Review Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jun 7, 2021; 27(21): 2710-2726
Published online Jun 7, 2021. doi: 10.3748/wjg.v27.i21.2710
Molecular alterations in pancreatic tumors
Michela Visani, Giorgia Acquaviva, Antonio De Leo, Viviana Sanza, Lidia Merlo, Thais Maloberti, Giovanni Tallini, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna–Molecular Diagnostic Unit, Azienda USL di Bologna, Bologna 40138, Italy
Antonio De Leo, Annalisa Pession, Giovanni Tallini, Dario de Biase, Division of Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40138, Italy
Alba A Brandes, Enrico Franceschi, Monica Di Battista, Medical Oncology Department, Azienda USL/IRCCS Istituto Delle Scienze Neurologiche di Bologna, Bologna 40139, Italy
Michele Masetti, Elio Jovine, Division of Surgery, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna 40133, Italy
Sirio Fiorino, Internal Medicine Unit, Budrio Hospital Azienda USL, Bologna 40133, Italy
Annalisa Pession, Dario de Biase, Department of Pharmacy and Biotechnology, University of Bologna, Bologna 40138, Italy
ORCID number: Michela Visani (0000-0002-9051-2231); Giorgia Acquaviva (0000-0002-8811-7865); Antonio De Leo (0000-0002-3761-5135); Viviana Sanza (0000-0001-9889-8776); Lidia Merlo (0000-0003-0301-1103); Thais Maloberti (0000-0002-8306-4653); Alba A Brandes (0000-0002-2503-9089); Enrico Franceschi (0000-0001-9332-4677); Monica Di Battista (0000-0001-0000-0001); Michele Masetti (0000-0003-2259-8675); Elio Jovine (0000-0002-1514-7772); Sirio Fiorino (0000-0001-5755-2197); Annalisa Pession (0000-0001-9035-9698); Giovanni Tallini (0000-0003-0113-6682); Dario de Biase (0000-0002-0609-8817).
Author contributions: de Biase D conceived and designed the study; Visani M, Acquaviva G, De Leo A, Sanza V, Merlo L, Maloberti T, Franceschi E, Di Battista M, Masetti M, Fiorino S and de Biase D performed the literature search and acquisition of the data; Brandes AA, Jovine E, Pession A and Tallini G performed the interpretation and the critical revision of the data; Visani M and de Biase D wrote the manuscript; all authors have read and approved the final manuscript.
Conflict-of-interest statement: Authors have nothing to declare.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Dario de Biase, MSc, PhD, Assistant Professor, Department of Pharmacy and Biotechnology, University of Bologna, viale Ercolani 4/2, Bologna 40138, Italy. dario.debiase@unibo.it
Received: January 17, 2021
Peer-review started: January 17, 2021
First decision: February 9, 2021
Revised: February 25, 2021
Accepted: April 14, 2021
Article in press: April 14, 2021
Published online: June 7, 2021

Abstract

Genetic alterations in pancreatic tumors can usually be classified in: (1) Mutational activation of oncogenes; (2) Inactivation of tumor suppressor genes; and (3) Inactivation of genome maintenance genes controlling the repair of DNA damage. Endoscopic ultrasound-guided fine-needle aspiration has improved pre-operative diagnosis, but the management of patients with a pancreatic lesion is still challenging. Molecular testing could help mainly in solving these “inconclusive” specimens. The introduction of multi-gene analysis approaches, such as next-generation sequencing, has provided a lot of useful information on the molecular characterization of pancreatic tumors. Different types of pancreatic tumors (e.g., pancreatic ductal adenocarcinomas, intraductal papillary mucinous neoplasms, solid pseudopapillary tumors) are characterized by specific molecular alterations. The aim of this review is to summarize the main molecular alterations found in pancreatic tumors.

Key Words: Pancreatic tumors, Molecular alterations, Pancreatic ductal adenocarcinomas, Intraductal papillary mucinous neoplasm, Mutations, Molecular markers

Core Tip: To date, in patients with pancreatic cancer there are no targetable molecules for personalized treatment in clinical practice. The introduction of multi-gene analysis approaches, such as massive parallel sequencing, has provided a lot of useful information regarding the molecular characterization of pancreatic tumors. However, a huge amount of data needs to be properly managed to determine the information that is useful and correct. A deeper knowledge of the molecular alterations characterizing pancreatic neoplasms may lead to new potential therapeutic targets for these tumors.



INTRODUCTION

Genetic alterations in pancreatic tumors are usually classified in: (1) Tumors with activation of oncogenes (e.g., KRAS mutation—more than 90% of pancreatic tumors); (2) Tumors harboring inactivation of tumor suppressor genes (e.g., p16/CDKN2A, TP53, and SMAD4); and (3) Tumors with inactivation of genes controlling the repair of DNA damage (e.g., hMLH1 and MSH2)[1].

KRAS gene mutations inhibit the ability of KRAS protein to hydrolyze GTP (guanosine-5'-triphosphate), leaving the protein constitutively active, mediating cell survival and differentiation. KRAS is the most common marker used for single-gene testing. Its use is strongly limited by the identification of mutations also in low-grade pancreaticobiliary dysplasia or chronic pancreatitis (about 10%)[2-5]. To date, the “The Papanicolaou Society of Cytopathology guidelines” does not encourage testing of KRAS in bile duct strictures and solid pancreatic masses as a useful “single-gene” test.

P53 protein is involved in cell-cycle regulation, the apoptotic process, and plays a crucial role in the maintenance of genomic stability. Mutations in the TP53 gene lead to inactivation of the normal protein function. In the presence of DNA damage, the functional loss of the p53 protein enhances cellular survival, facilitating the accumulation of further genetic mutations[6]. TP53 is mostly inactivated by single-point mutations[7]. TP53 gene inactivation is a very common event in pancreatic cancer (50% to 75% of pancreatic cancers harbor TP53 mutations)[7-9].

CDKN2A/p16 maps on chromosome 9p and encodes the protein p14ARF, an activator of p53 protein, and p16INK4a protein. This protein inhibits the progression of the cell cycle at the G1-S checkpoint binding of cyclin-dependent kinases (CDKs), as CDK4 and CDK6[10]. CDKN2A/p16 was the first tumor suppressor gene that was shown to undergo silencing and promoter hypermethylation in pancreatic cancer[11]. The Rb/p16 pathway is down-regulated in most pancreatic cancers, almost all through p16 gene inactivation[11]. Mutations in the CDKN2A gene are associated with an increased risk of cancers. Moreover, CDKN2A alterations are also frequently observed in cancer cell lines.

SMAD4/DPC4 gene is located on chromosome 18q and the protein acts in the signal transduction cascade, involving transforming growth factor β (TGF-β). A loss of SMAD4 protein leads to unregulated cellular proliferation[12].

The aim of this review is to summarize the main molecular alterations found in pancreatic tumors, both in solid (e.g., pancreatic ductal adenocarcinoma (PDAC), Table 1) and in cystic neoplasms (e.g., intraductal papillary mucinous neoplasms, IPMN).

Table 1 Main genetic alterations detectable in pancreatic ductal adenocarcinoma.
Type of pancreatic lesion
Genetic alteration
Reported frequency (%)
Type of alterations
Role in clinical practice
PDACKRAS70-90Point mutationsDiagnostic/prognostic
TP5350-75Point mutations/LOHPrognostic
CDKN2A/p1690-98Point mutations/LOHPrognostic/genetic surveillance
SMAD440-60Point mutations/LOHPrognostic
BRCA1/25-10Point mutationsPredictive/genetic surveillance
NTRK1-3< 1Gene fusionsPredictive
MSI< 2LOFPredictive
PDAC
Wide genome analysis

According to data obtained by whole-exome sequencing analysis, PDAC harbors an average of about 60 genetic alterations, and most of them are point mutations[1,8,13]. Based on these alterations, 12 cellular pathways genetically altered in pancreatic neoplasia have been identified[8]. Massive sequencing studies carried out on PDACs revealed that alterations may be found in some well-known genes (e.g., KRAS, CDKN2A, TP53, ARID1A, SMAD4) or in novel genes, that may be involved in DNA damage repair (e.g., ATM), chromatin modification (e.g., EPC1 and ARID2), or in neoplastic carcinogenesis (e.g., KDM6A and PREX2)[14]. Whole-exome sequencing analysis has defined some putative therapeutic targets (e.g., RBM10) associated with longer survival in patients with pancreatic cancers, others associated with improved survival (e.g., KRAS p.Q61H mutation), and others defining sensitivity to target therapies in PDAC models (e.g., BRAF mutations as sensitivity markers for treatment with vemurafenib)[15].

An expression analysis study led to the cluster of PDAC in 4 different subtypes: (1) Squamous; (2) Pancreatic progenitor; (3) Immunogenic; and (4) Aberrantly differentiated endocrine exocrine[16]. Also analysis of the structural genomic alterations in PDACs have been classified into four different subtypes: (1) “Stable”, when PDACs contain less than 50 structural variations; (2) “Locally rearranged”, when PDACs exhibit a focal event on one or two chromosomes; (3) “Scattered subtype”, when PDACs show a fewer number of chromosomal damages and less than 200 structural variations; and (4) “Unstable”, when PDACs exhibit more than 200 structural events[17].

KRAS

KRAS is the most frequently mutated oncogene in pancreatic cancers (> 95%) and the most frequent gene mutated in PDAC (from 70% to 95%)[8,15,18,19]. The acquisition of a KRAS mutation represents an early and initiating event in PDACs. However, the low frequency of progression of precursor lesions to PDAC suggests that additional alterations are needed for neoplastic progression[20]. In PDAC, the mutations in KRAS are not located only in exon 2, but they have also been found in other exons[14,15,21]. However, the mutations harbored in exon 2 exhibited a similar association with survival, while cases mutated in exon 3 seem to have a remarkably favorable prognosis[15]. Coexistent KRAS mutations were detected in the same pancreatic neoplastic mass more frequently than in other tumors[21-23].

KRAS analysis may be of particular interest in the case of doubtful or inconclusive diagnoses (e.g., specimens with cytological atypia or acellular specimens). It is well-established that cytopathology together with KRAS analysis allows improved diagnosis of PDAC in endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA) specimens[24-31]. Finding a KRAS mutation in EUS-FNA material may: (1) Indicate that a re-evaluation of the cytopathology report is needed (mainly if doubtful); (2) Indicate a second FNA or surgery[21]; and (3) Lead to a reduction in false-negative diagnoses[32].

A worse prognosis has been observed in patients with tumors harboring coexistent KRAS mutations together with TP53 alterations and/or loss of SMAD4 protein[33,34]. KRAS p.G12C variant is a mutation that could be targeted by KRAS-G12C-specific inhibitors[35]. PDAC which also harbors the p.G12C alteration may benefit from this treatment. KRAS-wild type PDACs represent a distinct molecular subtype of pancreatic cancer that could benefit from tailored treatments, including BRAF antagonists and MAPK inhibitors[36].

