Next Article in Journal
Zebrafish Patient-Derived Xenograft Model to Predict Treatment Outcomes of Colorectal Cancer Patients
Next Article in Special Issue
Tumor Budding Is an Independent Prognostic Factor in Pancreatic Adenocarcinoma and It Positively Correlates with PD-L1 Expression on Tumor Cells
Previous Article in Journal
Chemical Barrier Proteins in Human Body Fluids
Previous Article in Special Issue
Isolated Gastric Metastases of Pancreatic Ductal Adenocarcinoma following Radical Resection—Impact of Endosonography-Guided Fine Needle Aspiration Tract Seeding
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Intraductal Papillary Mucinous Neoplasms in Hereditary Cancer Syndromes

1
Department of Internal Medicine, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
2
College of Medicine, Ohio State University, Columbus, OH 43210, USA
3
Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
4
Division of Gastroenterology, Hepatology and Nutrition, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(7), 1475; https://doi.org/10.3390/biomedicines10071475
Submission received: 22 April 2022 / Revised: 17 June 2022 / Accepted: 20 June 2022 / Published: 22 June 2022
(This article belongs to the Special Issue Advances on Pancreatic Cancer)

Abstract

:
Hereditary pancreatic cancer, which includes patients with familial pancreatic cancer (FPC) and hereditary pancreatic cancer syndromes, accounts for about 10% of all pancreatic cancer diagnoses. The early detection of pre-cancerous pancreatic cysts has increasingly become a focus of interest in recent years as a potential avenue to lower pancreatic cancer incidence and mortality. Intraductal papillary mucinous cystic neoplasms (IPMNs) are recognized precursor lesions of pancreatic cancer. IPMNs have high prevalence in patients with hereditary pancreatic cancer and their relatives. While various somatic mutations have been identified in IPMNs, certain germline mutations associated with hereditary cancer syndromes have also been identified in IPMNs, suggesting a role in their formation. While the significance for the higher prevalence of IPMNs or similar germline mutations in these high-risk patients remain unclear, IPMNs do represent pre-malignant lesions that need close surveillance. This review summarizes the available literature on the incidence and prevalence of IPMNs in inherited genetic predisposition syndromes and FPC and speculates if IPMN and pancreatic cancer surveillance in these high-risk individuals needs to change.

1. Introduction

Pancreatic cancer is one of the leading causes of cancer-related deaths worldwide with a 5-year survival of less than 10% [1]. Dismal survival among these patients is related to late diagnosis and lack of effective treatment [2]. Somatic and germline genetic mutations have been implicated in pancreatic cancer development. Activating gene mutations in oncogene KRAS and inactivating mutations in tumor suppressor genes TP53, CDKN2A and SMAD4 play a crucial role in the formation of pancreatic cancer [3]. Germline mutations in genes associated with hereditary cancer syndromes account for up to 10% of “heritable” pancreatic cancer [4,5,6].
Heritable pancreatic cancer can be broadly divided into hereditary pancreatic cancer syndromes and familial pancreatic cancer (FPC). Pancreatic cancers arising in patients with inherited genetic predisposition (hereditary breast and ovarian cancer syndrome, Lynch syndrome, Peutz Jeghers syndrome) that increases the risk of pancreatic cancer are defined as the hereditary pancreatic cancer syndromes. The underlying mutations for heritable pancreatic cancer syndromes are predominantly inherited in autosomal dominant patterns and affect the next generation with 50% probability. Therefore, such patients have a high risk of developing pancreatic cancer and need committed pancreatic cancer screening. In FPC, pancreatic cancers arise in patients with strong family history of pancreatic cancer (at least two first-degree relatives (FDRs)) without any known genetic predisposition syndromes. The risk of pancreatic cancer in FPC increases with more affected blood relatives.
Pancreatic cancer screening through the International Cancer of the Pancreas Screening (CAPS) Consortium guidelines, American Gastroenterological Association (AGA) guidelines and the American College of Gastroenterology guidelines (ACG) is designed to detect pancreatic cancer early and improve survival in such high-risk patients [7,8,9]. Almost 85% of patients with pancreatic ductal adenocarcinoma (PDAC) have locally advanced or metastatic disease at diagnosis [10]. About 15% of pancreatic cancers arise from intraductal papillary mucinous neoplasms (IPMNs) [11,12,13]. Therefore, the early detection of pre-cancerous pancreatic cysts through screening has gained intense focus in recent years to potentially lower PDAC-associated mortality.
Pancreatic cancer screening has revealed a high prevalence of IPMNs in high-risk populations; among patients with FPC, 10–18% patients had IPMNs [14,15]. Finding a potentially pre-malignant lesion in patients at high risk of cancers is anxiety provoking. Due to the lack of data on the natural progression of these IPMNs or their relationship with IPMNs in the general population, their management is provider dependent. Consequently, some providers have a very low threshold for surgery. A meta-analysis reported that two out of three pancreatic surgeries in patients undergoing screening did not have high-risk lesions or cancer [16]. Understanding the natural progression of IPMNs in such high-risk patients is necessary to develop evidence-based management guidelines. The first step towards this end is to determine the incidence and prevalence of IPMNs in such patient population [17]. This review summarizes the available literature on the incidence and prevalence of IPMNs in inherited genetic predisposition syndromes and FPC and speculates if IPMN and pancreatic cancer surveillance in these high-risk individuals needs to change.

2. Origins of PDAC

PDAC comprises the majority of all pancreatic cancers and arises from the exocrine pancreatic ductal cells. There are five precursor lesions of PDAC (Table 1): pancreatic intraepithelial neoplasias (PanINs), IPMNs, mucinous cystic neoplasms (MCNs), intraductal oncocytic papillary neoplasms (IOPNs) and intraductal tubulopapillary neoplasms (ITPNs) [18,19]. PanINs are small (<1 cm) epithelial neoplasms within the pancreatic duct with a variable degree of atypia [20,21]. All PanINs have KRAS mutations, with CDKN2A/p16 mutations often found in high-grade PanIN lesions [22,23,24]. Due to their small size, PanINs are not identified via radiological modalities and are thus not ideal targets for PDAC screening.
MCNs and IPMNs are >1 cm lesions, often incidentally found during cross-sectional imaging. These lesions are ideal for PDAC screening, and multiple guidelines on pancreatic cystic lesions characterize them into high- and low-risk cysts to guide management [26,27,28,29,30]. IPMNs are papillary projections of mucin-producing epithelium in the main pancreatic duct (main-duct IPMNs) or its branches (branch-duct IPMNs). Main-duct IPMNs and branch-duct IPMNs have 36–100% and 6–50% risk of developing PDAC, respectively [31]. The development of PDACs within IPMNs has been hypothesized to occur in one of three ways [32]:
(i)
Sequential pathway—sequential acquisition of driver and tumor suppressor gene mutations in IPMNs leading to PDACs;
(ii)
Branch-off pathway—after acquisition of key early genetic alterations, PDAC acquires new mutations not present in the IPMN;
(iii)
De novo pathway—Distinct PDAC that has unique mutations compared with the concomitant IPMN.
Evidence for sequential and branch-off pathways is derived from the presence of similar genetic mutations in IPMNs and PDACs (Table 2) [33]. Several studies have reported that while KRAS and GNAS are highly specific for early IPMNs, the accumulation of other pathogenic variants in a stepwise and straightforward fashion may transform IPMNs into PDAC [34,35,36,37]. Additionally, PDACs that arise from sequential or branch-off pathways have worse prognosis than the de novo PDACs [38].
Although GNAS and KRAS mutations are highly specific and sensitive for the diagnosis of IPMNs [47], there is substantial genetic heterogeneity among various driver mutations, and these mutations can exist within the same lesion [46,48]. Additionally, low-grade IPMNs have more genetic heterogeneity than IPMNs that have high-grade dysplasia [49]. IPMNs are multifocal, and 20% of patients have recurrence or new disease after resection [50,51,52,53]. This “field defect” of the pancreas that IPMNs herald along with high genetic heterogeneity suggests that the genetic pathway from IPMNs to PDAC is more complex than previously hypothesized. MCNs are unifocal mucinous lesions that lack the risk of recurrence; thus, no PDAC surveillance is necessary after surgical resection.
ITPNs are >1 cm intraductal nodular masses with minimal cystic formation or mucin production and are associated with PDAC in more than 50% of the cases [54,55]. These are rare and account for only few overall PDAC cases [56]. IOPNs are >1 cm cystic nodular lesions with oncocytic features and ductal differentiation and are associated with PDAC in 60% of the cases [57,58]. IOPNs are newly defined, and their significance in PDAC is still being elucidated.

