Context.—

Tumoral (grossly visible) intraductal neoplasms of the bile ducts are still being characterized.

Objective.—

To investigate their morphologic, immunohistochemical, and molecular features.

Design.—

Forty-one cases were classified as gastric-, intestinal-, pancreatobiliary-type intraductal papillary neoplasm (IPN), intraductal oncocytic papillary neoplasm (IOPN), or intraductal tubulopapillary neoplasm (ITPN) on the basis of histology. All neoplasms were subjected to targeted next-generation sequencing.

Results.—

The mean age at diagnosis was 69 years (42–81 years); male to female ratio was 1.3. Most neoplasms (n = 23, 56%) were extrahepatic/large (mean size, 4.6 cm). The majority (n = 32, 78%) contained high-grade dysplasia, and 68% (n = 28) revealed invasion. All gastric-type IPNs (n = 9) and most ITPNs/IOPNs showed consistent colabeling for CK7/MUC6, which was less common among others (P = .004). Intestinal-type IPNs (n = 5) showed higher rates of CK20 expression than others (P < .001). Overall, the most commonly mutated genes included TP53 and APC, while copy number variants affected ELF3 and CDKN2A/B. All gastric-type IPNs contained an alteration affecting the Wnt signaling pathway; 7 of 9 (78%) showed aberrations in the MAPK pathway. Mutations in APC and KRAS were common in gastric-type IPNs as compared with others (P = .01 for both). SMAD4 was more frequently mutated in intestinal-type IPNs (P = .02). Pancreatobiliary-type IPNs (n = 14) exhibited frequent alterations in tumor suppressor genes including TP53, CDKN2A/B, and ARID2 (P = .04, P = .01 and P = .002, respectively). Of 6 IOPNs analyzed, 3 (50%) revealed ATP1B1-PRKACB fusion. ITPNs (n = 6) showed relatively few recurrent genetic aberrations. Follow-up information was available for 38 patients (median, 58.5 months). The ratio of disease-related deaths was higher for the cases with invasion (56% versus 10%).

Conclusions.—

Tumoral intraductal neoplasms of the bile ducts, similar to their counterparts in the pancreas, are morphologically and genetically heterogeneous.

Tumoral (grossly visible) intraductal neoplasms of the bile ducts are rare tumors, which exhibit variable histomorphology and behavior. The nomenclature for these biliary neoplasms has been inconsistent, and they have been referred to as papilloma, papillary adenoma, papillary carcinoma, papillomatosis, and biliary intraductal papillary mucinous neoplasm.1  The World Health Organization (WHO) currently endorses the term intraductal papillary neoplasm of the bile ducts to encompass all tumoral intraductal biliary neoplasms, although specific morphologic subtypes are commonly designated separately (see below).2  These biliary neoplasms follow a dysplasia-carcinoma sequence and, thus, are regarded as preinvasive neoplasms.3  Although the incidence of carcinoma development is not well established, the prevalence of invasive carcinoma among patients with a tumoral intraductal neoplasm varies by geographic region, ranging from 20% to 80%.38 

Tumoral intraductal neoplasms of the bile ducts can exhibit gastric, intestinal, pancreatobiliary, or oncocytic differentiation and some show predominantly tubular/tubulopapillary growth patterns.1,2,5,912  These morphologic features mirror the histology of the more common pancreatic intraductal papillary mucinous neoplasms (IPMNs),1214  intraductal oncocytic papillary neoplasm (IOPN),1519  and intraductal tubulopapillary neoplasm (ITPN).20,21  In the pancreas, detailed molecular studies have revealed that IPMNs, IOPN, and ITPN exhibit different genetic alterations. IPMNs frequently harbor alterations in KRAS, GNAS, and RNF43,2227  while IOPNs have recurrent alterations in ARHGAP26, ASXL1, EPHA8, and ERBB4.16,17  Three recent studies analyzing pancreatic and biliary IOPNs by RNA-based targeted sequencing also revealed specific variant fusions of DNAJB1-PRKACA, ATP1B1-PRKACA, or ATP1B1-PRKACB.19,28,29  Pancreatic ITPNs exhibit mutations in chromatin remodelling genes such as MLL1, MLL2, MLL3, and BAP1. A subset of the cases harbors phosphatidylinositol 3-kinase (PI3K) pathway (PIK3CA, PIK3CB, INPP4A, and PTEN) mutations or FGFR2 fusions.20  Therefore, pancreatic IOPNs and ITPNs are now classified as distinct entities, also recognized by the WHO as separate categories.30,31 