TP53

EUS-FNA sensitivity in the diagnosis of pancreatic malignant lesions can be improved by also implementing the evaluation of the TP53 gene[37-39]. P53 protein overexpression has been detected in specimens with pancreatic cancer but not in those with chronic pancreatitis. TP53 alterations have been detected in 50%-75% of PDACs. Combining the evaluation of p53 protein expression with histological examination improves the sensitivity of diagnosis of pancreatic cancers, with a high specificity [37,38]. The sensitivity of EUS-FNA in the diagnosis of PDAC is further improved by combining p53 and Ki67 staining[38].

The combination of p53 and CA19.9 increases the sensitivity of cytology but it can negatively affect the specificity[39]. A worse patient prognosis has been correlated with loss of p53 protein, mainly if p53 loss is combined with KRAS alterations and loss of SMAD4 protein expression[33,34]. In a recent study based on next-generation sequencing (NGS) results from the CONKO-001 phase III trial, it has been demonstrated that patients with TP53 mutated PDAC tumors benefit from adjuvant gemcitabine treatment[40].

SMAD4

Approximately 50% of pancreatic cancers show SMAD4 protein inactivation, due to homozygous deletion and intragenic mutations[10,41-43]. Loss of SMAD4 generally occurs late in pancreatic carcinogenesis, and it has been frequently observed in pancreatic adenocarcinomas, but not in extra-pancreatic lesions[44]. In reactive and inflammatory diseases of the pancreas (e.g., chronic pancreatitis) SMAD4 activity is usually preserved. Loss of SMAD4 protein has also been linked with an increased risk of developing metastases and worse prognosis[33,34,43,45,46]. In PDAC, SMAD4 mutations lead to the prevention of the normal transduction of TGF-β signals; TGF-β inhibitor has shown efficacy in a preclinical investigation[47].

A study by Hsieh et al[48] demonstrated that SMAD4 deficiency in PDAC cells led to higher sensitivity to gemcitabine. On the contrary, SMAD4 mutated cells were sensitive to gemcitabine similar to that in cells with wild-type SMAD4[48]. These data suggest that the SMAD4 copy number may be a therapeutic marker for PDAC treatment with gemcitabine[49]. In this study, it was observed that SMAD4 deficiency led to upregulation of cell cycle-related genes, such as CDK1, with consequent higher sensitivity to other agents modulating the cell cycle such as clofarabine, cytarabine, darinaparsin, and olaparib[48,49].

CDKN2A

More than 95% of sporadic pancreatic carcinomas harbor a CDKN2A gene inactivation due to intragenic mutation, usually coupled with the loss of the other allele, promoter hypermethylation, or homozygous deletion of both alleles[50-52]. CDKN2A is involved in familial pancreatic cancer, although CDKN2A germline mutations in patients with pancreatic cancer occur rarely (0.6%)[53,54]. Patients harboring CDKN2A mutations are more likely to report a family history of pancreatic cancer than those without CDKN2A alterations[54]. Surveillance protocols from age 40 are recommended for CDKN2A mutation carriers[55].

Decreased expression of p16 protein has been associated with a tendency for the tumor to be larger than in those with normal p16 expression levels. Pancreatic neoplasms with loss of p16 expression, due to mutations and/or promoter hypermethylation, are significantly larger. Moreover, the survival period in these patients was significantly shorter when compared to those with a pancreatic tumor characterized by intact p16 functions[33,56,57]. CDK4/6 is a potential target in CDKN2A-deficient tumors and the efficacy of CDK4 inhibitors has been confirmed in PDAC preclinical models[58].

BRCA1-2

Alterations in the BRCA pathway and defects in DNA maintenance (such as genomic instability and the BRCA mutational signature) may have implications for the therapeutic selection of patients with pancreatic tumors[17]. BRCA1/2 mutation frequencies range from 5% to 10%, and BRCA alterations have been detected both in sporadic and familial PDAC[1,59]. Patients with PDAC harboring germline BRCA1 or BRCA2 mutations showed a longer progression-free survival if treated with a PARP inhibitor[60,61]. According to the recommendation from the International Cancer of the Pancreas Screening Consortium, for BRCA1 a consensus of 69.9% was reached for recommending that BRCA1 mutation carriers undergo surveillance, whereas no consensus was reached on family history criteria for BRCA1 mutation carriers[55]. For carriers of BRCA2 mutations, the consensus (agreement of 93%) was to recommend surveillance for mutation carriers who have a blood relative with pancreatic cancer[55]. For BRCA2 mutation carriers with a germline variant (deleterious), the recommended age to initiate surveillance is generally 50 years[55]. With regard to the surveillance protocols for high-risk individuals, the consensus is that pancreatic imaging with magnetic resonance imaging (MRI)/magnetic retrograde cholangiopancreatography and/or EUS should be the first-line test for pancreatic surveillance[55]. Pancreatic computed tomography is reserved for individuals unable to undergo MRI or EUS[55].

Other alterations

Even if uncommon, MSI/dMMR (Microsatellite Instability/defective DNA mismatch repair) has been described in about 1%-2% of PDAC[62,63]. PDACs with MSI are usually associated with medullary histology and are rarely mutated in KRAS or TP53 genes[62,64]. National Comprehensive Cancer Network (NCCN) guidelines recommend the MSI and MMR tests in locally advanced and metastatic pancreatic carcinomas[65]. NCCN recommends the treatment of PDAC with the PD-1 (programmed cell death-1) inhibitor, Pembrolizumab, only in those patients with MSI-H (high-MSI) or dMMR advanced/metastatic pancreatic cancer[65].

BRAF mutations are an uncommon event in PDAC[1]. Some evidence suggests that patients with pancreas tumors harboring BRAFV600E mutations may benefit from treatment with RAF-MEK-targeted therapy[66].

The MGMT promoter can be hypermethylated in PDAC[34,67]. In 1998, treatment with temozolomide in advanced pancreatic cancer has been tried in a phase II study, but no relevant clinical response was observed[68]. ARID1A oncosuppressor protein deficiency was significantly associated with poor outcome in PDAC patients[15]. Amplifications and copy-number gains of oncogenes such as ERBB2, MET, and FGFR1 may be detected in pancreatic tumors[17]. The inactivation of several genes (e.g., ROBO1, ROBO2, SLIT2, and RNF43) leads to an aberrant WNT (Wingless-related integration site) signaling[15,17]. Unresectable non-metastatic pancreatic carcinomas may also harbor mutations in GRM8 and TRIM33 genes[69], while only a small fraction (approximately 5%) of pancreatic adenocarcinomas showed Cyclin E overexpression[70]. Different to solid pseudopapillary neoplasia (SPN), CTNNB1 mutations are uncommon in PDAC[15,71]. In PDAC, the frequency of NTRK (neurotrophic tropomyosin receptor kinase) fusion is very low (about 0.3%), but clinical trials revealed that the selective TRK (tropomyosin receptor kinase) inhibitors are also effective in PDAC harboring NTRK rearrangement[58]. NCCN guidelines suggest the use of a TRK inhibitor in NTRK gene fusion-positive advanced/metastatic pancreatic cancer[61,65].

Organoids are a preclinical model that is becoming increasingly important for studying tumor behavior because they can simulate metastases, microtumors, and the tumor microenvironment better than “classical” monolayer culture systems[72]. An interesting preclinical model for the study of PDAC is the set-up of three-dimensional (3D)-tumoroids in vitro culture systems[72-78]. These organoids can also be generated from a resected PDAC and are amenable to therapeutic screening as well as genetic and biochemical perturbation[77]. In a study by Boj et al[76], organoids derived from murine and human PDAC generated lesions similar to pancreatic intraepithelial neoplasia (PanIN) and progressed to invasive PDAC. Intriguingly, the expression of mutated KRAS protein (KRASG12D) in PDAC organoids was sufficient to induce a preinvasive neoplasm[76]. PDAC tumoroid cultures retain the capacity to maintain tumor stroma and characteristics of the primary tumor including the long-term (> 44 d) production of CEA (carcinoembryonic antigen) and CA19-9 (carbohydrate antigen 19-9)[74]. This 3D cell culture model of PDAC would help the diagnostics, investigation of genetic drivers, and identification of novel therapeutic targets[72]. Moreover, this culture could also allow clarification on how the immunosuppressive mechanism affects the growth and stasis of tumors[74]. Moreover, another important aspect is that organoids are suitable for storage in biobanks and used for further research, ensuring access to relevant sample numbers[78].

CYSTIC PANCREATIC TUMORS
IPMN

KRAS mutations are harbored by over 90% of low-grade PanIN[79], and mutant KRAS is sufficient to initiate the development of PanINs and IPMNs[20,80-83] (Table 2).

Table 2 Main genetic alterations detectable in cystic pancreatic lesions.
Type of pancreatic lesion
Genetic alteration
Reported frequency (%)
Type of alterations
Role in clinical practice
IPMNKRAS90Point mutationsDiagnostic
TP5310Point mutations/LOHPrognostic
GNAS40-60Point mutationsDiagnostic
RNF4325Point mutations/LOHDiagnostic
MCNKRAS0-25% (LG); 50-90% (HG)Point mutationsDiagnostic
CDKN2A/p160-10 (LG); 50 (HG)Point mutations/LOHPrognostic
TP530 (LG); 20-50 (HG)Point mutations/LOHPrognostic
SCNVHL40-60Point mutations/LOHDiagnostic

The distinction between IPMNs and mucinous cystic neoplasms (MCNs) from non-neoplastic pancreatic cysts may be helped by analysis of pancreatic cyst fluid. In fact, a KRAS mutation is highly specific for mucinous differentiation, but not for identifying MCNs[84]. KRAS alterations have a very high specificity but low sensitivity for MCNs and IPMNs (approximately 15% and 70%, respectively). If KRAS analysis is combined with that of GNAS, the sensitivity increases[85,86]. The differential diagnosis of cystic mucinous lesions (IPMN and MCN), mainly when the pre-operative cytology is non-diagnostic or when the CEA cyst fluid levels are indeterminate, may be helped by the analysis of KRAS and Loss-of-Heterozygosity (LOH)[87].

IPMNs rarely harbor mutations in the TP53 gene (approximately 10%). The overexpression of TP53 was more commonly observed in IPMNs of the pancreatobiliary type with invasion[88]. In a cohort of IPMN patients, the overexpression of TP53, together with loss of function of SMAD4, was strongly associated with patient survival[88]. In IPMNs, SMAD4 loss of function was rarely detected; SMAD4 loss is more common in IPMNs of the pancreatobiliary type with invasion[88].