3. Pancreatic Cancer Screening

The United States Preventative Services Task Force (USPSTF) guidelines do not recommend pancreatic cancer screening in the general population with average risk of pancreatic cancer [59]. However, high-risk individuals and patients with strong family history of pancreatic cancer require screening. The CAPS Consortium [7], AGA [8] and the ACG [9] have published guidelines on pancreatic cancer screening in such individuals (Table 3). The CAPS Consortium, AGA and ACG guidelines recommend screening patients with Peutz Jeghers syndrome and Familial Atypical Multiple Mole Melanoma (FAMMM), regardless of family history of pancreatic cancer. Both AGA and ACG guidelines also recommend pancreatic cancer screening in patients with Hereditary Pancreatitis regardless of family history of pancreatic cancer. All three guidelines recommend pancreatic cancer screening in BRCA2, PALB2, ATM and Lynch syndrome patients with at least one affected FDR. The National Comprehensive Cancer Network (NCCN) has similar guidelines for cancer screening in patients with hereditary genetic predisposition syndromes [60,61].
Among patients without such hereditary genetic predisposition syndromes, the guidelines differ in their recommendations. AGA guidelines recommend pancreatic cancer screening for patients with two affected blood relatives. The CAPS Consortium guidelines recommend pancreatic cancer screening among patients with two affected blood relatives provided one is an FDR. The ACG recommends cancer screening among patients with one affected FDR. These differences in cancer screening for FPC patients may be due to lack of data on FPC as described below. Screening should occur through endoscopic ultrasound (EUS), magnetic resonance imaging (MRI) or computed tomography (CT) at different ages depending on the underlying risk.
Overall, all pancreatic cancer screening guidelines focus only on family history of pancreatic cancer and omit history of IPMNs in family members. However, 42% patients with PDAC do not have a family history of PDAC and would be missed by these guidelines [62]. IPMNs, the potential pre-cursor lesions to PDAC, are more prevalent among patients with a history of hereditary genetic predisposition syndromes and individuals with FPC. While “familial” IPMNs have not been previously described, cases of hereditary IPMNs (affected mother and son) without any history of underlying extra-pancreatic malignancy or defined hereditary genetic predisposition syndrome have been reported [63]. While these data might not provide enough evidence, such findings suggest that perhaps hereditary genetic predisposition syndromes are related to IPMNs rather than PDAC. In turn, the cumulative genetic alterations in IPMNs may increase the risk of PDAC among such high-risk patients.

4. IPMNs in FPC

All pancreatic cancer screening guidelines recognize a strong family history of pancreatic cancer in the risk assessment for patients. The risk of pancreatic cancer increases by 4.6-, 6.4- and 32-fold for one, two and three or more affected FDRs, respectively [64]. The causative genetic mutations of FPC are still unknown. Numerous studies evaluating the efficacy of screening methods (endoscopic ultrasound, computer tomography, magnetic resonance imaging (MRI), endoscopic retrograde cholangiopancreatography) have utilized high-risk populations such as FPC patients. These studies have often noted a higher prevalence of IPMNs and other pancreas abnormalities such as cysts, duct dilations and nodules rather than pancreatic cancer (Table 4). Overall, these studies report IPMN incidence to be about 2-4-fold higher than that of pancreatic cancer.
Three studies of all the reports listed in Table 4 merit attention (Canto et al. 2002, Canto et al. 2006, and Ludwig et al. 2010) [14,65,66]. These studies had <10% non-FPC patients in their patient population, while the other reports included >30% of subjects with hereditary genetic predisposition syndrome. It is important to note that these three studies collectively report that the incidence of IPMNs is more than twice the incidence of pancreatic cancer among FPC patients. Currently, there is a lack of data purely focused on FPC patients. Additionally, Canto et al. 2006 reported that one-half of IPMNs detected on repeat screening had previous pancreatic abnormalities identified during initial screening [14]. Biopsy of an IPMN revealed in situ PDAC validating the sequential pathway of PDAC formation.
Table 4. The incidence of IPMNs and pancreatic cancer among patients with significant family history of pancreatic cancer.
Table 4. The incidence of IPMNs and pancreatic cancer among patients with significant family history of pancreatic cancer.
HRITotalIPMNsPC
Canto et al. 2002 [65]FPC or PJS381 (PJS)1 (FPC)
Canto et al. 2006 [14]FPC or PJS78 (72 FPC + 6 PJS)
149 controls
7
1 cyst in control
2
Poley et al. 2009 [67]FPC or genetic predisposition syndromes45 (13 FAMMM + 21 FPC + 3 HP + 2 PJS + 3 BRCA1 + 2 BRCA2 + 1 p53)72
Verna et al. 2010 [68]3 BRs or 2 FDRs or 2 BRs with 1 FDR or genetic predisposition syndrome2432
Ludwig et al. 2011 [66]1 FDR or 2 BRs or BRCA with FH of PC10951 (FDR)
Al-Sukhni et al. 2012 [69]2 FPCs or genetic predisposition syndromes or HP or FDR of double primary cancer patient262 (159 FPC + 7 PJS + 68 BRCA2 + 11 p16 + 5 BRCA1 + 2 HP + 10 double primary)15 (9 FPC, 4 BRCA2, 1 HP, 1 double primary)3 (2 FPC and 1 BRCA2)
Sud et al. 2014 [70]2 FDRs or 3 BRs or HP or PJS or p16 or Lynch with FH of PC1612
Chang et al. 2017 [71]Any BR303477/18 (pathological diagnosis)
Gangi et al. 2018 [72]2 BRs (including 1 FDR) or PJS or HP or FAMMM or BRCA2 mutations with FH58 (48 ≥ 1 FDR + 9 BRCA2 + 1 PJS)1 (2 FDR)0
HRI—high-risk individuals; IPMNs—intraductal papillary mucinous neoplasm; PC—pancreatic cancer; FPC—familial pancreatic cancer; PJS—Peutz Jeghers syndrome; BR—blood relative; FDR—first-degree relative; FH—family history; HP—hereditary pancreatitis; FAMMM—familial atypical multiple mole melanoma syndrome.

5. IPMNs in Hereditary Genetic Predisposition Syndromes

Along with FPC, certain hereditary genetic predisposition syndromes confer higher risk of pancreatic cancer in patients. Various case reports have identified IPMNs in patients with polycystic kidney disease [73,74], Lady Windermere Syndrome [75], parathyroid adenoma concerning for multiple endocrine neoplasia type 1 or type 2A, [76] cystic fibrosis [77], BRCA2 [78] and Lynch syndrome [79]. The underlying mutations in IPMNs were not analyzed in these case reports. Thus, the presence of IPMNs may be coincidental. Such case reports did not shed light on the prevalence of IPMNs or allowed a comparison to the prevalence of PDAC in these patients to be performed. Other studies have analyzed mutations in IPMNs and extrapancreatic malignancy among patients with hereditary syndromes and have reported similar genetic alterations (Table 5). A multivariate analysis revealed that germline mutations among patients with hereditary cancer syndromes was a predictor of the presence of IPMNs (relative risk, 3.2; 95% confidence interval (CI): 1.6–6.4) independent of family history of pancreatic cancer (p = 0.22) [80]. These data suggest that hereditary syndromes may predispose patients to IPMNs, which, in turn, increases the risk of PDAC formation.

5.1. McCune Albright Syndrome (MAS)

MAS is a rare, autosomal dominant syndrome resulting from an activating GNAS mutation. It presents with polycystic fibrous dysplasia, precocious puberty and cafe au lait spots. Genetic studies of IPMNs have noted that somatic GNAS mutations play an important role in IPMN formation. A total of 54 patients with MAS underwent contrast-enhanced MRI and magnetic resonance cholangiopancreatography (MRCP); IPMNs were identified in 25 (46%) patients [82]. The mean age of patients who had an IPMN detected was much lower, at 35.1 years, in this patient population than the mean age of 63 years in the general population [96,97]. Ten patients had worrisome or high-risk features; of note, among two patients who underwent surgical resection, one individual (age: 27 years) had a high-grade IPMN. These data suggest that underlying pathogenic germline GNAS mutations potentially increase the risk of IPMNs with advanced neoplasia in patients with MAS. In another study of 29 patients with MAS who underwent MRI/MRCP, 3 (16%) patients had an IPMN (mean age 27 years) [81]; one 50-year old patient with MAS had PDAC within an IPMN [83].

5.2. Lynch Syndrome/Hereditary Non-Polyposis Colorectal Cancer

Lynch syndrome is an autosomal dominant hereditary syndrome resulting from germline mutations in mismatch repair (MMR) genes (MLH1, MSH2, MSH6, PMS2 and EPCAM) that predispose patients to various malignancies, especially colon and endometrial cancer [98]. The relative risk of developing pancreatic cancer is from 9.0 to 11.0 for patients with these mutations [88,99]. Among 445 patients with PDAC, 9 patients had MMR gene deficiency and 3 of these patients had Lynch syndrome according to Bethesda criteria [100]. The study also reported that MMR deficiency occurred at a lower frequency in sporadic PDAC versus IPMN-related PDAC (1.6% vs. 6.9%, p = 0.02) [100]. Genetic analyses of IPMNs in Lynch syndrome patients have detected mutations in MSH2 and MSH6 [84,85,86]. While MMR gene deficiency occurs at a low frequency in IPMNs in the general population, the presence of similar MMR gene mutations in IPMNs and Lynch syndrome among these patients suggests that Lynch syndrome may activate IPMN formation, leading to PDAC formation [101].

5.3. Peutz Jeghers Syndrome (PJS)

PJS is an autosomal dominant hereditary syndrome characterized by mucocutaneous pigmentation and numerous gastrointestinal hamartomas. It is linked to germline mutations in the STK11 gene [102,103,104]. This mutation has been associated with an independent increase in the relative risk of 132 (95% CI: 44–261) for PDAC; as such, patients with PJS require screening regardless of family history [90]. A comparative mutational analysis of 22 IPMNs among patients with and without PJS demonstrated that STK11/LKB1 mutations were present in 100% (2/2) of samples from PJS patients versus only 25% (5/20) of samples from patients without PJS [89]. These data suggest that STK11/LKB1 mutation plays a role in IPMN formation. A molecular analysis of PDAC revealed that the loss of heterozygosity in tumor suppressor gene STK11/LKB1 plays an important role in both sporadic and familial pancreatic cancer [105]. Given the higher risk of PDAC in PJS and similar mutations in IPMNs and pancreatic cancer, STK11/LKB1 mutations noted in IPMNs among PJS patients may predispose this patient population to higher rates of progression to PDAC versus the general population.

