clinical implications of genomic alterations in the tumour and circulation of pancreatic cancer patients. Nature Communications. 2015;6:7686
  84. 84. Bettegowda C, Sausen M, Leary RJ, Kinde I, Wang Y, Agrawal N, et al. Detection of circulating tumor DNA in early- and late-stage human malignancies. Science Translational Medicine. 2014;6(224):224ra24
  85. 85. De Guire V, Robitaille R, Tétreault N, Guérin R, Ménard C, Bambace N, et al. Circulating miRNAs as sensitive and specific biomarkers for the diagnosis and monitoring of human diseases: Promises and challenges. Clinical Biochemistry. 2013;46(10):846-860
  86. 86. Yu J, Li A, Hong SM, Hruban RH, Goggins M. MicroRNA alterations of pancreatic intraepithelial neoplasias. Clinical Cancer Research: An Official Journal of the American Association for Cancer Research. 2012;18(4):981-992
  87. 87. Allenson K, Castillo J, San Lucas FA, Scelo G, Kim DU, Bernard V, et al. High prevalence of mutant KRAS in circulating exosome-derived DNA from early-stage pancreatic cancer patients. Annals of Oncology. 2017;28(4):741-747
  88. 88. Melo SA, Luecke LB, Kahlert C, Fernandez AF, Gammon ST, Kaye J, et al. Glypican-1 identifies cancer exosomes and detects early pancreatic cancer. Nature. 2015;523(7559):177-182
  89. 89. Versteijne E, van Dam JL, Suker M, Janssen QP, Groothuis K, Akkermans-Vogelaar JM, et al. Neoadjuvant chemoradiotherapy versus upfront surgery for resectable and borderline resectable pancreatic cancer: Long-term results of the Dutch randomized PREOPANC trial. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology. 2022;40(11):1220-1230
  90. 90. Ryan DP, Hong TS, Bardeesy N. Pancreatic adenocarcinoma. The New England Journal of Medicine. 2014;371(11):1039-1049
  91. 91. Kendrick ML, Cusati D. Total laparoscopic pancreaticoduodenectomy: Feasibility and outcome in an early experience. Archives of Surgery (Chicago, Ill: 1960). 2010;145(1):19-23
  92. 92. Palanivelu C, Jani K, Senthilnathan P, Parthasarathi R, Rajapandian S, Madhankumar MV. Laparoscopic pancreaticoduodenectomy: Technique and outcomes. Journal of the American College of Surgeons. 2007;205(2):222-230
  93. 93. Kleeff J, Diener MK, Z'Graggen K, Hinz U, Wagner M, Bachmann J, et al. Distal pancreatectomy: Risk factors for surgical failure in 302 consecutive cases. Annals of Surgery. 2007;245(4):573-582
  94. 94. Bineshfar N, Malekpour Alamdari N, Rostami T, Mirahmadi A, Zeinalpour A. The effect of preoperative biliary drainage on postoperative complications of pancreaticoduodenectomy: A triple center retrospective study. BMC Surgery. 2022;22(1):399
  95. 95. Wang Q , Gurusamy KS, Lin H, Xie X, Wang C. Preoperative biliary drainage for obstructive jaundice. The Cochrane Database of Systematic Reviews. 2008;(3):CD005444
  96. 96. van der Gaag NA, Rauws EA, van Eijck CH, Bruno MJ, van der Harst E, Kubben FJ, et al. Preoperative biliary drainage for cancer of the head of the pancreas. The New England Journal of Medicine. 2010;362(2):129-137
  97. 97. Greco SH, August DA, Shah MM, Chen C, Moore DF, Masanam M, et al. Neoadjuvant therapy is associated with lower margin positivity rates after pancreaticoduodenectomy in T1 and T2 pancreatic head cancers: An analysis of the National Cancer Database. Surgery Open Science. 2021;3:22-28
  98. 98. Versteijne E, Vogel JA, Besselink MG, Busch ORC, Wilmink JW, Daams JG, et al. Meta-analysis comparing upfront surgery with neoadjuvant treatment in patients with resectable or borderline resectable pancreatic cancer. The British Journal of Surgery. 2018;105(8):946-958
  99. 99. Heinrich S, Schäfer M, Weber A, Hany TF, Bhure U, Pestalozzi BC, et al. Neoadjuvant chemotherapy generates a significant tumor response in resectable pancreatic cancer without increasing morbidity: Results of a prospective phase II trial. Annals of Surgery. 2008;248(6):1014-1022
  100. 100. Oba A, Ho F, Bao QR, Al-Musawi MH, Schulick RD, Del Chiaro M. Neoadjuvant treatment in pancreatic cancer. Frontiers in Oncology. 2020;10:245
  101. 101. Colucci G, Giuliani F, Gebbia V, Biglietto M, Rabitti P, Uomo G, et al. Gemcitabine alone or with cisplatin for the treatment of patients with locally advanced and/or metastatic pancreatic carcinoma: A prospective, randomized phase III study of the Gruppo Oncologia dell'Italia Meridionale. Cancer. 2002;94(4):902-910
  102. 102. Scheithauer W, Schüll B, Ulrich-Pur H, Schmid K, Raderer M, Haider K, et al. Biweekly high-dose gemcitabine alone or in combination with capecitabine in patients with metastatic pancreatic adenocarcinoma: A randomized phase II trial. Annals of Oncology: Official Journal of the European Society for Medical Oncology. 2003;14(1):97-104
  103. 103. Tempero M, Plunkett W, Ruiz Van Haperen V, Hainsworth J, Hochster H, Lenzi R, et al. Randomized phase II comparison of dose-intense gemcitabine: Thirty-minute infusion and fixed dose rate infusion in patients with pancreatic adenocarcinoma. Journal of Clinical Oncology: Official Journal of the American society of Clinical Oncology. 2003;21(18):3402-3408
  104. 104. Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. The New England Journal of Medicine. 2013;369(18):1691-1703
  105. 105. Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. The New England Journal of Medicine. 2011;364(19):1817-1825

Written By

Abdullah Esmail, Mohamed Badheeb and Maen Abdelrahim

Submitted: 21 February 2023 Reviewed: 15 March 2023 Published: 26 July 2023