The GNAS gene encodes the α-subunit of the stimulatory G-protein (Gαs). This subunit regulates the adenylate cyclase activity through Gαs-coupled receptors. Alterations in GNAS may determine the characteristic IPMN phenotype[89]. GNAS activating mutations are reported prevalently in IPMN (approximately 40%-60%)[85,88,90-92] and invasive pancreatic cancers, only if arising in association with an IPMN[79,93]. In the majority of IPMNs (approximately 90%), at least one of the KRAS or GNAS genes harbor mutations[94], and in about half of IPMN (approximately 40%), a GNAS alteration coexists with a KRAS mutation[90]. The combination of KRAS and GNAS mutations helps in distinguishing between a serous cystic neoplasm (SCN) and an IPMN with high sensitivity and specificity. In fact, if most IPMNs have a GNAS and/or a KRAS alteration, no SCNs harbor either mutation. Besides, detecting a mutation in the GNAS gene in cyst fluid may help to distinguish IPMNs from MCNs[95].

RNF43 mutation frequency in IPMN is about 25%, ranging from 10% to 75%[93,96]. These mutations are often inactivating alterations and are found in association with LOH.

CDKN2A/p16 inactivating mutations have also been found in IPMNs with high-grade dysplasia, other than in pancreatic adenocarcinoma[97].

Germline mutations of STK11/LKB1 genes have been associated with IPMNs and invasive pancreatic cancer[98,99]. Besides, somatic mutations of STK11/LKB1 are observed in about 5% of patients with sporadic IPMNs and pancreatic cancers[98,99].

A high amount of DNA and high-amplitude mutations in the pancreatic cyst fluid may be indicators of malignancy, helping to identifying malignant cystic lesions[86].

The use of NGS in pancreatic cyst fluid allows high sensitivity and specificity in classifying pancreatic cancers, mainly for the diagnosis of IPMN with advanced neoplasia[100].

MCN

MCNs harbor alterations also commonly found in PDAC. MCNs frequently have KRAS gene mutations, mainly in MCNs with high-grade dysplasia[101]. The diagnosis of mucinous cysts may be helped by the presence of a KRAS mutation in cyst fluid[86]. p16/CDKN2A expression is altered in MCN, even if in a lower percentage (about 15%) if compared to that of PDAC[102]. P53 has been reported with aberrant expression in MCN with high-grade dysplasia[103]. TP53 alterations are often associated with aggressiveness and seem to be involved in progression to PDAC[92]. Mutations of RNF43 have been reported in MCN[92]. PIK3CA alterations were described with very low frequency in MCN and in association with high-grade dysplasia and invasive adenocarcinoma[104]. As in IPMN, no VHL mutations were detected in MCN[92], but, different to IPMN, the GNAS gene is not mutated in MCN[96]. The LOH of Dpc4/Smad4 contributes to MCN progression in mice with KRAS-G12D mutation[105], confirming that the SMAD4 gene acts as a PDAC tumor suppressor[106]. Whole-exome sequencing performed on a cohort of MCN revealed that the tumors harbored about 16 somatic mutations per tumor, lower than the number of mutations observed in IPMN (about 27 per tumor)[92].

SCN

VHL mutations are frequently reported in SCN (from 40% up to 60% of cases) [100,107]. SCNs usually do not harbor alterations in genes frequently mutated in IPMN, or MCN (i.e., KRAS, GNAS, TP53), helping to distinguish SCNs from the other mucinous neoplasia[100,108]. The lack of CTNNB1 mutations allows the differentiation of SCN from SPN. Moreover, SCNs do not harbor mutations in genes frequently altered in neuroendocrine pancreatic tumors[109]. Whole exome sequencing analysis performed on a cohort of eight SCNs revealed that almost all tumors harbored a LOH on chromosome 3p[92]. An average of only 10 non-synonymous somatic mutations was detected in SCNs[92], far less than the average observed in PDAC.

OTHER PANCREATIC TUMORS
SPN

The CTNNB1 gene (codons from 32 to 37) encodes for a region that plays a crucial role in the regulation of the β-catenin protein[110,111]. Alterations within this CTNNB1 region usually block β-catenin phosphorylation, inhibiting degradation of the protein[112]. CTNNB1 mutations are characteristic of pancreatic SPN[71,113,114]. Different to PDAC, SPNs are not mutated in KRAS, TP53, or SMAD4 genes, and CTNNB1 mutations are the main molecular alteration detected[92]. DNA array CGH (comparative genomic hybridization) performed on a pediatric case of SPN revealed a loss in chromosome band 11p15.5, a chromosomal region encoding for the HRAS gene[115]. As suggested by Selenica et al[116], even if inhibition of the Wnt pathway may be an intuitive therapeutic option for this disease, the evidence that clinically advanced Wnt pathway inhibitors target components upstreaming β-catenin activity is a clear limitation. For this reason, future drugs should be designed to target the β-catenin protein directly[116] (Table 3).

Table 3 Main genetic alterations detectable in other pancreatic tumors.
Type of pancreatic lesion
Genetic alteration
Reported frequency (%)
Type of alterations
Role in clinical practice
SPNCTNNB190-100Point mutationsDiagnostic
PanNETMEN170Point mutations/LOHDiagnostic/prognostic
VHL25Point mutations/LOHDiagnostic
AACCTNNB15-25Point mutationsLOFDiagnostic
MSI5-15LOFPredictive?
Pancreatic neuroendocrine tumors

Pancreatic neuroendocrine tumors (PanNETs) show a distinct landscape of molecular alterations if compared to the other pancreatic tumors. The mutation frequency in PanNET was lower than that observed in poorly differentiated neuroendocrine carcinomas (45% and 83%, respectively)[117], and the incidence of mutations was higher in PanNET with a high risk of progression than those with low risk[117]. MEN1 alterations (both mutations and LOH) have been found in up to 70% of PanNETs and have been associated with a better prognosis[118,119]. PanNETs harbor alterations in those genes involved in the chromatin remodeling complex, such as loss of ATRX and DAXX proteins expression[118]. Mutations in the mTOR (mammalian target of rapamycin) pathway genes (e.g., PIK3CA, PTEN, and TSC2) have been detected in PanNET[118]. For example, PTEN and TSC2 genes have been observed as inactivating in primary PanNET and their low protein levels were associated with shorter overall and disease-free survival[120]. VHL inactivation, due to deletion or methylation, was also observed in up to 25% of PanNETs[121,122].

Acinar cell carcinomas

Activating mutations in CTNNB1 and inactivating mutations in APC genes have been observed in up to 25% of acinar cell carcinomas (ACCs). KRAS mutations are an uncommon event in this type of neoplasia[123]. Nevertheless, TP53 mutations were also found only in a low fraction of ACCs[123-125], deletion of the TP53 region (chromosome band 17p13.1) was detected by FISH in about half of a cohort of 54 ACCs[125]. Alterations found in IPMN (e.g., GNAS and RNF43) or in PanNET (e.g., MEN1) were rarely found in ACC[126] A genome-wide analysis performed on ACCs revealed a median number of 137 point mutations per tumor and COL12A1, FRY, FRYL, and PLB1 were the most frequently mutated genes[124]. Intriguingly, a genome-wide analysis identified no recurrent point mutations in ACCs[124]. Microsatellite instability was detected in a fraction of ACC ranging from 7% to 14% of cases[127]. ACC with MSI did not exhibit distinct morphological or clinical features[128].

CONCLUSION

To date, there are no targetable molecules available for personalized patient treatment of pancreatic tumors in clinical practice, as stated by the current European Society for Medical Oncology (ESMO) guidelines[129]. EUS-FNA has improved pre-operative diagnosis[130-132], but the management of patients with a pancreatic lesion is still challenging. In fact, in a subset of cases, such as lesions with atypical/suspicious cytopathologic features, the pre-operative diagnosis remains inconclusive[133]. Performing a molecular characterization could help mainly in solving these “inconclusive” specimens. The introduction of multi-gene analysis approaches, such as NGS, has provided a lot of useful information regarding the molecular characterization of pancreatic tumors[134]. EUS-FNAC (FNA cytology) is a useful diagnostic tool for pancreatic lesions[135] and molecular analysis can be successfully performed on cytological smears[21]. However, it has becoming increasingly crucial to have sufficient material for histological, immunohistochemical, and molecular characterization. Pancreatic FNAB (FNA biopsy) provides enough material to allow proper histological assessment, immunostaining, and molecular techniques [e.g., NGS, digital polymerase chain reaction (PCR)][136-138].

A huge amount of data needs to be properly managed to determine the information that is useful and correct[139]. The recent ESMO guidelines have outlined their indications for the use of NGS in the characterization of metastatic cancers. As regards PDAC, it is not currently recommended to perform multigene NGS in daily practice[140]. However, ESMO encourages multigene sequencing in order to get access to innovative drugs. Moreover, NGS can be an alternative technique to PCR-based assays if it is not associated with extra cost for the public health care system if the patient is informed about the putative benefits of this analysis[140]. In conclusion, a deeper knowledge of the molecular alterations characterizing pancreatic neoplasms may lead to new potential therapeutic targets for these tumors.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Italy

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): C, C, C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Aykan NF, Chin YK, Labori KJ, Shiryajev YN S-Editor: Gao CC L-Editor: Webster JR P-Editor: Ma YJ