5.4. Hereditary Breast and Ovarian Cancer Syndrome (HBOC)

BRCA1 and BRCA2 are tumor suppressor genes that code for a protein responsible for the repair of breaks in DNA strands [106]. Mutations in the BRCA2 gene predispose patients to breast and ovarian cancer and confer a relative risk from 3.0 to 9.0 to develop pancreatic cancer [107]. The pathogenesis of BRCA2 gene mutations that lead to PDAC remains unclear. Pancreatic cancer patients with a family history of pancreatic cancer have a higher prevalence of BRCA2 gene mutation than pancreatic cancer patients without a family history of pancreatic cancer (17–19% [108,109] vs. 5–10% [110,111], respectively). Screening all BRCA2 patients demonstrated a 17% prevalence of IPMNs compared with 1% in the general population [112]. The same study reported a 4% prevalence of PDAC among BRCA2 patients, which was considerably lower than the prevalence of IPMNs.
Similar to BRCA2, BRCA1 mutations also increase the risk of pancreatic cancer (relative risk, 2.26; 95% CI: 1.26–4.06) [113]. When patients with a BRCA1 mutation were screened using cross-sectional imaging, 8.3% (3/36) of individuals had an IPMN [112]. While both BRCA1 and BRCA2 mutations have been extensively studied relative to pancreatic cancer risk, these screening studies have also demonstrated a higher prevalence of IPMNs in patients with BRCA mutations.

5.5. Familial Atypical Multiple Mole Melanoma (FAMMM)

FAMMM is an autosomal dominant syndrome associated with a germline mutation in CDKN2A gene p16-Leiden resulting in familial melanomas and multiple atypical nevi. The CDKN2A p16-Leiden gene mutation predisposes patients to PDAC with a relative risk from 13.0 to 39.0 [114,115,116,117]. Among 19 families with FAMMM syndrome, 15 patients from 7 families had pancreatic cancer versus 0 patients from 8 families without FAMMM syndrome [114]. In a recent study, 77 patients with the p16-Leiden germline mutation were screened using MRI/MRCP, and 9% (7/77) had pancreatic cancer, while 11% (9/77) had cystic lesions that were not otherwise categorized [118]. Overall, FAMMM patients have a 17% cumulative risk of developing pancreatic cancer by age 75 [114]. The higher prevalence of IPMNs compared with pancreatic cancer among FAMMM patients suggests that the high risk of pancreatic cancer may be mediated through increased IPMN formation from the p16-Leiden mutation.

5.6. Familial Adenomatosis Polyposis (FAP)

FAP is an autosomal dominant hereditary syndrome secondary to germline mutations of the adenomatous polyposis coli (APC) gene resulting in early colonic carcinogenesis. APC gene mutation confers a relative risk of 4.46 (95% CI 1.2–11.4) to develop pancreatic cancer [119]. Genetic analyses of IPMNs among FAP patients have reported an absence of the APC protein that is typical of the adenomas in FAP patients elsewhere [91,92,93]. This body of evidence suggests that APC mutations that transform normal mucosa to adenomas to colon cancer may also play a role in increasing the risk of PDAC through mutations in IPMNs.

5.7. Carney Complex (CNC)

CNC is a hereditary syndrome that increases the risk of various tumors due to an alteration in the PRKAR1A gene mutation. In a CNC patient registry with 354 patients, 3 patients had an IPMN with a PRKAR1A mutation and 1 patient had PDAC [95]. This mutation has not typically been identified in IPMNs and may suggest a potential pathway for increased risk of PDAC in CNC patients.

6. Summary

IPMNs have an estimated prevalence of ~5% in the general population [120]. While previous single-center studies have suggested similar rates of IPMNs in the general population and high-risk patients, the current review suggests IPMN prevalence to be much higher among patients with hereditary genetic predisposition syndromes and FPC [17,121]. This is likely explained by a detection bias, since hereditary pancreatic cancer patients undergo more frequent imaging surveillance. Alternatively, the higher prevalence of pre-malignant lesions and pancreatic cancer in hereditary pancreatic cancer patients could be explained by a direct tumorigenesis pathway encompassing IPMNs and pancreatic cancer. One recent study reported that, compared with the general population, IPMNs among patients with FPC had shorter duration of progression to PDAC. This progression persisted despite the absence of traditional risk factors such as smoking, obesity, alcohol consumption and diabetes [122]. Nearly half of the high-risk individuals who have no pancreatic lesions may develop a neoplastic lesion in 11 months, and about half of high-risk individuals with prior known lesions may show rapid growth with progression beyond the pancreas within 21 months [123]. However, more studies are needed to corroborate these findings. There is growing evidence that IPMNs are the result of a neoplastic “field-defect” involving the entire ductal epithelium of the pancreas [124]. Mutations associated with hereditary pancreatic cancers may be contributing to such field-defect, resulting in IPMNs in some foci of the ductal epithelium while generating pancreatic cancer at another focus. Moreover, some of the higher-risk IPMNs may also progress to pancreatic cancer through the sequential pathway. Continued research is essential to better understand the prevalence and progression of IPMNs to pancreatic cancer in these patient populations; in the meantime, providers should continue to closely monitor high-risk patients as per current guidelines [7,8,9].

Author Contributions

D.R.A. was the primary author. D.R.A. and S.G.K. provided the study concept and design. S.R. and T.C. assisted with tables and performed revisions of the manuscript; P.P.S. and T.M.P. provided critical revision of the final manuscript. S.G.K. was provided critical revisions and input in the writing process. S.G.K. and D.R.A. provided approval of the final manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research study received no external funding.