The similarities between tumoral intraductal neoplasms of the bile ducts and the pancreas raise the question of whether they are analogous or even identical tumors. However, data regarding the genetic features of tumoral intraductal neoplasms of the bile ducts are still limited by the rarity of these neoplasms. In one of the first studies analyzing the genetic landscape of tumoral intraductal neoplasms of the bile ducts, whole exome sequencing showed them to be genetically distinct from cholangiocarcinomas, frequently harboring APC and CTNNB1 mutations.32  Another study found that tumoral intraductal neoplasms of the bile ducts may show heterogeneous molecular alterations, with activation of Ras–mitogen-activated protein kinase (MAPK), Wnt/β-catenin, and G-protein-coupled receptor (GPCR)–cAMP signaling.33  Recently, Goeppert et al34  reported that intraductal papillary neoplasms of the bile ducts are enriched in mutations typical for extrahepatic cholangiocarcinoma, whereas intraductal tubulopapillary neoplasms of the bile ducts exhibit few genetic alterations, suggesting distinct oncogenic mechanisms specific to these distinct precursor subtypes. They also found that papillary and tubulopapillary neoplasms differ in their DNA methylation profiles.

In this study, we aimed to further define the histologic, immunohistochemical, and genetic underpinnings of tumoral intraductal neoplasms of the bile ducts through the analysis of 41 cases.

With approval of the institutional review boards, the surgical pathology and consultation files of the authors between 1994 and 2018 were searched for cases of tumoral (grossly visible) intraductal neoplasms of the bile ducts with or without an associated invasive carcinoma. Tumoral intraductal neoplasms that were not entirely submitted for microscopic evaluation as well as dysplastic changes that did not form grossly visible lesions (nontumoral dysplasia, biliary intraepithelial neoplasia) were excluded.35  Available gross photographs and descriptions as well as all histologic sections were re-evaluated to confirm the diagnoses and further characterize the spectrum of architecture (eg, papillary, tubulopapillary, and tubular), morphologic differentiation (eg, pancreatobiliary, gastric, intestinal, and oncocytic), and presence, pattern, and extent of invasive carcinoma by using the current WHO criteria for pancreatic and biliary intraductal neoplasms.2,30,31,36  Available medical records, including imaging study reports, were reviewed to obtain clinical data regarding age, sex, tumor size, location (intrahepatic or extrahepatic), type of surgical intervention, margin status, and date of last follow-up. Contributing physicians were contacted to obtain this information for consultation cases.

Immunohistochemical Labeling

Representative full-faced formalin-fixed, paraffin-embedded tissue sections from each case were immunolabeled for MUC1, MUC2, MUC5AC, MUC6, CK7, CK20, and CDX2 by using the standard avidin-biotin peroxidase method (Supplemental Table 1; see supplemental digital content at https://meridian.allenpress.com/aplm in the December 2023 table of contents). The controls used were as follows: Nonneoplastic tissues and carcinomas from the pancreas (MUC1 and CK7), colon (MUC2, CK20 and CDX2), and stomach (MUC5AC and MUC6) served as controls. The staining reaction was evaluated in tumor cell membranes and cytoplasm (MUC1, MUC2, MUC5AC, MUC6), cytoplasm (CK7 and CK20), and nucleus (CDX2). Moderate to strong staining of at least 10% of the lesional cells was considered to represent a positive result.

Targeted Next-Generation Sequencing and Molecular Pathway Annotation

A representative formalin-fixed, paraffin-embedded block of neoplastic and normal control tissue from each case was selected for next-generation sequencing by the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) platform. For each case, twenty 10-μm sections of tissue were cut. Owing to small size of the invasive carcinoma component in most cases, only the intraductal component was needle microdissected. DNA was extracted from intraductal neoplasms and normal tissues. Deep coverage, targeted next-generation sequencing was performed for a panel of 468 genes known to undergo somatic genomic alterations in cancer (Supplemental Table 2).16,20  Briefly, massively parallel sequencing libraries (Kapa Biosystems, New England Biolabs) with barcoded universal primers were generated from 115 to 250 ng of genomic DNA from each case. After amplification and DNA quantification, the barcoded libraries were subjected to solution-phase hybrid capture with synthetic biotinylated DNA probes (Nimblegen Seq-Cap) that target exons from all key genes and introns known to harbor recurrent translocation breakpoints. Each hybrid capture pool was sequenced with deep coverage in a single paired-end lane of an Illumina flow cell. The sequencing data were analyzed for single-nucleotide variants (SNVs), small insertion/deletions (indels), and copy number variations. Somatic SNVs and indels were called by using MuTect and the SomaticIndelDetector tools in the Genome Analysis Toolkit.37,38  All candidate mutations and indels were reviewed manually with the Integrative Genomics Viewer.39  The mean coverage was 400× for tumor DNA and 50× for normal DNA. Altered genes were categorized into molecular pathways, based on review of the GeneCards Human Gene integrative database (Supplemental Table 3).