References
1.  Cancer Genome Atlas Research Network. Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma. Cancer Cell 2017; 32: 185-203. e13.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1247]  [Cited by in F6Publishing: 1153]  [Article Influence: 164.7]  [Reference Citation Analysis (0)]
2.  Layfield LJ, Ehya H, Filie AC, Hruban RH, Jhala N, Joseph L, Vielh P, Pitman MB;  Papanicolaou Society of Cytopathology. Utilization of ancillary studies in the cytologic diagnosis of biliary and pancreatic lesions: the Papanicolaou Society of Cytopathology guidelines for pancreatobiliary cytology. Diagn Cytopathol. 2014;42:351-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 71]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
3.  Lüttges J, Reinecke-Lüthge A, Möllmann B, Menke MA, Clemens A, Klimpfinger M, Sipos B, Klöppel G. Duct changes and K-ras mutations in the disease-free pancreas: analysis of type, age relation and spatial distribution. Virchows Arch. 1999;435:461-468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 96]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
4.  Löhr M, Klöppel G, Maisonneuve P, Lowenfels AB, Lüttges J. Frequency of K-ras mutations in pancreatic intraductal neoplasias associated with pancreatic ductal adenocarcinoma and chronic pancreatitis: a meta-analysis. Neoplasia. 2005;7:17-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 224]  [Cited by in F6Publishing: 226]  [Article Influence: 11.9]  [Reference Citation Analysis (0)]
5.  Löhr M, Maisonneuve P, Lowenfels AB. K-Ras mutations and benign pancreatic disease. Int J Pancreatol. 2000;27:93-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 47]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
6.  Vogelstein B, Kinzler KW. Cancer genes and the pathways they control. Nat Med. 2004;10:789-799.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2905]  [Cited by in F6Publishing: 2685]  [Article Influence: 134.3]  [Reference Citation Analysis (0)]
7.  Forbes SA, Beare D, Gunasekaran P, Leung K, Bindal N, Boutselakis H, Ding M, Bamford S, Cole C, Ward S, Kok CY, Jia M, De T, Teague JW, Stratton MR, McDermott U, Campbell PJ. COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res. 2015;43:D805-D811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1815]  [Cited by in F6Publishing: 1787]  [Article Influence: 178.7]  [Reference Citation Analysis (0)]
8.  Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, Mankoo P, Carter H, Kamiyama H, Jimeno A, Hong SM, Fu B, Lin MT, Calhoun ES, Kamiyama M, Walter K, Nikolskaya T, Nikolsky Y, Hartigan J, Smith DR, Hidalgo M, Leach SD, Klein AP, Jaffee EM, Goggins M, Maitra A, Iacobuzio-Donahue C, Eshleman JR, Kern SE, Hruban RH, Karchin R, Papadopoulos N, Parmigiani G, Vogelstein B, Velculescu VE, Kinzler KW. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801-1806.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3082]  [Cited by in F6Publishing: 2881]  [Article Influence: 180.1]  [Reference Citation Analysis (0)]
9.  Scarpa A, Capelli P, Mukai K, Zamboni G, Oda T, Iacono C, Hirohashi S. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am J Pathol. 1993;142:1534-1543.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Maitra A, Hruban RH. Pancreatic cancer. Annu Rev Pathol. 2008;3:157-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 537]  [Cited by in F6Publishing: 559]  [Article Influence: 34.9]  [Reference Citation Analysis (0)]
11.  Schutte M, Hruban RH, Geradts J, Maynard R, Hilgers W, Rabindran SK, Moskaluk CA, Hahn SA, Schwarte-Waldhoff I, Schmiegel W, Baylin SB, Kern SE, Herman JG. Abrogation of the Rb/p16 tumor-suppressive pathway in virtually all pancreatic carcinomas. Cancer Res. 1997;57:3126-3130.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Siegel PM, Massagué J. Cytostatic and apoptotic actions of TGF-beta in homeostasis and cancer. Nat Rev Cancer. 2003;3:807-821.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1240]  [Cited by in F6Publishing: 1212]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
13.  Wang L, Tsutsumi S, Kawaguchi T, Nagasaki K, Tatsuno K, Yamamoto S, Sang F, Sonoda K, Sugawara M, Saiura A, Hirono S, Yamaue H, Miki Y, Isomura M, Totoki Y, Nagae G, Isagawa T, Ueda H, Murayama-Hosokawa S, Shibata T, Sakamoto H, Kanai Y, Kaneda A, Noda T, Aburatani H. Whole-exome sequencing of human pancreatic cancers and characterization of genomic instability caused by MLH1 haploinsufficiency and complete deficiency. Genome Res. 2012;22:208-219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 93]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
14.  Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N;  Australian Pancreatic Cancer Genome Initiative; Kakkar N, Zhao F, Wu YQ, Wang M, Muzny DM, Fisher WE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, Chang K, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, Sam M, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Capelli P, Corbo V, Scardoni M, Tortora G, Tempero MA, Mann KM, Jenkins NA, Perez-Mancera PA, Adams DJ, Largaespada DA, Wessels LF, Rust AG, Stein LD, Tuveson DA, Copeland NG, Musgrove EA, Scarpa A, Eshleman JR, Hudson TJ, Sutherland RL, Wheeler DA, Pearson JV, McPherson JD, Gibbs RA, Grimmond SM. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature. 2012;491:399-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1442]  [Cited by in F6Publishing: 1521]  [Article Influence: 126.8]  [Reference Citation Analysis (0)]
15.  Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, Choti MA, Yeo CJ, McCue P, White MA, Knudsen ES. Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 2015;6:6744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 704]  [Cited by in F6Publishing: 763]  [Article Influence: 84.8]  [Reference Citation Analysis (0)]
16.  Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, Miller DK, Christ AN, Bruxner TJ, Quinn MC, Nourse C, Murtaugh LC, Harliwong I, Idrisoglu S, Manning S, Nourbakhsh E, Wani S, Fink L, Holmes O, Chin V, Anderson MJ, Kazakoff S, Leonard C, Newell F, Waddell N, Wood S, Xu Q, Wilson PJ, Cloonan N, Kassahn KS, Taylor D, Quek K, Robertson A, Pantano L, Mincarelli L, Sanchez LN, Evers L, Wu J, Pinese M, Cowley MJ, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chantrill LA, Mawson A, Humphris J, Chou A, Pajic M, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Lovell JA, Merrett ND, Toon CW, Epari K, Nguyen NQ, Barbour A, Zeps N, Moran-Jones K, Jamieson NB, Graham JS, Duthie F, Oien K, Hair J, Grützmann R, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Rusev B, Capelli P, Salvia R, Tortora G, Mukhopadhyay D, Petersen GM;  Australian Pancreatic Cancer Genome Initiative; Munzy DM, Fisher WE, Karim SA, Eshleman JR, Hruban RH, Pilarsky C, Morton JP, Sansom OJ, Scarpa A, Musgrove EA, Bailey UM, Hofmann O, Sutherland RL, Wheeler DA, Gill AJ, Gibbs RA, Pearson JV, Biankin AV, Grimmond SM. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2480]  [Cited by in F6Publishing: 2210]  [Article Influence: 276.3]  [Reference Citation Analysis (0)]
17.  Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, Quinn MC, Robertson AJ, Fadlullah MZ, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wani S, Wilson PJ, Markham E, Cloonan N, Anderson MJ, Fink JL, Holmes O, Kazakoff SH, Leonard C, Newell F, Poudel B, Song S, Taylor D, Waddell N, Wood S, Xu Q, Wu J, Pinese M, Cowley MJ, Lee HC, Jones MD, Nagrial AM, Humphris J, Chantrill LA, Chin V, Steinmann AM, Mawson A, Humphrey ES, Colvin EK, Chou A, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Pettitt JA, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, Graham JS, Niclou SP, Bjerkvig R, Grützmann R, Aust D, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Falconi M, Zamboni G, Tortora G, Tempero MA;  Australian Pancreatic Cancer Genome Initiative; Gill AJ, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Pearson JV, Biankin AV, Grimmond SM. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature. 2015;518:495-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1917]  [Cited by in F6Publishing: 1771]  [Article Influence: 196.8]  [Reference Citation Analysis (1)]
18.  Hruban RH, van Mansfeld AD, Offerhaus GJ, van Weering DH, Allison DC, Goodman SN, Kensler TW, Bose KK, Cameron JL, Bos JL. K-ras oncogene activation in adenocarcinoma of the human pancreas. A study of 82 carcinomas using a combination of mutant-enriched polymerase chain reaction analysis and allele-specific oligonucleotide hybridization. Am J Pathol. 1993;143:545-554.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Aung KL, Fischer SE, Denroche RE, Jang GH, Dodd A, Creighton S, Southwood B, Liang SB, Chadwick D, Zhang A, O'Kane GM, Albaba H, Moura S, Grant RC, Miller JK, Mbabaali F, Pasternack D, Lungu IM, Bartlett JMS, Ghai S, Lemire M, Holter S, Connor AA, Moffitt RA, Yeh JJ, Timms L, Krzyzanowski PM, Dhani N, Hedley D, Notta F, Wilson JM, Moore MJ, Gallinger S, Knox JJ. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin Cancer Res. 2018;24:1344-1354.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 333]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
20.  Morris JP 4th, Wang SC, Hebrok M. KRAS, Hedgehog, Wnt and the twisted developmental biology of pancreatic ductal adenocarcinoma. Nat Rev Cancer. 2010;10:683-695.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 425]  [Cited by in F6Publishing: 435]  [Article Influence: 31.1]  [Reference Citation Analysis (0)]
21.  de Biase D, Visani M, Baccarini P, Polifemo AM, Maimone A, Fornelli A, Giuliani A, Zanini N, Fabbri C, Pession A, Tallini G. Next generation sequencing improves the accuracy of KRAS mutation analysis in endoscopic ultrasound fine needle aspiration pancreatic lesions. PLoS One. 2014;9:e87651.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 59]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
22.  Gormally E, Caboux E, Vineis P, Hainaut P. Circulating free DNA in plasma or serum as biomarker of carcinogenesis: practical aspects and biological significance. Mutat Res. 2007;635:105-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 323]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
23.  Visani M, de Biase D, Baccarini P, Fabbri C, Polifemo AM, Zanini N, Pession A, Tallini G. Multiple KRAS mutations in pancreatic adenocarcinoma: molecular features of neoplastic clones indicate the selection of divergent populations of tumor cells. Int J Surg Pathol. 2013;21:546-552.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 18]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
24.  Bournet B, Souque A, Senesse P, Assenat E, Barthet M, Lesavre N, Aubert A, O'Toole D, Hammel P, Levy P, Ruszniewski P, Bouisson M, Escourrou J, Cordelier P, Buscail L. Endoscopic ultrasound-guided fine-needle aspiration biopsy coupled with KRAS mutation assay to distinguish pancreatic cancer from pseudotumoral chronic pancreatitis. Endoscopy. 2009;41:552-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 60]  [Article Influence: 4.0]  [Reference Citation Analysis (0)]