Conflicts of Interest

Stanich P.P. receives research support from Emtora Biosciences, Freenome Holdings Inc., Janssen Pharmaceuticals Inc., Pfizer Inc. and the PTEN Research foundation. Krishna S.G. is PI of an investigator-initiated study. The study is in part funded by a grant to The Ohio State University Wexner Medical Center from Mauna Kea Technologies, Paris, France.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Kamisawa, T.; Wood, L.D.; Itoi, T.; Takaori, K. Pancreatic cancer. Lancet 2016, 388, 73–85. [Google Scholar] [CrossRef]
  3. Raphael, B.J.; Hruban, R.H.; Aguirre, A.J.; Moffitt, R.A.; Yeh, J.J.; Stewart, C.; Robertson, A.G.; Cherniack, A.D.; Gupta, M.; Getz, G. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 2017, 32, 185–203.e113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Zhen, D.B.; Rabe, K.G.; Gallinger, S.; Syngal, S.; Schwartz, A.G.; Goggins, M.G.; Hruban, R.H.; Cote, M.L.; McWilliams, R.R.; Roberts, N.J.; et al. BRCA1, BRCA2, PALB2, and CDKN2A mutations in familial pancreatic cancer: A PACGENE study. Genet. Med. 2015, 17, 569–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hiripi, E.; Bermejo, J.L.; Li, X.; Sundquist, J.; Hemminki, K. Familial association of pancreatic cancer with other malignancies in Swedish families. Br. J. Cancer 2009, 101, 1792–1797. [Google Scholar] [CrossRef] [Green Version]
  6. Brune, K.A.; Lau, B.; Palmisano, E.; Canto, M.; Goggins, M.G.; Hruban, R.H.; Klein, A.P. Importance of age of onset in pancreatic cancer kindreds. J. Natl. Cancer Inst. 2010, 102, 119–126. [Google Scholar] [CrossRef] [Green Version]
  7. Canto, M.I.; Harinck, F.; Hruban, R.H.; Offerhaus, G.J.; Poley, J.-W.; Kamel, I.; Nio, Y.; Schulick, R.S.; Bassi, C.; Kluijt, I.; et al. International Cancer of the Pancreas Screening (CAPS) Consortium summit on the management of patients with increased risk for familial pancreatic cancer. Gut 2013, 62, 339–347. [Google Scholar] [CrossRef]
  8. Aslanian, H.R.; Lee, J.H.; Canto, M.I. AGA Clinical Practice Update on Pancreas Cancer Screening in High-Risk Individuals: Expert Review. Gastroenterology 2020, 159, 358–362. [Google Scholar] [CrossRef]
  9. Syngal, S.; Brand, R.E.; Church, J.M.; Giardiello, F.M.; Hampel, H.L.; Burt, R.W. ACG clinical guideline: Genetic testing and management of hereditary gastrointestinal cancer syndromes. Am. J. Gastroenterol. 2015, 110, 223–263. [Google Scholar] [CrossRef] [Green Version]
  10. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1049. [Google Scholar] [CrossRef]
  11. Riall, T.S.; Stager, V.M.; Nealon, W.H.; Townsend, C.M.; Kuo, Y.-F.; Goodwin, J.S.; Freeman, J.L. Incidence of Additional Primary Cancers in Patients with Invasive Intraductal Papillary Mucinous Neoplasms and Sporadic Pancreatic Adenocarcinomas. J. Am. Coll. Surg. 2007, 204, 803–813. [Google Scholar] [CrossRef] [PubMed]
  12. Lubezky, N.; Ben-Haim, M.; Nakache, R.; Lahat, G.; Blachar, A.; Brazowski, E.; Santo, E.; Klausner, J.M. Clinical presentation can predict disease course in patients with intraductal papillary mucinous neoplasm of the pancreas. World J. Surg. 2010, 34, 126–132. [Google Scholar] [CrossRef] [PubMed]
  13. Singhi, A.D.; Koay, E.J.; Chari, S.T.; Maitra, A. Early Detection of Pancreatic Cancer: Opportunities and Challenges. Gastroenterology 2019, 156, 2024–2040. [Google Scholar] [CrossRef] [Green Version]
  14. Canto, M.I.; Goggins, M.; Hruban, R.H.; Petersen, G.M.; Giardiello, F.M.; Yeo, C.; Fishman, E.K.; Brune, K.; Axilbund, J.; Griffin, C.; et al. Screening for Early Pancreatic Neoplasia in High-Risk Individuals: A Prospective Controlled Study. Clin. Gastroenterol. Hepatol. 2006, 4, 766–781. [Google Scholar] [CrossRef]
  15. Shi, C.; Klein, A.P.; Goggins, M.; Maitra, A.; Canto, M.; Ali, S.; Schulick, R.; Palmisano, E.; Hruban, R.H. Increased prevalence of precursor lesions in familial pancreatic cancer patients. Clin. Cancer Res. 2009, 15, 7737–7743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Paiella, S.; Capurso, G.; Butturini, G.; Pezzilli, R.; Carrara, S.; Falconi, M.; Zerbi, A. Surveillance for pancreatic cancer in high-risk individuals: First-round screening results of a multicentric italian program. Pancreatology 2017, 17, S2. [Google Scholar] [CrossRef]
  17. Shah, I.; Silva-Santisteban, A.; Germansky, K.A.; Wadhwa, V.; Tung, N.; Huang, D.C.; Kandasamy, C.; Mlabasati, J.; Bilal, M.; Sawhney, M.S. Incidence and Prevalence of Intraductal Papillary Mucinous Neoplasms in Individuals with BRCA1 and BRCA2 Pathogenic Variant. J. Clin. Gastroenterol. 2022. [Google Scholar] [CrossRef]
  18. Singh, M.; Maitra, A. Precursor Lesions of Pancreatic Cancer: Molecular Pathology and Clinical Implications. Pancreatology 2007, 7, 9–19. [Google Scholar] [CrossRef]
  19. Singhi, A.D.; Maitra, A. The molecular pathology of precursor lesions of pancreatic cancer. In Pancreatic Cancer; Springer: Berlin/Heidelberg, Germany, 2018; pp. 147–176. [Google Scholar]
  20. Basturk, O.; Hong, S.-M.; Wood, L.D.; Adsay, N.V.; Albores-Saavedra, J.; Biankin, A.V.; Brosens, L.A.; Fukushima, N.; Goggins, M.; Hruban, R.H. A revised classification system and recommendations from the Baltimore consensus meeting for neoplastic precursor lesions in the pancreas. Am. J. Surg. Pathol. 2015, 39, 1730. [Google Scholar] [CrossRef]
  21. Yamao, K.; Yanagisawa, A.; Takahashi, K.; Kimura, W.; Doi, R.; Fukushima, N.; Ohike, N.; Shimizu, M.; Hatori, T.; Nobukawa, B. Clinicopathological features and prognosis of mucinous cystic neoplasm with ovarian-type stroma: A multi-institutional study of the Japan pancreas society. Pancreas 2011, 40, 67–71. [Google Scholar] [CrossRef]
  22. Kanda, M.; Matthaei, H.; Wu, J.; Hong, S.M.; Yu, J.; Borges, M.; Hruban, R.H.; Maitra, A.; Kinzler, K.; Vogelstein, B. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 2012, 142, 730–733.e739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Feldmann, G.; Beaty, R.; Hruban, R.H.; Maitra, A. Molecular genetics of pancreatic intraepithelial neoplasia. J. Hepato Biliary Pancreat. Surg. 2007, 14, 224–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Caldas, C.; Hahn, S.A.; da Costa, L.T.; Redston, M.S.; Schutte, M.; Seymour, A.B.; Weinstein, C.L.; Hruban, R.H.; Yeo, C.J.; Kern, S.E. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat. Genet. 1994, 8, 27–32. [Google Scholar] [CrossRef] [PubMed]
  25. Springer, S.; Wang, Y.; Dal Molin, M.; Masica, D.L.; Jiao, Y.; Kinde, I.; Blackford, A.; Raman, S.P.; Wolfgang, C.L.; Tomita, T. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology 2015, 149, 1501–1510. [Google Scholar] [CrossRef] [PubMed]
  26. Vege, S.S.; Ziring, B.; Jain, R.; Moayyedi, P.; Adams, M.A.; Dorn, S.D.; Dudley-Brown, S.L.; Flamm, S.L.; Gellad, Z.F.; Gruss, C.B. American gastroenterological association institute guideline on the diagnosis and management of asymptomatic neoplastic pancreatic cysts. Gastroenterology 2015, 148, 819–822. [Google Scholar] [CrossRef] [Green Version]
  27. Tanaka, M.; Fernández-del Castillo, C.; Kamisawa, T.; Jang, J.Y.; Levy, P.; Ohtsuka, T.; Salvia, R.; Shimizu, Y.; Tada, M.; Wolfgang, C.L. Revisions of international consensus Fukuoka guidelines for the management of IPMN of the pancreas. Pancreatology 2017, 17, 738–753. [Google Scholar] [CrossRef]
  28. Elta, G.H.; Enestvedt, B.K.; Sauer, B.G.; Lennon, A.M. ACG clinical guideline: Diagnosis and management of pancreatic cysts. Off. J. Am. Coll. Gastroenterol. ACG 2018, 113, 464–479. [Google Scholar] [CrossRef]
  29. European Study Group on Cystic Tumours of the Pancreas. European evidence-based guidelines on pancreatic cystic neoplasms. Gut 2018, 67, 789–804. [Google Scholar] [CrossRef]