Fusion Assay

A representative formalin-fixed, paraffin-embedded block of neoplastic tissue from IOPNs with enough material (n = 6) was selected for the MSK-Fusion assay. The MSK-Fusion assay is a custom targeted, RNA-based panel that uses Archer Anchored Multiplex PCR (polymerase chain reaction) technology and next-generation sequencing to detect gene fusions in 62 genes (including DNAJB1, PRKACA, and ATP1B1) known to be involved in chromosomal rearrangements.4042  These custom assays have been validated and approved for clinical use at Memorial Sloan Kettering Cancer Center (New York, New York) by the New York State Department of Health Clinical Laboratory Evaluation Program.

Statistical Analysis

Statistical comparisons of binomial data were performed by using the Fisher exact test. Statistical comparisons of continuous data were made with the Student t test. Survival comparisons were made by log-rank test of Kaplan-Meier survival curves. Unsupervised hierarchical clustering of the cases, based on molecular pathways, was performed by the Ward method using Euclidean distance measurements.

The clinicopathologic features of the 41 cases analyzed are summarized in Table 1.

Table 1

Clinical Characteristics

Clinical Characteristics
Clinical Characteristics

Clinical Findings

The study group included 18 females and 23 males who ranged from 42 to 81 years in age (mean, 69 years). Presenting symptoms included jaundice, dark urine, pruritus, bloating, abdominal pain, loss of appetite, nausea, vomiting, and weight loss. One patient had primary sclerosing cholangitis. Three patients had a history of a prior malignancy, including one each with breast carcinoma, colonic adenocarcinoma, and prostatic carcinoma.

Tumors were relatively evenly distributed throughout the biliary tract: Of the 41 tumors, 44% (n = 18) were intrahepatic, 27% (n = 11) were hilar, and 29% (n = 12) were located within the distal bile duct. All patients were treated by surgical resection, and none received neoadjuvant chemotherapy. Nine of 11 patients with disease recurrence or metastases received chemotherapy and/or radiation therapy.

Pathologic Findings

The tumors ranged from 1.2 to 14 cm (mean, 4.6 cm) in greatest dimension. The intraductal nature of the tumor was specifically documented in 39 of 41 cases (95%) (Figure 1, A and B). Most tumors were described as friable/papillary (n = 19, 46%) or polypoid (n = 11, 27%) masses. Nine (22%) were described as predominantly solid and 2 as cystic (5%). None of the tumors were accompanied by grossly visible mucin in the lumen.

Figure 1

A, Intrahepatic intraductal papillary neoplasm of the bile ducts, a papillary tumor within a dilated intrahepatic bile duct (between green stars). B, Extrahepatic intraductal papillary neoplasm of the bile ducts, with friable papillary projections, arising from the distal common bile duct (significantly dilated). Main PD appears unremarkable. Abbreviation: PD, pancreatic duct.

Figure 1

A, Intrahepatic intraductal papillary neoplasm of the bile ducts, a papillary tumor within a dilated intrahepatic bile duct (between green stars). B, Extrahepatic intraductal papillary neoplasm of the bile ducts, with friable papillary projections, arising from the distal common bile duct (significantly dilated). Main PD appears unremarkable. Abbreviation: PD, pancreatic duct.

Close modal

Most tumors (n = 29, 71%) displayed a predominantly papillary or tubulopapillary growth pattern, and the remainder (n = 12, 29%) displayed a tubular growth pattern. Pancreatobiliary differentiation was most common (n = 14, 34%), whereas intestinal differentiation was least common (n = 5, 12%) (Figure 2, A through F). Seven of 41 cases (17%) showed the characteristic features of an IOPN and were composed of arborizing papillae punctuated by intraepithelial lumina and lined by 1 to 2 cell layers of oncocytic cells with large, uniform nuclei and single, prominent nucleoli resembling pancreatic IOPN (Figure 3, A and B).15,17,18  Six of 41 cases (15%) had a predominantly tubular growth pattern characterized by back-to-back small glands lined by cuboidal cells with minimal to modest amounts of cytoplasm, lacking obvious intracellular mucin, and round to ovoid nuclei reminiscent of pancreatic ITPN (Figure 3, C and D).20 