25.  Bournet B, Selves J, Grand D, Danjoux M, Hanoun N, Cordelier P, Buscail L. Endoscopic ultrasound-guided fine-needle aspiration biopsy coupled with a KRAS mutation assay using allelic discrimination improves the diagnosis of pancreatic cancer. J Clin Gastroenterol. 2015;49:50-56.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 42]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
26.  Ginestà MM, Mora J, Mayor R, Farré A, Peinado MA, Busquets J, Serrano T, Capellá G, Fabregat J. Genetic and epigenetic markers in the evaluation of pancreatic masses. J Clin Pathol. 2013;66:192-197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
27.  Maluf-Filho F, Kumar A, Gerhardt R, Kubrusly M, Sakai P, Hondo F, Matuguma SE, Artifon E, Monteiro da Cunha JE, César Machado MC, Ishioka S, Forero E. Kras mutation analysis of fine needle aspirate under EUS guidance facilitates risk stratification of patients with pancreatic mass. J Clin Gastroenterol. 2007;41:906-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 51]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
28.  Ogura T, Yamao K, Sawaki A, Mizuno N, Hara K, Hijioka S, Niwa Y, Tajika M, Kondo S, Shimizu Y, Bhatia V, Higuchi K, Hosoda W, Yatabe Y. Clinical impact of K-ras mutation analysis in EUS-guided FNA specimens from pancreatic masses. Gastrointest Endosc. 2012;75:769-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 50]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
29.  Pellisé M, Castells A, Ginès A, Solé M, Mora J, Castellví-Bel S, Rodríguez-Moranta F, Fernàndez-Esparrach G, Llach J, Bordas JM, Navarro S, Piqué JM. Clinical usefulness of KRAS mutational analysis in the diagnosis of pancreatic adenocarcinoma by means of endosonography-guided fine-needle aspiration biopsy. Aliment Pharmacol Ther. 2003;17:1299-1307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 56]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
30.  Tada M, Komatsu Y, Kawabe T, Sasahira N, Isayama H, Toda N, Shiratori Y, Omata M. Quantitative analysis of K-ras gene mutation in pancreatic tissue obtained by endoscopic ultrasonography-guided fine needle aspiration: clinical utility for diagnosis of pancreatic tumor. Am J Gastroenterol. 2002;97:2263-2270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 111]  [Cited by in F6Publishing: 121]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
31.  Takahashi K, Yamao K, Okubo K, Sawaki A, Mizuno N, Ashida R, Koshikawa T, Ueyama Y, Kasugai K, Hase S, Kakumu S. Differential diagnosis of pancreatic cancer and focal pancreatitis by using EUS-guided FNA. Gastrointest Endosc. 2005;61:76-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 100]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
32.  Bournet B, Gayral M, Torrisani J, Selves J, Cordelier P, Buscail L. Role of endoscopic ultrasound in the molecular diagnosis of pancreatic cancer. World J Gastroenterol. 2014;20:10758-10768.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 29]  [Cited by in F6Publishing: 26]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
33.  Oshima M, Okano K, Muraki S, Haba R, Maeba T, Suzuki Y, Yachida S. Immunohistochemically detected expression of 3 major genes (CDKN2A/p16, TP53, and SMAD4/DPC4) strongly predicts survival in patients with resectable pancreatic cancer. Ann Surg. 2013;258:336-346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 174]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
34.  Masetti M, Acquaviva G, Visani M, Tallini G, Fornelli A, Ragazzi M, Vasuri F, Grifoni D, Di Giacomo S, Fiorino S, Lombardi R, Tuminati D, Ravaioli M, Fabbri C, Bacchi-Reggiani ML, Pession A, Jovine E, de Biase D. Long-term survivors of pancreatic adenocarcinoma show low rates of genetic alterations in KRAS, TP53 and SMAD4. Cancer Biomark. 2018;21:323-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
35.  Gillson J, Ramaswamy Y, Singh G, Gorfe AA, Pavlakis N, Samra J, Mittal A, Sahni S. Small Molecule KRAS Inhibitors: The Future for Targeted Pancreatic Cancer Therapy? Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
36.  Luchini C, Paolino G, Mattiolo P, Piredda ML, Cavaliere A, Gaule M, Melisi D, Salvia R, Malleo G, Shin JI, Cargnin S, Terrazzino S, Lawlor RT, Milella M, Scarpa A. KRAS wild-type pancreatic ductal adenocarcinoma: molecular pathology and therapeutic opportunities. J Exp Clin Cancer Res. 2020;39:227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 40]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
37.  Itoi T, Takei K, Sofuni A, Itokawa F, Tsuchiya T, Kurihara T, Nakamura K, Moriyasu F, Tsuchida A, Kasuya K. Immunohistochemical analysis of p53 and MIB-1 in tissue specimens obtained from endoscopic ultrasonography-guided fine needle aspiration biopsy for the diagnosis of solid pancreatic masses. Oncol Rep. 2005;13:229-234.  [PubMed]  [DOI]  [Cited in This Article: ]
38.  Jahng AW, Reicher S, Chung D, Varela D, Chhablani R, Dev A, Pham B, Nieto J, Venegas RJ, French SW, Stabile BE, Eysselein VE. Staining for p53 and Ki-67 increases the sensitivity of EUS-FNA to detect pancreatic malignancy. World J Gastrointest Endosc. 2010;2:362-368.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 20]  [Cited by in F6Publishing: 22]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
39.  Mu DQ, Wang GF, Peng SY. p53 protein expression and CA19.9 values in differential cytological diagnosis of pancreatic cancer complicated with chronic pancreatitis and chronic pancreatitis. World J Gastroenterol. 2003;9:1815-1818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
40.  Sinn M, Sinn BV, Treue D, Keilholz U, Damm F, Schmuck R, Lohneis P, Klauschen F, Striefler JK, Bahra M, Bläker H, Bischoff S, Pelzer U, Oettle H, Riess H, Budczies J, Denkert C. TP53 Mutations Predict Sensitivity to Adjuvant Gemcitabine in Patients with Pancreatic Ductal Adenocarcinoma: Next-Generation Sequencing Results from the CONKO-001 Trial. Clin Cancer Res. 2020;26:3732-3739.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 22]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
41.  Iacobuzio-Donahue CA, Wilentz RE, Argani P, Yeo CJ, Cameron JL, Kern SE, Hruban RH. Dpc4 protein in mucinous cystic neoplasms of the pancreas: frequent loss of expression in invasive carcinomas suggests a role in genetic progression. Am J Surg Pathol. 2000;24:1544-1548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 136]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
42.  Hahn SA, Schutte M, Hoque AT, Moskaluk CA, da Costa LT, Rozenblum E, Weinstein CL, Fischer A, Yeo CJ, Hruban RH, Kern SE. DPC4, a candidate tumor suppressor gene at human chromosome 18q21.1. Science. 1996;271:350-353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1723]  [Cited by in F6Publishing: 1635]  [Article Influence: 58.4]  [Reference Citation Analysis (0)]
43.  Tascilar M, Skinner HG, Rosty C, Sohn T, Wilentz RE, Offerhaus GJ, Adsay V, Abrams RA, Cameron JL, Kern SE, Yeo CJ, Hruban RH, Goggins M. The SMAD4 protein and prognosis of pancreatic ductal adenocarcinoma. Clin Cancer Res. 2001;7:4115-4121.  [PubMed]  [DOI]  [Cited in This Article: ]
44.  Schutte M, Hruban RH, Hedrick L, Cho KR, Nadasdy GM, Weinstein CL, Bova GS, Isaacs WB, Cairns P, Nawroz H, Sidransky D, Casero RA Jr, Meltzer PS, Hahn SA, Kern SE. DPC4 gene in various tumor types. Cancer Res. 1996;56:2527-2530.  [PubMed]  [DOI]  [Cited in This Article: ]
45.  Blackford A, Serrano OK, Wolfgang CL, Parmigiani G, Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Eshleman JR, Goggins M, Jaffee EM, Iacobuzio-Donahue CA, Maitra A, Cameron JL, Olino K, Schulick R, Winter J, Herman JM, Laheru D, Klein AP, Vogelstein B, Kinzler KW, Velculescu VE, Hruban RH. SMAD4 gene mutations are associated with poor prognosis in pancreatic cancer. Clin Cancer Res. 2009;15:4674-4679.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 259]  [Cited by in F6Publishing: 273]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
46.  Singh P, Srinivasan R, Wig JD. SMAD4 genetic alterations predict a worse prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas. 2012;41:541-546.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 41]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
47.  Shi L, Sheng J, Wang M, Luo H, Zhu J, Zhang B, Liu Z, Yang X. Combination Therapy of TGF-β Blockade and Commensal-derived Probiotics Provides Enhanced Antitumor Immune Response and Tumor Suppression. Theranostics. 2019;9:4115-4129.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 56]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
48.  Hsieh YY, Liu TP, Chou CJ, Chen HY, Lee KH, Yang PM. Integration of Bioinformatics Resources Reveals the Therapeutic Benefits of Gemcitabine and Cell Cycle Intervention in SMAD4-Deleted Pancreatic Ductal Adenocarcinoma. Genes (Basel). 2019;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 10]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
49.  Dardare J, Witz A, Merlin JL, Gilson P, Harlé A. SMAD4 and the TGFβ Pathway in Patients with Pancreatic Ductal Adenocarcinoma. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 47]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
50.  Bartsch D, Shevlin DW, Tung WS, Kisker O, Wells SA Jr, Goodfellow PJ. Frequent mutations of CDKN2 in primary pancreatic adenocarcinomas. Genes Chromosomes Cancer. 1995;14:189-195.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 60]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
51.  Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, Weinstein CL, Hruban RH, Yeo CJ, Kern SE. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma. Nat Genet. 1994;8:27-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 785]  [Cited by in F6Publishing: 742]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
52.  Rozenblum E, Schutte M, Goggins M, Hahn SA, Panzer S, Zahurak M, Goodman SN, Sohn TA, Hruban RH, Yeo CJ, Kern SE. Tumor-suppressive pathways in pancreatic carcinoma. Cancer Res. 1997;57:1731-1734.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Bartsch DK, Sina-Frey M, Lang S, Wild A, Gerdes B, Barth P, Kress R, Grützmann R, Colombo-Benkmann M, Ziegler A, Hahn SA, Rothmund M, Rieder H. CDKN2A germline mutations in familial pancreatic cancer. Ann Surg. 2002;236:730-737.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 121]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
54.  McWilliams RR, Wieben ED, Rabe KG, Pedersen KS, Wu Y, Sicotte H, Petersen GM. Prevalence of CDKN2A mutations in pancreatic cancer patients: implications for genetic counseling. Eur J Hum Genet. 2011;19:472-478.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 90]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
55.  Goggins M, Overbeek KA, Brand R, Syngal S, Del Chiaro M, Bartsch DK, Bassi C, Carrato A, Farrell J, Fishman EK, Fockens P, Gress TM, van Hooft JE, Hruban RH, Kastrinos F, Klein A, Lennon AM, Lucas A, Park W, Rustgi A, Simeone D, Stoffel E, Vasen HFA, Cahen DL, Canto MI, Bruno M;  International Cancer of the Pancreas Screening (CAPS) consortium. Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the International Cancer of the Pancreas Screening (CAPS) Consortium. Gut. 2020;69:7-17.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 284]  [Article Influence: 71.0]  [Reference Citation Analysis (0)]