  30. Megibow, A.J.; Baker, M.E.; Morgan, D.E.; Kamel, I.R.; Sahani, D.V.; Newman, E.; Brugge, W.R.; Berland, L.L.; Pandharipande, P.V. Management of incidental pancreatic cysts: A white paper of the ACR Incidental Findings Committee. J. Am. Coll. Radiol. 2017, 14, 911–923. [Google Scholar] [CrossRef]
  31. Tanaka, M. Intraductal Papillary Mucinous Neoplasm of the Pancreas as the Main Focus for Early Detection of Pancreatic Adenocarcinoma. Pancreas 2018, 47, 544–550. [Google Scholar] [CrossRef]
  32. Omori, Y.; Ono, Y.; Tanino, M.; Karasaki, H.; Yamaguchi, H.; Furukawa, T.; Enomoto, K.; Ueda, J.; Sumi, A.; Katayama, J.; et al. Pathways of Progression from Intraductal Papillary Mucinous Neoplasm to Pancreatic Ductal Adenocarcinoma Based on Molecular Features. Gastroenterology 2019, 156, 647–661.e642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Fischer, C.G.; Wood, L.D. From somatic mutation to early detection: Insights from molecular characterization of pancreatic cancer precursor lesions. J. Pathol. 2018, 246, 395–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Wu, J.; Matthaei, H.; Maitra, A.; Dal Molin, M.; Wood, L.D.; Eshleman, J.R.; Goggins, M.; Canto, M.I.; Schulick, R.D.; Edil, B.H.; et al. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci. Transl. Med. 2011, 3, 92ra66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Tamura, K.; Ohtsuka, T.; Date, K.; Fujimoto, T.; Matsunaga, T.; Kimura, H.; Watanabe, Y.; Miyazaki, T.; Ohuchida, K.; Takahata, S.; et al. Distinction of Invasive Carcinoma Derived from Intraductal Papillary Mucinous Neoplasms from Concomitant Ductal Adenocarcinoma of the Pancreas Using Molecular Biomarkers. Pancreas 2016, 45, 826–835. [Google Scholar] [CrossRef]
  36. Tan, M.C.; Basturk, O.; Brannon, A.R.; Bhanot, U.; Scott, S.N.; Bouvier, N.; LaFemina, J.; Jarnagin, W.R.; Berger, M.F.; Klimstra, D.; et al. GNAS and KRAS Mutations Define Separate Progression Pathways in Intraductal Papillary Mucinous Neoplasm-Associated Carcinoma. J. Am. Coll. Surg. 2015, 220, 845–854.e841. [Google Scholar] [CrossRef] [Green Version]
  37. Furukawa, T.; Kuboki, Y.; Tanji, E.; Yoshida, S.; Hatori, T.; Yamamoto, M.; Shibata, N.; Shimizu, K.; Kamatani, N.; Shiratori, K. Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas. Sci. Rep. 2011, 1, 161. [Google Scholar] [CrossRef] [Green Version]
  38. Yamaguchi, K.; Kanemitsu, S.; Hatori, T.; Maguchi, H.; Shimizu, Y.; Tada, M.; Nakagohri, T.; Hanada, K.; Osanai, M.; Noda, Y.; et al. Pancreatic ductal adenocarcinoma derived from IPMN and pancreatic ductal adenocarcinoma concomitant with IPMN. Pancreas 2011, 40, 571–580. [Google Scholar] [CrossRef]
  39. Uemura, K.; Hiyama, E.; Murakami, Y.; Kanehiro, T.; Ohge, H.; Sueda, T.; Yokoyama, T. Comparative analysis of K-ras point mutation, telomerase activity, and p53 overexpression in pancreatic tumours. Oncol. Rep. 2003, 10, 277–283. [Google Scholar] [CrossRef]
  40. Schönleben, F.; Qiu, W.; Bruckman, K.C.; Ciau, N.T.; Li, X.; Lauerman, M.H.; Frucht, H.; Chabot, J.A.; Allendorf, J.D.; Remotti, H.E. BRAF and KRAS gene mutations in intraductal papillary mucinous neoplasm/carcinoma (IPMN/IPMC) of the pancreas. Cancer Lett. 2007, 249, 242–248. [Google Scholar] [CrossRef] [Green Version]
  41. Reid, M.D.; Saka, B.; Balci, S.; Goldblum, A.S.; Adsay, N.V. Molecular genetics of pancreatic neoplasms and their morphologic correlates: An update on recent advances and potential diagnostic applications. Am. J. Clin. Pathol. 2014, 141, 168–180. [Google Scholar] [CrossRef] [Green Version]
  42. Amato, E.; Molin, M.D.; Mafficini, A.; Yu, J.; Malleo, G.; Rusev, B.; Fassan, M.; Antonello, D.; Sadakari, Y.; Castelli, P. Targeted next-generation sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas. J. Pathol. 2014, 233, 217–227. [Google Scholar] [CrossRef] [PubMed]
  43. Sakamoto, H.; Kuboki, Y.; Hatori, T.; Yamamoto, M.; Sugiyama, M.; Shibata, N.; Shimizu, K.; Shiratori, K.; Furukawa, T. Clinicopathological significance of somatic RNF43 mutation and aberrant expression of ring finger protein 43 in intraductal papillary mucinous neoplasms of the pancreas. Mod. Pathol. 2015, 28, 261–267. [Google Scholar] [CrossRef] [PubMed]
  44. Abe, T.; Fukushima, N.; Brune, K.; Boehm, C.; Sato, N.; Matsubayashi, H.; Canto, M.; Petersen, G.M.; Hruban, R.H.; Goggins, M. Genome-wide allelotypes of familial pancreatic adenocarcinomas and familial and sporadic intraductal papillary mucinous neoplasms. Clin. Cancer Res. 2007, 13, 6019–6025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Mueller, J.; Gansauge, S.; Mattfeldt, T. P53 mutation but not p16/MTS1 mutation occurs in intraductal papillary mucinous tumors of the pancreas. Hepatogastroenterology 2003, 50, 541–544. [Google Scholar]
  46. Noë, M.; Niknafs, N.; Fischer, C.G.; Hackeng, W.M.; Beleva Guthrie, V.; Hosoda, W.; Debeljak, M.; Papp, E.; Adleff, V.; White, J.R.; et al. Genomic characterization of malignant progression in neoplastic pancreatic cysts. Nat. Commun. 2020, 11, 4085. [Google Scholar] [CrossRef]
  47. Ren, R.; Krishna, S.G.; Chen, W.; Frankel, W.L.; Shen, R.; Zhao, W.; Avenarius, M.R.; Garee, J.; Caruthers, S.; Jones, D. Activation of the RAS pathway through uncommon BRAF mutations in mucinous pancreatic cysts without KRAS mutation. Mod. Pathol. 2021, 34, 438–444. [Google Scholar] [CrossRef]
  48. Attiyeh, M.; Zhang, L.; Iacobuzio-Donahue, C.; Allen, P.; Imam, R.; Basturk, O.; Klimstra, D.S.; Sigel, C.S. Simple mucinous cysts of the pancreas have heterogeneous somatic mutations. Hum. Pathol. 2020, 101, 1–9. [Google Scholar] [CrossRef]
  49. Fischer, C.G.; Beleva Guthrie, V.; Braxton, A.M.; Zheng, L.; Wang, P.; Song, Q.; Griffin, J.F.; Chianchiano, P.E.; Hosoda, W.; Niknafs, N.; et al. Intraductal Papillary Mucinous Neoplasms Arise from Multiple Independent Clones, Each with Distinct Mutations. Gastroenterology 2019, 157, 1123–1137.e1122. [Google Scholar] [CrossRef] [Green Version]
  50. He, J.; Cameron, J.L.; Ahuja, N.; Makary, M.A.; Hirose, K.; Choti, M.A.; Schulick, R.D.; Hruban, R.H.; Pawlik, T.M.; Wolfgang, C.L. Is it necessary to follow patients after resection of a benign pancreatic intraductal papillary mucinous neoplasm? J. Am. Coll. Surg. 2013, 216, 657–665. [Google Scholar] [CrossRef] [Green Version]
  51. Miyasaka, Y.; Ohtsuka, T.; Tamura, K.; Mori, Y.; Shindo, K.; Yamada, D.; Takahata, S.; Ishigami, K.; Ito, T.; Tokunaga, S.; et al. Predictive Factors for the Metachronous Development of High-risk Lesions in the Remnant Pancreas After Partial Pancreatectomy for Intraductal Papillary Mucinous Neoplasm. Ann. Surg. 2016, 263, 1180–1187. [Google Scholar] [CrossRef]
  52. Tanaka, M.; Fernández-del Castillo, C.; Adsay, V.; Chari, S.; Falconi, M.; Jang, J.-Y.; Kimura, W.; Levy, P.; Pitman, M.B.; Schmidt, C.M. International consensus guidelines 2012 for the management of IPMN and MCN of the pancreas. Pancreatology 2012, 12, 183–197. [Google Scholar] [CrossRef] [PubMed]
  53. Munigala, S.; Gelrud, A.; Agarwal, B. Risk of pancreatic cancer in patients with pancreatic cyst. Gastrointest. Endosc. 2016, 84, 81–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Rooney, S.L.; Shi, J. Intraductal tubulopapillary neoplasm of the pancreas: An update from a pathologist’s perspective. Arch. Pathol. Lab. Med. 2016, 140, 1068–1073. [Google Scholar] [CrossRef] [Green Version]
  55. Basturk, O.; Adsay, V.; Askan, G.; Dhall, D.; Zamboni, G.; Shimizu, M.; Cymes, K.; Carneiro, F.; Balci, S.; Sigel, C. Intraductal tubulopapillary neoplasm of the pancreas: A clinicopathologic and immunohistochemical analysis of 33 cases. Am. J. Surg. Pathol. 2017, 41, 313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Yamaguchi, H.; Kuboki, Y.; Hatori, T.; Yamamoto, M.; Shiratori, K.; Kawamura, S.; Kobayashi, M.; Shimizu, M.; Ban, S.; Koyama, I. Somatic mutations in PIK3CA and activation of AKT in intraductal tubulopapillary neoplasms of the pancreas. Am. J. Surg. Pathol. 2011, 35, 1812–1817. [Google Scholar] [CrossRef] [PubMed]