56.  Ohtsubo K, Watanabe H, Yamaguchi Y, Hu YX, Motoo Y, Okai T, Sawabu N. Abnormalities of tumor suppressor gene p16 in pancreatic carcinoma: immunohistochemical and genetic findings compared with clinicopathological parameters. J Gastroenterol. 2003;38:663-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
57.  Gerdes B, Ramaswamy A, Ziegler A, Lang SA, Kersting M, Baumann R, Wild A, Moll R, Rothmund M, Bartsch DK. p16INK4a is a prognostic marker in resected ductal pancreatic cancer: an analysis of p16INK4a, p53, MDM2, an Rb. Ann Surg. 2002;235:51-59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 70]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
58.  Qian Y, Gong Y, Fan Z, Luo G, Huang Q, Deng S, Cheng H, Jin K, Ni Q, Yu X, Liu C. Molecular alterations and targeted therapy in pancreatic ductal adenocarcinoma. J Hematol Oncol. 2020;13:130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 152]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
59.  Wong W, Raufi AG, Safyan RA, Bates SE, Manji GA. BRCA Mutations in Pancreas Cancer: Spectrum, Current Management, Challenges and Future Prospects. Cancer Manag Res. 2020;12:2731-2742.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 55]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
60.  Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, Park JO, Hochhauser D, Arnold D, Oh DY, Reinacher-Schick A, Tortora G, Algül H, O'Reilly EM, McGuinness D, Cui KY, Schlienger K, Locker GY, Kindler HL. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N Engl J Med. 2019;381:317-327.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1137]  [Cited by in F6Publishing: 1311]  [Article Influence: 262.2]  [Reference Citation Analysis (0)]
61.  Sohal DPS, Kennedy EB, Cinar P, Conroy T, Copur MS, Crane CH, Garrido-Laguna I, Lau MW, Johnson T, Krishnamurthi S, Moravek C, O'Reilly EM, Philip PA, Pant S, Shah MA, Sahai V, Uronis HE, Zaidi N, Laheru D. Metastatic Pancreatic Cancer: ASCO Guideline Update. J Clin Oncol. 2020;JCO2001364.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 122]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
62.  Luchini C, Brosens LAA, Wood LD, Chatterjee D, Shin JI, Sciammarella C, Fiadone G, Malleo G, Salvia R, Kryklyva V, Piredda ML, Cheng L, Lawlor RT, Adsay V, Scarpa A. Comprehensive characterisation of pancreatic ductal adenocarcinoma with microsatellite instability: histology, molecular pathology and clinical implications. Gut. 2021;70:148-156.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 122]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
63.  Hu ZI, Shia J, Stadler ZK, Varghese AM, Capanu M, Salo-Mullen E, Lowery MA, Diaz LA Jr, Mandelker D, Yu KH, Zervoudakis A, Kelsen DP, Iacobuzio-Donahue CA, Klimstra DS, Saltz LB, Sahin IH, O'Reilly EM. Evaluating Mismatch Repair Deficiency in Pancreatic Adenocarcinoma: Challenges and Recommendations. Clin Cancer Res. 2018;24:1326-1336.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 239]  [Article Influence: 39.8]  [Reference Citation Analysis (0)]
64.  Bazzichetto C, Luchini C, Conciatori F, Vaccaro V, Di Cello I, Mattiolo P, Falcone I, Ferretti G, Scarpa A, Cognetti F, Milella M. Morphologic and Molecular Landscape of Pancreatic Cancer Variants as the Basis of New Therapeutic Strategies for Precision Oncology. Int J Mol Sci. 2020;21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 24]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
65.  National Comprehensive Cancer Network®  NCCN guidelines for patients: Pancreatic Cancer. National Comprehensive Cancer Network®, 2019.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Pishvaian MJ, Blais EM, Brody JR, Lyons E, DeArbeloa P, Hendifar A, Mikhail S, Chung V, Sahai V, Sohal DPS, Bellakbira S, Thach D, Rahib L, Madhavan S, Matrisian LM, Petricoin EF 3rd. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: a retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol. 2020;21:508-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 274]  [Article Influence: 68.5]  [Reference Citation Analysis (0)]
67.  Peng DF, Kanai Y, Sawada M, Ushijima S, Hiraoka N, Kitazawa S, Hirohashi S. DNA methylation of multiple tumor-related genes in association with overexpression of DNA methyltransferase 1 (DNMT1) during multistage carcinogenesis of the pancreas. Carcinogenesis. 2006;27:1160-1168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 115]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
68.  Moore MJ, Feld R, Hedley D, Oza A, Siu LL. A phase II study of temozolomide in advanced untreated pancreatic cancer. Invest New Drugs. 1998;16:77-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
69.  Valero V 3rd, Saunders TJ, He J, Weiss MJ, Cameron JL, Dholakia A, Wild AT, Shin EJ, Khashab MA, O'Broin-Lennon AM, Ali SZ, Laheru D, Hruban RH, Iacobuzio-Donahue CA, Herman JM, Wolfgang CL. Reliable Detection of Somatic Mutations in Fine Needle Aspirates of Pancreatic Cancer With Next-generation Sequencing: Implications for Surgical Management. Ann Surg. 2016;263:153-161.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 41]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
70.  Calhoun ES, Jones JB, Ashfaq R, Adsay V, Baker SJ, Valentine V, Hempen PM, Hilgers W, Yeo CJ, Hruban RH, Kern SE. BRAF and FBXW7 (CDC4, FBW7, AGO, SEL10) mutations in distinct subsets of pancreatic cancer: potential therapeutic targets. Am J Pathol. 2003;163:1255-1260.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 170]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
71.  Zeng G, Germinaro M, Micsenyi A, Monga NK, Bell A, Sood A, Malhotra V, Sood N, Midda V, Monga DK, Kokkinakis DM, Monga SP. Aberrant Wnt/beta-catenin signaling in pancreatic adenocarcinoma. Neoplasia. 2006;8:279-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 168]  [Article Influence: 9.3]  [Reference Citation Analysis (0)]
72.  Doctor A, Seifert V, Ullrich M, Hauser S, Pietzsch J. Three-Dimensional Cell Culture Systems in Radiopharmaceutical Cancer Research. Cancers (Basel). 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 27]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
73.  Lee JH, Kim SK, Khawar IA, Jeong SY, Chung S, Kuh HJ. Microfluidic co-culture of pancreatic tumor spheroids with stellate cells as a novel 3D model for investigation of stroma-mediated cell motility and drug resistance. J Exp Clin Cancer Res. 2018;37:4.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 114]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
74.  Finnberg NK, Gokare P, Lev A, Grivennikov SI, MacFarlane AW 4th, Campbell KS, Winters RM, Kaputa K, Farma JM, Abbas AE, Grasso L, Nicolaides NC, El-Deiry WS. Application of 3D tumoroid systems to define immune and cytotoxic therapeutic responses based on tumoroid and tissue slice culture molecular signatures. Oncotarget. 2017;8:66747-66757.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
75.  Ehlen L, Arndt J, Treue D, Bischoff P, Loch FN, Hahn EM, Kotsch K, Klauschen F, Beyer K, Margonis GA, Kreis ME, Kamphues C. Novel methods for in vitro modeling of pancreatic cancer reveal important aspects for successful primary cell culture. BMC Cancer. 2020;20:417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 13]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
76.  Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, Gracanin A, Oni T, Yu KH, van Boxtel R, Huch M, Rivera KD, Wilson JP, Feigin ME, Öhlund D, Handly-Santana A, Ardito-Abraham CM, Ludwig M, Elyada E, Alagesan B, Biffi G, Yordanov GN, Delcuze B, Creighton B, Wright K, Park Y, Morsink FH, Molenaar IQ, Borel Rinkes IH, Cuppen E, Hao Y, Jin Y, Nijman IJ, Iacobuzio-Donahue C, Leach SD, Pappin DJ, Hammell M, Klimstra DS, Basturk O, Hruban RH, Offerhaus GJ, Vries RG, Clevers H, Tuveson DA. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160:324-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1150]  [Cited by in F6Publishing: 1340]  [Article Influence: 134.0]  [Reference Citation Analysis (0)]
77.  Baker LA, Tiriac H, Tuveson DA. Generation and Culture of Human Pancreatic Ductal Adenocarcinoma Organoids from Resected Tumor Specimens. Methods Mol Biol. 2019;1882:97-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 23]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
78.  Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18:407-418.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 813]  [Cited by in F6Publishing: 894]  [Article Influence: 149.0]  [Reference Citation Analysis (0)]
79.  Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, Hruban RH, Maitra A, Kinzler K, Vogelstein B, Goggins M. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 2012; 142: 730-733. e9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 459]  [Cited by in F6Publishing: 484]  [Article Influence: 40.3]  [Reference Citation Analysis (0)]
80.  Guerra C, Schuhmacher AJ, Cañamero M, Grippo PJ, Verdaguer L, Pérez-Gallego L, Dubus P, Sandgren EP, Barbacid M. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 2007;11:291-302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 872]  [Cited by in F6Publishing: 889]  [Article Influence: 52.3]  [Reference Citation Analysis (0)]
81.  Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA, Kawaguchi Y, Johann D, Liotta LA, Crawford HC, Putt ME, Jacks T, Wright CV, Hruban RH, Lowy AM, Tuveson DA. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell. 2003;4:437-450.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1827]  [Cited by in F6Publishing: 1773]  [Article Influence: 84.4]  [Reference Citation Analysis (0)]
82.  Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D. RAS oncogenes: weaving a tumorigenic web. Nat Rev Cancer. 2011;11:761-774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1238]  [Cited by in F6Publishing: 1256]  [Article Influence: 96.6]  [Reference Citation Analysis (0)]
83.  Seidler B, Schmidt A, Mayr U, Nakhai H, Schmid RM, Schneider G, Saur D. A Cre-loxP-based mouse model for conditional somatic gene expression and knockdown in vivo by using avian retroviral vectors. Proc Natl Acad Sci USA. 2008;105:10137-10142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 117]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
84.  Nikiforova MN, Khalid A, Fasanella KE, McGrath KM, Brand RE, Chennat JS, Slivka A, Zeh HJ, Zureikat AH, Krasinskas AM, Ohori NP, Schoedel KE, Navina S, Mantha GS, Pai RK, Singhi AD. Integration of KRAS testing in the diagnosis of pancreatic cystic lesions: a clinical experience of 618 pancreatic cysts. Mod Pathol. 2013;26:1478-1487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 116]  [Cited by in F6Publishing: 101]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