  57. Adsay, N.V.; Adair, C.F.; Heffess, C.S.; Klimstra, D.S. Intraductal oncocytic papillary neoplasms of the pancreas. Am. J. Surg. Pathol. 1996, 20, 980–994. [Google Scholar] [CrossRef]
  58. Marchegiani, G.; Mino-Kenudson, M.; Ferrone, C.R.; Warshaw, A.L.; Lillemoe, K.D.; Fernández-del Castillo, C. Oncocytic-type intraductal papillary mucinous neoplasms: A unique malignant pancreatic tumor with good long-term prognosis. J. Am. Coll. Surg. 2015, 220, 839–844. [Google Scholar] [CrossRef]
  59. Owens, D.K.; Davidson, K.W.; Krist, A.H.; Barry, M.J.; Cabana, M.; Caughey, A.B.; Curry, S.J.; Doubeni, C.A.; Epling, J.W.; Kubik, M. Screening for pancreatic cancer: US preventive services task force reaffirmation recommendation statement. JAMA 2019, 322, 438–444. [Google Scholar]
  60. Tempero, M.A. NCCN Guidelines Updates: Pancreatic Cancer. J. Natl. Compr. Cancer Netw. J. Natl. Compr. Cancer Netw. 2019, 17, 603–605. [Google Scholar] [CrossRef]
  61. Tempero, M.A.; Malafa, M.P.; Al-Hawary, M.; Behrman, S.W.; Benson, A.B.; Cardin, D.B.; Chiorean, E.G.; Chung, V.; Czito, B.; Del Chiaro, M.; et al. Pancreatic Adenocarcinoma, Version 2.2021, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2021, 19, 439–457. [Google Scholar] [CrossRef]
  62. Mandelker, D.; Zhang, L.; Kemel, Y.; Stadler, Z.K.; Joseph, V.; Zehir, A.; Pradhan, N.; Arnold, A.; Walsh, M.F.; Li, Y.; et al. Mutation Detection in Patients with Advanced Cancer by Universal Sequencing of Cancer-Related Genes in Tumor and Normal DNA vs Guideline-Based Germline Testing. JAMA 2017, 318, 825–835. [Google Scholar] [CrossRef] [PubMed]
  63. Denost, Q.; Chafai, N.; Arrive, L.; Mourra, N.; Paye, F. Hereditary intraductal papillary mucinous neoplasm of the pancreas. Clin. Res. Hepatol. Gastroenterol. 2012, 36, e23–e25. [Google Scholar] [CrossRef] [PubMed]
  64. Klein, A.P.; Brune, K.A.; Petersen, G.M.; Goggins, M.; Tersmette, A.C.; Offerhaus, G.J.; Griffin, C.; Cameron, J.L.; Yeo, C.J.; Kern, S.; et al. Prospective risk of pancreatic cancer in familial pancreatic cancer kindreds. Cancer Res. 2004, 64, 2634–2638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Canto, M.I.; Goggins, M.; Yeo, C.J.; Griffin, C.; Axilbund, J.E.; Brune, K.; Ali, S.Z.; Jagannath, S.; Petersen, G.M.; Fishman, E.K.; et al. Screening for pancreatic neoplasia in high-risk individuals: An EUS-based approach. Clin. Gastroenterol. Hepatol. 2004, 2, 606–621. [Google Scholar] [CrossRef]
  66. Ludwig, E.; Olson, S.H.; Bayuga, S.; Simon, J.; Schattner, M.A.; Gerdes, H.; Allen, P.J.; Jarnagin, W.R.; Kurtz, R.C. Feasibility and yield of screening in relatives from familial pancreatic cancer families. Am. J. Gastroenterol. 2011, 106, 946–954. [Google Scholar] [CrossRef] [Green Version]
  67. Poley, J.-W.; Kluijt, I.; Gouma, D.J.; Harinck, F.; Wagner, A.; Aalfs, C.; Van Eijck, C.; Cats, A.; Kuipers, E.; Nio, Y. The yield of first-time endoscopic ultrasonography in screening individuals at a high risk of developing pancreatic cancer. Off. J. Am. Coll. Gastroenterol. ACG 2009, 104, 2175–2181. [Google Scholar] [CrossRef]
  68. Verna, E.C.; Hwang, C.; Stevens, P.D.; Rotterdam, H.; Stavropoulos, S.N.; Sy, C.D.; Prince, M.A.; Chung, W.K.; Fine, R.L.; Chabot, J.A.; et al. Pancreatic Cancer Screening in a Prospective Cohort of High-Risk Patients: A Comprehensive Strategy of Imaging and Genetics. Clin. Cancer Res. 2010, 16, 5028. [Google Scholar] [CrossRef] [Green Version]
  69. Al-Sukhni, W.; Borgida, A.; Rothenmund, H.; Holter, S.; Semotiuk, K.; Grant, R.; Wilson, S.; Moore, M.; Narod, S.; Jhaveri, K. Screening for pancreatic cancer in a high-risk cohort: An eight-year experience. J. Gastrointest. Surg. 2012, 16, 771–783. [Google Scholar] [CrossRef]
  70. Sud, A.; Wham, D.; Catalano, M.; Guda, N.M. Promising Outcomes of Screening for Pancreatic Cancer by Genetic Testing and Endoscopic Ultrasound. Pancreas 2014, 43, 458–461. [Google Scholar] [CrossRef]
  71. Chang, M.-C.; Wu, C.-H.; Yang, S.-H.; Liang, P.-C.; Chen, B.-B.; Jan, I.S.; Chang, Y.-T.; Jeng, Y.-M. Pancreatic cancer screening in different risk individuals with family history of pancreatic cancer-a prospective cohort study in Taiwan. Am. J. Cancer Res. 2017, 7, 357–369. [Google Scholar]
  72. Gangi, A.; Malafa, M.; Klapman, J. Endoscopic Ultrasound–Based Pancreatic Cancer Screening of High-Risk Individuals: A Prospective Observational Trial. Pancreas 2018, 47, 586–591. [Google Scholar] [CrossRef] [PubMed]
  73. Sato, Y.; Mukai, M.; Sasaki, M.; Kitao, A.; Yoneda, N.; Kobayashi, D.; Imamura, Y.; Nakanuma, Y. Intraductal papillary–mucinous neoplasm of the pancreas associated with polycystic liver and kidney disease. Pathol. Int. 2009, 59, 201–204. [Google Scholar] [CrossRef] [PubMed]
  74. Pipaliya, N.; Rathi, C.; Parikh, P.; Patel, R.; Ingle, M.; Sawant, P. A Rare Case of an Intraductal Papillary Mucinous Neoplasm of Pancreas Fistulizing Into Duodenum with Adult Polycystic Kidney Disease. Gastroenterol. Res. 2015, 8, 197–200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Nguyen, V.X.; Decker, G.A.; Das, A.; Harrison, M.E.; Silva, A.C.; Ocal, I.T.; Collins, J.M.; Nguyen, C.C. The Natural History of a Branch Duct Intraductal Papillary Mucinous Neoplasm in a Patient with Lady Windermere Syndrome: A Case Reports. JOP J. Pancreas 2010, 11, 249–254. [Google Scholar]
  76. Gilshtein, H.; Mekel, M.; Kluger, Y. IPMN and Parathyroid Adenoma: An Interesting Association. JOP J. Pancreas 2012, 13, 542. [Google Scholar]
  77. Pagliari, D.; Saviano, A.; Serricchio, M.; Dal Lago, A.; Brizi, M.; Manfredi, R.; Costamagna, G.; Attili, F. The association of pancreatic cystosis and IPMN in cystic fibrosis: Case report and literature review. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 5179–5184. [Google Scholar]
  78. Bhandari, B.S.; Kaila, V.; Saad, A.B.; Khalil, E.; Som, A.; Munjal, A.; Kannadath, B.S.; Thosani, N.C. Intraductal Papillary Mucinous Neoplasm (IPMN) and BRCA Mutation: A Case Report: 2827. Off. J. Am. Coll. Gastroenterol. ACG 2017, 112, S1519. [Google Scholar] [CrossRef]
  79. Flanagan, M.R.; Jayaraj, A.; Xiong, W.; Yeh, M.M.; Raskind, W.H.; Pillarisetty, V.G. Pancreatic intraductal papillary mucinous neoplasm in a patient with Lynch syndrome. World J. Gastroenterol. 2015, 21, 2820–2825. [Google Scholar] [CrossRef]
  80. Roch, A.M.; Al-Temimi, M.H.; Nguyen, T.; House, M.G.; Zyromski, N.J.; Nakeeb, A.; Schmidt, C.M.; Ceppa, E.P. Patients with deleterious germline mutations: A heterogeneous population for pancreatic cancer screening? HPB 2020, 22, S13. [Google Scholar] [CrossRef]
  81. Gaujoux, S.; Salenave, S.; Ronot, M.; Rangheard, A.-S.; Cros, J.; Belghiti, J.; Sauvanet, A.; Ruszniewski, P.; Chanson, P. Hepatobiliary and Pancreatic Neoplasms in Patients With McCune-Albright Syndrome. J. Clin. Endocrinol. Metab. 2014, 99, E97–E101. [Google Scholar] [CrossRef] [Green Version]
  82. Robinson, C.; Estrada, A.; Zaheer, A.; Singh, V.K.; Wolfgang, C.L.; Goggins, M.G.; Hruban, R.H.; Wood, L.D.; Noë, M.; Montgomery, E.A.; et al. Clinical and Radiographic Gastrointestinal Abnormalities in McCune-Albright Syndrome. J. Clin. Endocrinol. Metab. 2018, 103, 4293–4303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Gaujoux, S.; Pasmant, E.; Silve, C.; Mehsen-Cetre, N.; Coriat, R.; Rouquette, A.; Douset, B.; Prat, F.; Leroy, K. McCune Albright syndrome is a genetic predisposition to intraductal papillary and mucinous neoplasms of the pancreas associated pancreatic cancer in relation with GNAS somatic mutation—a case report. Medicine 2019, 98, e18102. [Google Scholar] [CrossRef] [PubMed]
  84. Sparr, J.A.; Bandipalliam, P.; Redston, M.S.; Syngal, S. Intraductal papillary mucinous neoplasm of the pancreas with loss of mismatch repair in a patient with Lynch syndrome. Am. J. Surg. Pathol. 2009, 33, 309–312. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Lee, S.H.; Kim, W.Y.; Hwang, D.-Y.; Han, H.S. Intraductal papillary mucinous neoplasm of the ileal heterotopic pancreas in a patient with hereditary non-polyposis colorectal cancer: A case report. World J. Gastroenterol. 2015, 21, 7916–7920. [Google Scholar] [CrossRef]
  86. Hackeng, W.M.; de Guerre, L.E.V.M.; Kuypers, K.C.; Snoek, A.M.; Morsink, F.H.; Offerhaus, G.J.A.; Brosens, L.A.A. Pseudomyxoma Peritonei After a Total Pancreatectomy for Intraductal Papillary Mucinous Neoplasm with Colloid Carcinoma in Lynch Syndrome. Pancreas 2019, 48, 135–138. [Google Scholar] [CrossRef]
  87. Leoz, M.L.; Sánchez, A.; Carballal, S.; Ruano, L.; Ocaña, T.; Pellisé, M.; Castells, A.; Balaguer, F.; Moreira, L. Hereditary gastric and pancreatic cancer predisposition syndromes. Gastroenterol. Y Hepatol. 2016, 39, 481–493. [Google Scholar] [CrossRef]
  88. Kastrinos, F.; Mukherjee, B.; Tayob, N.; Wang, F.; Sparr, J.; Raymond, V.M.; Bandipalliam, P.; Stoffel, E.M.; Gruber, S.B.; Syngal, S. Risk of pancreatic cancer in families with Lynch syndrome. JAMA 2009, 302, 1790–1795. [Google Scholar] [CrossRef]
  89. Sato, N.; Rosty, C.; Jansen, M.; Fukushima, N.; Ueki, T.; Yeo, C.J.; Cameron, J.L.; Iacobuzio-Donahue, C.A.; Hruban, R.H.; Goggins, M. STK11/LKB1 Peutz-Jeghers Gene Inactivation in Intraductal Papillary-Mucinous Neoplasms of the Pancreas. Am. J. Pathol. 2001, 159, 2017–2022. [Google Scholar] [CrossRef] [Green Version]
  90. Giardiello, F.M.; Brensinger, J.D.; Tersmette, A.C.; Goodman, S.N.; Petersen, G.M.; Booker, S.V.; Cruz-Correa, M.; Offerhaus, J.A. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 2000, 119, 1447–1453. [Google Scholar] [CrossRef] [Green Version]
  91. Chetty, R.; Salahshor, S.; Bapat, B.; Berk, T.; Croitoru, M.; Gallinger, S. Intraductal papillary mucinous neoplasm of the pancreas in a patient with attenuated familial adenomatous polyposis. J. Clin. Pathol. 2005, 58, 97–101. [Google Scholar] [CrossRef]
  92. Maire, F.; Hammel, P.; Terris, B.; Olschwang, S.; O’Toole, D.; Sauvanet, A.; Palazzo, L.; Ponsot, P.; Laplane, B.; Lévy, P. Intraductal papillary and mucinous pancreatic tumour: A new extracolonic tumour in familial adenomatous polyposis. Gut 2002, 51, 446–449. [Google Scholar] [CrossRef] [PubMed]
  93. Sudo, T.; Murakami, Y.; Uemura, K.; Hayashidani, Y.; Takesue, Y.; Sueda, T. Development of an Intraductal Papillary-Mucinous Neoplasm of the Pancreas in a Patient With Familial Adenomatous Polyposis. Pancreas 2005, 31, 428–429. [Google Scholar] [CrossRef] [PubMed]
  94. Moussata, D.; Senouci, L.; Berger, F.; Scoazec, J.-Y.; Pinson, S.; Walter, T.; Lombard-Bohas, C.; Saurin, J.-C. Familial Adenomatous Polyposis and Pancreatic Cancer. Pancreas 2015, 44, 512–513. [Google Scholar] [CrossRef] [PubMed]
  95. Gaujoux, S.; Tissier, F.; Ragazzon, B.; Rebours, V.; Saloustros, E.; Perlemoine, K.; Vincent-Dejean, C.; Meurette, G.; Cassagnau, E.; Dousset, B.; et al. Pancreatic Ductal and Acinar Cell Neoplasms in Carney Complex: A Possible New Association. J. Clin. Endocrinol. Metab. 2011, 96, E1888–E1895. [Google Scholar] [CrossRef] [Green Version]
  96. Hruban, R.H.; Pitman, M.B.; Klimstra, D.S. Tumors of the Pancreas; American Registry of Pathology in Collaboration with the Armed Force: Washington, DC, USA, 2007. [Google Scholar]
  97. Zamboni, G.; Hirabayashi, K.; Castelli, P.; Lennon, A.M. Precancerous lesions of the pancreas. Best Pract. Res. Clin. Gastroenterol. 2013, 27, 299–322. [Google Scholar] [CrossRef]
  98. Vasen, H.F.; Watson, P.; Mecklin, J.P.; Lynch, H.T. New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC. Gastroenterology 1999, 116, 1453–1456. [Google Scholar] [CrossRef]
  99. Win, A.K.; Young, J.P.; Lindor, N.M.; Tucker, K.M.; Ahnen, D.J.; Young, G.P.; Buchanan, D.D.; Clendenning, M.; Giles, G.G.; Winship, I. Colorectal and other cancer risks for carriers and noncarriers from families with a DNA mismatch repair gene mutation: A prospective cohort study. J. Clin. Oncol. 2012, 30, 958. [Google Scholar] [CrossRef] [Green Version]
  100. Lupinacci, R.M.; Goloudina, A.; Buhard, O.; Bachet, J.-B.; Maréchal, R.; Demetter, P.; Cros, J.; Bardier-Dupas, A.; Collura, A.; Cervera, P.; et al. Prevalence of Microsatellite Instability in Intraductal Papillary Mucinous Neoplasms of the Pancreas. Gastroenterology 2018, 154, 1061–1065. [Google Scholar] [CrossRef]
  101. Handra-Luca, A.; Couvelard, A.; Degott, C.; Fléjou, J.-F. Correlation between patterns of DNA mismatch repair hmlh1 and hmsh2 protein expression and progression of dysplasia in intraductal papillary mucinous neoplasms of the pancreas. Virchows Arch. 2004, 444, 235–238. [Google Scholar] [CrossRef]
  102. Hemminki, A.; Tomlinson, I.; Markie, D.; Järvinen, H.; Sistonen, P.; Björkqvist, A.-M.; Knuutila, S.; Salovaara, R.; Bodmer, W.; Shibata, D. Localization of a susceptibility locus for Peutz-Jeghers syndrome to 19p using comparative genomic hybridization and targeted linkage analysis. Nat. Genet. 1997, 15, 87–90. [Google Scholar] [CrossRef]
  103. Hemminki, A.; Markie, D.; Tomlinson, I.; Avizienyte, E.; Roth, S.; Loukola, A.; Bignell, G.; Warren, W.; Aminoff, M.; Höglund, P. A serine/threonine kinase gene defective in Peutz–Jeghers syndrome. Nature 1998, 391, 184–187. [Google Scholar] [CrossRef] [PubMed]
  104. Jenne, D.E.; Reomann, H.; Nezu, J.-I.; Friedel, W.; Loff, S.; Jeschke, R.; Müller, O.; Back, W.; Zimmer, M. Peutz-Jeghers syndrome is caused by mutations in a novel serine threoninekinase. Nat. Genet. 1998, 18, 38–43. [Google Scholar] [CrossRef] [PubMed]
  105. Su, G.H.; Hruban, R.H.; Bansal, R.K.; Bova, G.S.; Tang, D.J.; Shekher, M.C.; Westerman, A.M.; Entius, M.M.; Goggins, M.; Yeo, C.J.; et al. Germline and Somatic Mutations of the STK11/LKB1 Peutz-Jeghers Gene in Pancreatic and Biliary Cancers. Am. J. Pathol. 1999, 154, 1835–1840. [Google Scholar] [CrossRef] [Green Version]
  106. Sharan, S.K.; Morimatsu, M.; Albrecht, U.; Lim, D.-S.; Regel, E.; Dinh, C.; Sands, A.; Eichele, G.; Hasty, P.; Bradley, A. Embryonic lethality and radiation hypersensitivity mediated by Rad51 in mice lacking Brca2. Nature 1997, 386, 804–810. [Google Scholar] [CrossRef] [PubMed]
  107. Consortium, B. Cancer risks in BRCA2 mutation carriers. J. Natl. Cancer Inst. 1999, 91, 1310–1316. [Google Scholar] [CrossRef]
  108. Murphy, K.M.; Brune, K.A.; Griffin, C.; Sollenberger, J.E.; Petersen, G.M.; Bansal, R.; Hruban, R.H.; Kern, S.E. Evaluation of candidate genes MAP2K4, MADH4, ACVR1B, and BRCA2 in familial pancreatic cancer: Deleterious BRCA2 mutations in 17%. Cancer Res. 2002, 62, 3789–3793. [Google Scholar]
  109. Hahn, S.A.; Greenhalf, B.; Ellis, I.; Sina-Frey, M.; Rieder, H.; Korte, B.; Gerdes, B.; Kress, R.; Ziegler, A.; Raeburn, J.A. BRCA2 germline mutations in familial pancreatic carcinoma. J. Natl. Cancer Inst. 2003, 95, 214–221. [Google Scholar] [CrossRef]
  110. Goggins, M.; Schutte, M.; Lu, J.; Moskaluk, C.A.; Weinstein, C.L.; Petersen, G.M.; Yeo, C.J.; Jackson, C.E.; Lynch, H.T.; Hruban, R.H. Germline BRCA2 gene mutations in patients with apparently sporadic pancreatic carcinomas. Cancer Res. 1996, 56, 5360–5364. [Google Scholar]
  111. Ozcelik, H.; Schmocker, B.; Di Nicola, N.; Shi, X.; Langer, B.; Moore, M.; Taylor, B.; Narod, S.; Darlington, G.; Andrulis, I. Increased carrier rate of germline BRCA2 6174delT mutations in Jewish individuals with pancreatic cancer. Nat. Genet. 1997, 16, 17–18. [Google Scholar] [CrossRef]