85.  Molin MD, Matthaei H, Wu J, Blackford A, Debeljak M, Rezaee N, Wolfgang CL, Butturini G, Salvia R, Bassi C, Goggins MG, Kinzler KW, Vogelstein B, Eshleman JR, Hruban RH, Maitra A. Clinicopathological correlates of activating GNAS mutations in intraductal papillary mucinous neoplasm (IPMN) of the pancreas. Ann Surg Oncol. 2013;20:3802-3808.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 135]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
86.  Khalid A, Zahid M, Finkelstein SD, LeBlanc JK, Kaushik N, Ahmad N, Brugge WR, Edmundowicz SA, Hawes RH, McGrath KM. Pancreatic cyst fluid DNA analysis in evaluating pancreatic cysts: a report of the PANDA study. Gastrointest Endosc. 2009;69:1095-1102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 330]  [Cited by in F6Publishing: 357]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
87.  Al-Haddad M, DeWitt J, Sherman S, Schmidt CM, LeBlanc JK, McHenry L, Coté G, El Chafic AH, Luz L, Stuart JS, Johnson CS, Klochan C, Imperiale TF. Performance characteristics of molecular (DNA) analysis for the diagnosis of mucinous pancreatic cysts. Gastrointest Endosc. 2014;79:79-87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 89]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
88.  Kuboki Y, Shimizu K, Hatori T, Yamamoto M, Shibata N, Shiratori K, Furukawa T. Molecular biomarkers for progression of intraductal papillary mucinous neoplasm of the pancreas. Pancreas. 2015;44:227-235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 51]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
89.  Komatsu H, Tanji E, Sakata N, Aoki T, Motoi F, Naitoh T, Katayose Y, Egawa S, Unno M, Furukawa T. A GNAS mutation found in pancreatic intraductal papillary mucinous neoplasms induces drastic alterations of gene expression profiles with upregulation of mucin genes. PLoS One. 2014;9:e87875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 49]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
90.  Amato E, Molin MD, Mafficini A, Yu J, Malleo G, Rusev B, Fassan M, Antonello D, Sadakari Y, Castelli P, Zamboni G, Maitra A, Salvia R, Hruban RH, Bassi C, Capelli P, Lawlor RT, Goggins M, Scarpa A. Targeted next-generation sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas. J Pathol. 2014;233:217-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 249]  [Cited by in F6Publishing: 234]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
91.  Rosenbaum MW, Jones M, Dudley JC, Le LP, Iafrate AJ, Pitman MB. Next-generation sequencing adds value to the preoperative diagnosis of pancreatic cysts. Cancer Cytopathol. 2017;125:41-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 68]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
92.  Wu J, Jiao Y, Dal Molin M, Maitra A, de Wilde RF, Wood LD, Eshleman JR, Goggins MG, Wolfgang CL, Canto MI, Schulick RD, Edil BH, Choti MA, Adsay V, Klimstra DS, Offerhaus GJ, Klein AP, Kopelovich L, Carter H, Karchin R, Allen PJ, Schmidt CM, Naito Y, Diaz LA Jr, Kinzler KW, Papadopoulos N, Hruban RH, Vogelstein B. Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways. Proc Natl Acad Sci USA. 2011;108:21188-21193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 489]  [Cited by in F6Publishing: 457]  [Article Influence: 35.2]  [Reference Citation Analysis (0)]
93.  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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 321]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
94.  Singhi AD, Nikiforova MN, Fasanella KE, McGrath KM, Pai RK, Ohori NP, Bartholow TL, Brand RE, Chennat JS, Lu X, Papachristou GI, Slivka A, Zeh HJ, Zureikat AH, Lee KK, Tsung A, Mantha GS, Khalid A. Preoperative GNAS and KRAS testing in the diagnosis of pancreatic mucinous cysts. Clin Cancer Res. 2014;20:4381-4389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 131]  [Article Influence: 13.1]  [Reference Citation Analysis (0)]
95.  Wu J, Matthaei H, Maitra A, Dal Molin M, Wood LD, Eshleman JR, Goggins M, Canto MI, Schulick RD, Edil BH, Wolfgang CL, Klein AP, Diaz LA Jr, Allen PJ, Schmidt CM, Kinzler KW, Papadopoulos N, Hruban RH, Vogelstein B. Recurrent GNAS mutations define an unexpected pathway for pancreatic cyst development. Sci Transl Med. 2011;3:92ra66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 590]  [Cited by in F6Publishing: 565]  [Article Influence: 47.1]  [Reference Citation Analysis (0)]
96.  Lee JH, Kim Y, Choi JW, Kim YS. KRAS, GNAS, and RNF43 mutations in intraductal papillary mucinous neoplasm of the pancreas: a meta-analysis. Springerplus. 2016;5:1172.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 61]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
97.  Abe T, Fukushima N, Brune K, Boehm C, Sato N, Matsubayashi H, Canto M, Petersen GM, Hruban RH, Goggins M. Genome-wide allelotypes of familial pancreatic adenocarcinomas and familial and sporadic intraductal papillary mucinous neoplasms. Clin Cancer Res. 2007;13:6019-6025.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
98.  Su GH, Hruban RH, Bansal RK, Bova GS, Tang DJ, Shekher MC, Westerman AM, Entius MM, Goggins M, Yeo CJ, Kern SE. Germline and somatic mutations of the STK11/LKB1 Peutz-Jeghers gene in pancreatic and biliary cancers. Am J Pathol. 1999;154:1835-1840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 270]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
99.  Sato N, Rosty C, Jansen M, Fukushima N, Ueki T, Yeo CJ, Cameron JL, Iacobuzio-Donahue CA, Hruban RH, Goggins M. STK11/LKB1 Peutz-Jeghers gene inactivation in intraductal papillary-mucinous neoplasms of the pancreas. Am J Pathol. 2001;159:2017-2022.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 202]  [Cited by in F6Publishing: 199]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
100.  Singhi AD, McGrath K, Brand RE, Khalid A, Zeh HJ, Chennat JS, Fasanella KE, Papachristou GI, Slivka A, Bartlett DL, Dasyam AK, Hogg M, Lee KK, Marsh JW, Monaco SE, Ohori NP, Pingpank JF, Tsung A, Zureikat AH, Wald AI, Nikiforova MN. Preoperative next-generation sequencing of pancreatic cyst fluid is highly accurate in cyst classification and detection of advanced neoplasia. Gut. 2018;67:2131-2141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 210]  [Cited by in F6Publishing: 219]  [Article Influence: 36.5]  [Reference Citation Analysis (0)]
101.  Yoshizawa K, Nagai H, Sakurai S, Hironaka M, Morinaga S, Saitoh K, Fukayama M. Clonality and K-ras mutation analyses of epithelia in intraductal papillary mucinous tumor and mucinous cystic tumor of the pancreas. Virchows Arch. 2002;441:437-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 62]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
102.  Kim SG, Wu TT, Lee JH, Yun YK, Issa JP, Hamilton SR, Rashid A. Comparison of epigenetic and genetic alterations in mucinous cystic neoplasm and serous microcystic adenoma of pancreas. Mod Pathol. 2003;16:1086-1094.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 62]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
103.  Jimenez RE, Warshaw AL, Z'graggen K, Hartwig W, Taylor DZ, Compton CC, Fernández-del Castillo C. Sequential accumulation of K-ras mutations and p53 overexpression in the progression of pancreatic mucinous cystic neoplasms to malignancy. Ann Surg. 1999;230:501-9; discussion 509.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 142]  [Cited by in F6Publishing: 152]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]
104.  Garcia-Carracedo D, Chen ZM, Qiu W, Huang AS, Tang SM, Hruban RH, Su GH. PIK3CA mutations in mucinous cystic neoplasms of the pancreas. Pancreas. 2014;43:245-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 48]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
105.  Izeradjene K, Combs C, Best M, Gopinathan A, Wagner A, Grady WM, Deng CX, Hruban RH, Adsay NV, Tuveson DA, Hingorani SR. Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell. 2007;11:229-243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 284]  [Cited by in F6Publishing: 261]  [Article Influence: 15.4]  [Reference Citation Analysis (0)]
106.  Bardeesy N, Cheng KH, Berger JH, Chu GC, Pahler J, Olson P, Hezel AF, Horner J, Lauwers GY, Hanahan D, DePinho RA. Smad4 is dispensable for normal pancreas development yet critical in progression and tumor biology of pancreas cancer. Genes Dev. 2006;20:3130-3146.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 482]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
107.  Springer S, Wang Y, Dal Molin M, Masica DL, Jiao Y, Kinde I, Blackford A, Raman SP, Wolfgang CL, Tomita T, Niknafs N, Douville C, Ptak J, Dobbyn L, Allen PJ, Klimstra DS, Schattner MA, Schmidt CM, Yip-Schneider M, Cummings OW, Brand RE, Zeh HJ, Singhi AD, Scarpa A, Salvia R, Malleo G, Zamboni G, Falconi M, Jang JY, Kim SW, Kwon W, Hong SM, Song KB, Kim SC, Swan N, Murphy J, Geoghegan J, Brugge W, Fernandez-Del Castillo C, Mino-Kenudson M, Schulick R, Edil BH, Adsay V, Paulino J, van Hooft J, Yachida S, Nara S, Hiraoka N, Yamao K, Hijioka S, van der Merwe S, Goggins M, Canto MI, Ahuja N, Hirose K, Makary M, Weiss MJ, Cameron J, Pittman M, Eshleman JR, Diaz LA Jr, Papadopoulos N, Kinzler KW, Karchin R, Hruban RH, Vogelstein B, Lennon AM. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology. 2015;149:1501-1510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 310]  [Cited by in F6Publishing: 291]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
108.  Wood LD, Hruban RH. Pathology and molecular genetics of pancreatic neoplasms. Cancer J. 2012;18:492-501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 96]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
109.  Charville GW, Kao CS. Serous Neoplasms of the Pancreas: A Comprehensive Review. Arch Pathol Lab Med. 2018;142:1134-1140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
110.  Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and cadherin pathways. Science. 2004;303:1483-1487.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1944]  [Cited by in F6Publishing: 1972]  [Article Influence: 98.6]  [Reference Citation Analysis (0)]
111.  Rubinfeld B, Robbins P, El-Gamil M, Albert I, Porfiri E, Polakis P. Stabilization of beta-catenin by genetic defects in melanoma cell lines. Science. 1997;275:1790-1792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 957]  [Cited by in F6Publishing: 952]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
112.  Tanaka Y, Kato K, Notohara K, Hojo H, Ijiri R, Miyake T, Nagahara N, Sasaki F, Kitagawa N, Nakatani Y, Kobayashi Y. Frequent beta-catenin mutation and cytoplasmic/nuclear accumulation in pancreatic solid-pseudopapillary neoplasm. Cancer Res. 2001;61:8401-8404.  [PubMed]  [DOI]  [Cited in This Article: ]
113.  Abraham SC, Klimstra DS, Wilentz RE, Yeo CJ, Conlon K, Brennan M, Cameron JL, Wu TT, Hruban RH. Solid-pseudopapillary tumors of the pancreas are genetically distinct from pancreatic ductal adenocarcinomas and almost always harbor beta-catenin mutations. Am J Pathol. 2002;160:1361-1369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 363]  [Cited by in F6Publishing: 301]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