  112. Roch, A.M.; Schneider, J.; Carr, R.A.; Lancaster, W.P.; House, M.G.; Zyromski, N.J.; Nakeeb, A.; Schmidt, C.M.; Ceppa, E.P. Are BRCA1 and BRCA2 gene mutation patients underscreened for pancreatic adenocarcinoma? J. Surg. Oncol. 2019, 119, 777–783. [Google Scholar] [CrossRef]
  113. Thompson, D.; Easton, D.F. Cancer incidence in BRCA1 mutation carriers. J. Natl. Cancer Inst. 2002, 94, 1358–1365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Vasen, H.; Gruis, N.; Frants, R.; van Der Velden, P.; Hille, E.; Bergman, W. Risk of developing pancreatic cancer in families with familial atypical multiple mole melanoma associated with a specific 19 deletion of p16 (p16-Leiden). Int. J. Cancer 2000, 87, 809–811. [Google Scholar] [CrossRef]
  115. Goldstein, A.M.; Fraser, M.C.; Struewing, J.P.; Hussussian, C.J.; Ranade, K.; Zametkin, D.P.; Fontaine, L.S.; Organic, S.M.; Dracopoli, N.C.; Clark Jr, W.H. Increased risk of pancreatic cancer in melanoma-prone kindreds with p16 INK4 mutations. N. Engl. J. Med. 1995, 333, 970–975. [Google Scholar] [CrossRef] [PubMed]
  116. Hille, E.T.; van Duijn, E.; Gruis, N.A.; Rosendaal, F.R.; Bergman, W.; Vandenbroucke, J.P. Excess cancer mortality in six Dutch pedigrees with the familial atypical multiple mole-melanoma syndrome from 1830 to 1994. J. Investig. Dermatol. 1998, 110, 788–792. [Google Scholar] [CrossRef] [Green Version]
  117. Borg, A.k.; Sandberg, T.; Nilsson, K.; Johannsson, O.; Klinker, M.; Måsbäck, A.; Westerdahl, J.; Olsson, H.k.; Ingvar, C. High frequency of multiple melanomas and breast and pancreas carcinomas in CDKN2A mutation-positive melanoma families. J. Natl. Cancer Inst. 2000, 92, 1260–1266. [Google Scholar] [CrossRef] [Green Version]
  118. Vasen, H.F.A.; Wasser, M.; van Mil, A.; Tollenaar, R.A.; Konstantinovski, M.; Gruis, N.A.; Bergman, W.; Hes, F.J.; Hommes, D.W.; Offerhaus, G.J.A.; et al. Magnetic Resonance Imaging Surveillance Detects Early-Stage Pancreatic Cancer in Carriers of a p16-Leiden Mutation. Gastroenterology 2011, 140, 850–856. [Google Scholar] [CrossRef]
  119. Giardiello, F.; Offerhaus, G.; Lee, D.; Krush, A.; Tersmette, A.; Booker, S.; Kelley, N.; Hamilton, S. Increased risk of thyroid and pancreatic carcinoma in familial adenomatous polyposis. Gut 1993, 34, 1394–1396. [Google Scholar] [CrossRef] [Green Version]
  120. Zerboni, G.; Signoretti, M.; Crippa, S.; Falconi, M.; Arcidiacono, P.G.; Capurso, G. Systematic review and meta-analysis: Prevalence of incidentally detected pancreatic cystic lesions in asymptomatic individuals. Pancreatology 2019, 19, 2–9. [Google Scholar] [CrossRef]
  121. Kromrey, M.-L.; Bülow, R.; Hübner, J.; Paperlein, C.; Lerch, M.M.; Ittermann, T.; Völzke, H.; Mayerle, J.; Kühn, J.-P. Prospective study on the incidence, prevalence and 5-year pancreatic-related mortality of pancreatic cysts in a population-based study. Gut 2018, 67, 138–145. [Google Scholar] [CrossRef] [Green Version]
  122. Abe, K.; Kitago, M.; Kosaki, K.; Yamada, M.; Iwasaki, E.; Kawasaki, S.; Mizukami, K.; Momozawa, Y.; Terao, C.; Yagi, H.; et al. Genomic analysis of familial pancreatic cancers and intraductal papillary mucinous neoplasms: A cross-sectional study. Cancer Sci. 2022, 113, 1821–1829. [Google Scholar] [CrossRef]
  123. Overbeek, K.A.; Goggins, M.G.; Dbouk, M.; Levink, I.J.M.; Koopmann, B.D.M.; Chuidian, M.; Konings, I.C.A.W.; Paiella, S.; Earl, J.; Fockens, P.; et al. Timeline of Development of Pancreatic Cancer and Implications for Successful Early Detection in High-Risk Individuals. Gastroenterology 2022, 162, 772–785.e774. [Google Scholar] [CrossRef] [PubMed]
  124. Miller, F.H.; Vendrami, C.L.; Recht, H.S.; Wood, C.G.; Mittal, P.; Keswani, R.N.; Gabriel, H.; Borhani, A.A.; Nikolaidis, P.; Hammond, N.A. Pancreatic Cystic Lesions and Malignancy: Assessment, Guidelines, and the Field Defect. RadioGraphics 2022, 42, 87–105. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of pre-cursor lesions.
Table 1. Summary of pre-cursor lesions.
LesionSomatic Mutations [13,18,20,23,25]SizeRisk of Development into PDACPrevalence of Progression to PDAC [16,17]
PanINKRAS, CDKN2A/p(16) (High Grade)<1 cm<1% (Low grade), 40% (High grade)~80%
IPMNKRAS, GNAS, CDKN2A/p16, TP53, SMAD4>1 cm6–50% (Branch duct), 36–100% (Main duct)15%
MCNATM/GL13>1 cm<1%5–10%
IOPNPRKACA, PRKACB>1 cm60%<1%
ITPN->1 cm50%<1%
PanIN—pancreatic intraepithelial neoplasia; IPMN—intraductal papillary mucinous cystic neoplasm; MCN—mucinous cystic neoplasm; IOPN—intraductal oncocytic papillary neoplasm; ITPN—intraductal tubulopapillary neoplasm.
Table 2. Prevalence of somatic mutations in IPMNs that are typically found in PDACs.
Table 2. Prevalence of somatic mutations in IPMNs that are typically found in PDACs.
Genetic MutationPrevalence in IPMNs
KRAS30–80% [39,40]
GNAS40–79% [25,37,41,42]
RNF4314–38% [25,37,42,43]
CDKN2A/p16, TP53, SMAD440% [44,45]
GLI328% [46] (Combined prevalence in IPMNs/MCNs)
ATM17% [46] (Combined prevalence in IPMNs/MCNs)
IPMN—intraductal papillary mucinous neoplasm; MCN—mucinous cystic neoplasm.
Table 3. Screening guidelines for hereditary pancreatic cancer patients according to the International Cancer of the Pancreas Screening (CAPS) Consortium guidelines, American Gastroenterological Association (AGA) guidelines and the American College of Gastroenterology (ACG) guidelines [7,8,9].
Table 3. Screening guidelines for hereditary pancreatic cancer patients according to the International Cancer of the Pancreas Screening (CAPS) Consortium guidelines, American Gastroenterological Association (AGA) guidelines and the American College of Gastroenterology (ACG) guidelines [7,8,9].
Hereditary Genetic Predisposition SyndromesAffected Family Members
No FH1 FDR2 BR
Peutz Jeghers syndromeCAPS/AGA/ACG
FAMMM (CDKN2A/p16)CAPS/AGA/ACG
Hereditary pancreatitis (PRSS1)AGA/ACG
BRCA2 CAPS/AGA/ACG
BRCA1 AGA/ACG
PALB2 CAPS/AGA/ACG
ATM CAPS/AGA/ACG
Lynch syndrome CAPS/AGA/ACG
None ACGCAPS */AGA
* Includes at least one first-degree relative (FH—family history; FDR—first-degree relative; BR—blood relative).
Table 5. Prevalence/incidence of IPMNs and pancreatic cancer in hereditary genetic predisposition syndromes.
Table 5. Prevalence/incidence of IPMNs and pancreatic cancer in hereditary genetic predisposition syndromes.
Gene MutationIPMNsPC
MASGNAS16–46% [81,82]1 case [83]
LynchMLH1, MSH2, MSH63 cases [84,85,86]0.7–3.7% [87,88]
PJSSTK11/LKB1100% [89]11–36% [90]
FAPAPC, MUTYH3 cases [91,92,93]3% (4/127 including 2 endocrine carcinomas, 1 acinar cell carcinoma, 1 pancreatoblastoma) [94]
CNCPRKAR1A0.8% [95]1.7% (6/354 with only 1 PDAC) [95]
MAS—McCune Albright syndrome; PJS—Peutz Jeghers syndrome; FAP—familial adenomatosis polyposis; CNC—Carney complex syndrome; IPMNs—intraductal papillary mucinous neoplasms; PC—pancreatic cancer; PDAC—pancreatic ductal adenocarcinoma.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ardeshna, D.R.; Rangwani, S.; Cao, T.; Pawlik, T.M.; Stanich, P.P.; Krishna, S.G. Intraductal Papillary Mucinous Neoplasms in Hereditary Cancer Syndromes. Biomedicines 2022, 10, 1475. https://doi.org/10.3390/biomedicines10071475

AMA Style

Ardeshna DR, Rangwani S, Cao T, Pawlik TM, Stanich PP, Krishna SG. Intraductal Papillary Mucinous Neoplasms in Hereditary Cancer Syndromes. Biomedicines. 2022; 10(7):1475. https://doi.org/10.3390/biomedicines10071475

Chicago/Turabian Style

Ardeshna, Devarshi R., Shiva Rangwani, Troy Cao, Timothy M. Pawlik, Peter P. Stanich, and Somashekar G. Krishna. 2022. "Intraductal Papillary Mucinous Neoplasms in Hereditary Cancer Syndromes" Biomedicines 10, no. 7: 1475. https://doi.org/10.3390/biomedicines10071475

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

Article Metrics

Back to TopTop