114.  Kubota Y, Kawakami H, Natsuizaka M, Kawakubo K, Marukawa K, Kudo T, Abe Y, Kubo K, Kuwatani M, Hatanaka Y, Mitsuhashi T, Matsuno Y, Sakamoto N. CTNNB1 mutational analysis of solid-pseudopapillary neoplasms of the pancreas using endoscopic ultrasound-guided fine-needle aspiration and next-generation deep sequencing. J Gastroenterol. 2015;50:203-210.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 24]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
115.  Kempski HM, Austin N, Chatters SJ, Toomey SM, Chalker J, Anderson J, Sebire NJ. Previously unidentified complex cytogenetic changes found in a pediatric case of solid-pseudopapillary neoplasm of the pancreas. Cancer Genet Cytogenet. 2006;164:54-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
116.  Selenica P, Raj N, Kumar R, Brown DN, Arqués O, Reidy D, Klimstra D, Snuderl M, Serrano J, Palmer HG, Weigelt B, Reis-Filho JS, Scaltriti M. Solid pseudopapillary neoplasms of the pancreas are dependent on the Wnt pathway. Mol Oncol. 2019;13:1684-1692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 15]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
117.  Vijayvergia N, Boland PM, Handorf E, Gustafson KS, Gong Y, Cooper HS, Sheriff F, Astsaturov I, Cohen SJ, Engstrom PF. Molecular profiling of neuroendocrine malignancies to identify prognostic and therapeutic markers: a Fox Chase Cancer Center Pilot Study. Br J Cancer. 2016;115:564-570.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
118.  Jiao Y, Shi C, Edil BH, de Wilde RF, Klimstra DS, Maitra A, Schulick RD, Tang LH, Wolfgang CL, Choti MA, Velculescu VE, Diaz LA Jr, Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos N. DAXX/ATRX, MEN1, and mTOR pathway genes are frequently altered in pancreatic neuroendocrine tumors. Science. 2011;331:1199-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1227]  [Cited by in F6Publishing: 1235]  [Article Influence: 95.0]  [Reference Citation Analysis (0)]
119.  Corbo V, Dalai I, Scardoni M, Barbi S, Beghelli S, Bersani S, Albarello L, Doglioni C, Schott C, Capelli P, Chilosi M, Boninsegna L, Becker KF, Falconi M, Scarpa A. MEN1 in pancreatic endocrine tumors: analysis of gene and protein status in 169 sporadic neoplasms reveals alterations in the vast majority of cases. Endocr Relat Cancer. 2010;17:771-783.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 118]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
120.  Missiaglia E, Dalai I, Barbi S, Beghelli S, Falconi M, della Peruta M, Piemonti L, Capurso G, Di Florio A, delle Fave G, Pederzoli P, Croce CM, Scarpa A. Pancreatic endocrine tumors: expression profiling evidences a role for AKT-mTOR pathway. J Clin Oncol. 2010;28:245-255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 391]  [Cited by in F6Publishing: 391]  [Article Influence: 26.1]  [Reference Citation Analysis (0)]
121.  Schmitt AM, Schmid S, Rudolph T, Anlauf M, Prinz C, Klöppel G, Moch H, Heitz PU, Komminoth P, Perren A. VHL inactivation is an important pathway for the development of malignant sporadic pancreatic endocrine tumors. Endocr Relat Cancer. 2009;16:1219-1227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 77]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
122.  Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, Rusev B, Scardoni M, Antonello D, Barbi S, Sikora KO, Cingarlini S, Vicentini C, McKay S, Quinn MC, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, McLean S, Nourse C, Nourbakhsh E, Wilson PJ, Anderson MJ, Fink JL, Newell F, Waddell N, Holmes O, Kazakoff SH, Leonard C, Wood S, Xu Q, Nagaraj SH, Amato E, Dalai I, Bersani S, Cataldo I, Dei Tos AP, Capelli P, Davì MV, Landoni L, Malpaga A, Miotto M, Whitehall VL, Leggett BA, Harris JL, Harris J, Jones MD, Humphris J, Chantrill LA, Chin V, Nagrial AM, Pajic M, Scarlett CJ, Pinho A, Rooman I, Toon C, Wu J, Pinese M, Cowley M, Barbour A, Mawson A, Humphrey ES, Colvin EK, Chou A, Lovell JA, Jamieson NB, Duthie F, Gingras MC, Fisher WE, Dagg RA, Lau LM, Lee M, Pickett HA, Reddel RR, Samra JS, Kench JG, Merrett ND, Epari K, Nguyen NQ, Zeps N, Falconi M, Simbolo M, Butturini G, Van Buren G, Partelli S, Fassan M;  Australian Pancreatic Cancer Genome Initiative; Khanna KK, Gill AJ, Wheeler DA, Gibbs RA, Musgrove EA, Bassi C, Tortora G, Pederzoli P, Pearson JV, Biankin AV, Grimmond SM. Whole-genome landscape of pancreatic neuroendocrine tumours. Nature. 2017;543:65-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 643]  [Cited by in F6Publishing: 578]  [Article Influence: 82.6]  [Reference Citation Analysis (0)]
123.  Rigaud G, Moore PS, Zamboni G, Orlandini S, Taruscio D, Paradisi S, Lemoine NR, Klöppel G, Scarpa A. Allelotype of pancreatic acinar cell carcinoma. Int J Cancer. 2000;88:772-777.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
124.  Jäkel C, Bergmann F, Toth R, Assenov Y, van der Duin D, Strobel O, Hank T, Klöppel G, Dorrell C, Grompe M, Moss J, Dor Y, Schirmacher P, Plass C, Popanda O, Schmezer P. Genome-wide genetic and epigenetic analyses of pancreatic acinar cell carcinomas reveal aberrations in genome stability. Nat Commun. 2017;8:1323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
125.  La Rosa S, Bernasconi B, Frattini M, Tibiletti MG, Molinari F, Furlan D, Sahnane N, Vanoli A, Albarello L, Zhang L, Notohara K, Casnedi S, Chenard MP, Adsay V, Asioli S, Capella C, Sessa F. TP53 alterations in pancreatic acinar cell carcinoma: new insights into the molecular pathology of this rare cancer. Virchows Arch. 2016;468:289-296.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 19]  [Cited by in F6Publishing: 10]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
126.  Jiao Y, Yonescu R, Offerhaus GJ, Klimstra DS, Maitra A, Eshleman JR, Herman JG, Poh W, Pelosof L, Wolfgang CL, Vogelstein B, Kinzler KW, Hruban RH, Papadopoulos N, Wood LD. Whole-exome sequencing of pancreatic neoplasms with acinar differentiation. J Pathol. 2014;232:428-435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 114]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
127.  Thompson ED, Wood LD. Pancreatic Neoplasms With Acinar Differentiation: A Review of Pathologic and Molecular Features. Arch Pathol Lab Med. 2020;144:808-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 20]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
128.  Liu W, Shia J, Gönen M, Lowery MA, O'Reilly EM, Klimstra DS. DNA mismatch repair abnormalities in acinar cell carcinoma of the pancreas: frequency and clinical significance. Pancreas. 2014;43:1264-1270.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 30]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
129.  Ducreux M, Cuhna AS, Caramella C, Hollebecque A, Burtin P, Goéré D, Seufferlein T, Haustermans K, Van Laethem JL, Conroy T, Arnold D;  ESMO Guidelines Committee. Cancer of the pancreas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;26 Suppl 5:v56-v68.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 905]  [Cited by in F6Publishing: 844]  [Article Influence: 93.8]  [Reference Citation Analysis (0)]
130.  Hong SK, Loren DE, Rogart JN, Siddiqui AA, Sendecki JA, Bibbo M, Coben RM, Meckes DP, Kowalski TE. Targeted cyst wall puncture and aspiration during EUS-FNA increases the diagnostic yield of premalignant and malignant pancreatic cysts. Gastrointest Endosc. 2012;75:775-782.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 92]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
131.  Dumonceau JM, Polkowski M, Larghi A, Vilmann P, Giovannini M, Frossard JL, Heresbach D, Pujol B, Fernández-Esparrach G, Vazquez-Sequeiros E, Ginès A;  European Society of Gastrointestinal Endoscopy. Indications, results, and clinical impact of endoscopic ultrasound (EUS)-guided sampling in gastroenterology: European Society of Gastrointestinal Endoscopy (ESGE) Clinical Guideline. Endoscopy. 2011;43:897-912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 181]  [Cited by in F6Publishing: 225]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
132.  Jenssen C, Hocke M, Fusaroli P, Gilja OH, Buscarini E, Havre RF, Ignee A, Saftoiu A, Vilmann P, Burmester E, Nolsøe CP, Nürnberg D, D'Onofrio M, Lorentzen T, Piscaglia F, Sidhu PS, Dietrich CF. EFSUMB Guidelines on Interventional Ultrasound (INVUS), Part IV - EUS-guided Interventions: General aspects and EUS-guided sampling (Long Version). Ultraschall Med. 2016;37:E33-E76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 36]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
133.  Varadarajulu S, Fockens P, Hawes RH. Best practices in endoscopic ultrasound-guided fine-needle aspiration. Clin Gastroenterol Hepatol. 2012;10:697-703.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 57]  [Cited by in F6Publishing: 65]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
134.  Pishvaian MJ, Bender RJ, Halverson D, Rahib L, Hendifar AE, Mikhail S, Chung V, Picozzi VJ, Sohal D, Blais EM, Mason K, Lyons EE, Matrisian LM, Brody JR, Madhavan S, Petricoin EF 3rd. Molecular Profiling of Patients with Pancreatic Cancer: Initial Results from the Know Your Tumor Initiative. Clin Cancer Res. 2018;24:5018-5027.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 95]  [Cited by in F6Publishing: 136]  [Article Influence: 22.7]  [Reference Citation Analysis (0)]
135.  Ko SH, Pyo JS, Son BK, Lee HY, Oh IW, Chung KH. Comparison between Conventional Smear and Liquid-Based Preparation in Endoscopic Ultrasonography-Fine Needle Aspiration Cytology of Pancreatic Lesions. Diagnostics (Basel). 2020;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
136.  Fabbri C, Fornelli A, Fuccio L, Giovanelli S, Tarantino I, Antonini F, Liotta R, Frazzoni L, Gusella P, La Marca M, Barresi L, Macarri G, Traina M, De Biase D, Fiorino S, Jovine E, Larghi A, Cennamo V. High diagnostic adequacy and accuracy of the new 20G procore needle for EUS-guided tissue acquisition: Results of a large multicentre retrospective study. Endosc Ultrasound. 2019;8:261-268.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
137.  Sho S, Court CM, Kim S, Braxton DR, Hou S, Muthusamy VR, Watson RR, Sedarat A, Tseng HR, Tomlinson JS. Digital PCR Improves Mutation Analysis in Pancreas Fine Needle Aspiration Biopsy Specimens. PLoS One. 2017;12:e0170897.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
138.  de Biase D, Visani M, Acquaviva G, Fornelli A, Masetti M, Fabbri C, Pession A, Tallini G. The Role of Next-Generation Sequencing in the Cytologic Diagnosis of Pancreatic Lesions. Arch Pathol Lab Med. 2018;142:458-464.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 18]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
139.  de Biase D, Fassan M, Malapelle U. Next-Generation Sequencing in Tumor Diagnosis and Treatment. Diagnostics (Basel). 2020;10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
140.  Mosele F, Remon J, Mateo J, Westphalen CB, Barlesi F, Lolkema MP, Normanno N, Scarpa A, Robson M, Meric-Bernstam F, Wagle N, Stenzinger A, Bonastre J, Bayle A, Michiels S, Bièche I, Rouleau E, Jezdic S, Douillard JY, Reis-Filho JS, Dienstmann R, André F. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020;31:1491-1505.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 569]  [Cited by in F6Publishing: 558]  [Article Influence: 139.5]  [Reference Citation Analysis (0)]