Figure 2

Pancreatobiliary-type intraductal papillary neoplasm of the bile ducts with complex, interconnecting papillae (A) lined by atypical cuboidal cells that have little cytoplasmic mucin and enlarged round nuclei (B). Gastric-type intraductal papillary neoplasm of the bile ducts with relatively simple and typically tubular growth pattern (C). The tubules are lined by simple epithelium that resembles gastric foveolar epithelium (D). Intestinal-type intraductal papillary neoplasm of the bile ducts has a papillary (villous) growth pattern (E) and reveals pseudostratified columnar cells with a basophilic appearance and apical goblet-like mucin (F) (hematoxylin-eosin, original magnification ×40 [A, C, and E]; original magnification ×200 [B, D, and F]).

Figure 2

Pancreatobiliary-type intraductal papillary neoplasm of the bile ducts with complex, interconnecting papillae (A) lined by atypical cuboidal cells that have little cytoplasmic mucin and enlarged round nuclei (B). Gastric-type intraductal papillary neoplasm of the bile ducts with relatively simple and typically tubular growth pattern (C). The tubules are lined by simple epithelium that resembles gastric foveolar epithelium (D). Intestinal-type intraductal papillary neoplasm of the bile ducts has a papillary (villous) growth pattern (E) and reveals pseudostratified columnar cells with a basophilic appearance and apical goblet-like mucin (F) (hematoxylin-eosin, original magnification ×40 [A, C, and E]; original magnification ×200 [B, D, and F]).

Close modal
Figure 3

Intraductal oncocytic papillary neoplasm of bile ducts with complex papillary projections lined by stratified cuboidal or columnar cells (A). The cells have distinctive oncocytic cytoplasm and nuclei with single, prominent, eccentric nucleoli. Intracellular lumens are also present (B). Intraductal tubulopapillary neoplasm of bile ducts composed of back-to-back tubular glands or punctuated solid areas (C). The cuboidal tumor cells have modest amounts of cytoplasm without obvious intracellular mucin content. The nuclei are small, round to oval, and moderately to markedly atypical (D) (hematoxylin-eosin, original magnification ×40 [A and C]; original magnification ×200 [B and D]).

Figure 3

Intraductal oncocytic papillary neoplasm of bile ducts with complex papillary projections lined by stratified cuboidal or columnar cells (A). The cells have distinctive oncocytic cytoplasm and nuclei with single, prominent, eccentric nucleoli. Intracellular lumens are also present (B). Intraductal tubulopapillary neoplasm of bile ducts composed of back-to-back tubular glands or punctuated solid areas (C). The cuboidal tumor cells have modest amounts of cytoplasm without obvious intracellular mucin content. The nuclei are small, round to oval, and moderately to markedly atypical (D) (hematoxylin-eosin, original magnification ×40 [A and C]; original magnification ×200 [B and D]).

Close modal

Most tumors (n = 32, 78%) contained areas of high-grade dysplasia and 28 of 41 (68%) harbored an associated invasive carcinoma component. Of note, tumors with gastric differentiation were predominantly noninvasive (67% versus 22% in other subtypes, P = .02) and tumors with pancreatobiliary differentiation were predominantly invasive (93% versus 56% in other subtypes, P = .03). The invasive component was of tubular type in 26 of 28 cases (93%) and colloid type in 2 cases (7%), both with intestinal differentiation. One biliary IOPN showed stromal mucin accumulation around the invasive component, resembling a colloid carcinoma. Of the known 30 cases, lymphovascular invasion and perineural invasion was detected in 7 (23%) and 8 (27%) cases, respectively. A lymph node dissection was performed in 26 cases; only 1 case was associated with lymph node metastases.

Immunohistochemical Findings

Immunohistochemical staining results did not consistently distinguish between different morphologic subtypes (Table 2). All morphologic subtypes showed staining for CK7 and MUC1. Although 2 of 5 intestinal-type IPNs (40%) showed MUC2 staining, this marker was also positive in 3 of 13 pancreatobiliary-type IPNs (23%) and 3 of 6 IOPNs (50%). Similarly, MUC5AC and MUC6 were also expressed in most subtypes. Only CK20 showed higher rates of expression in intestinal-type IPNs than other subtypes (80% versus 9%; P < .001). CDX2 was not sensitive for any of the subtypes. Of note, all gastric-type IPNs as well as a vast majority (4 of 6, 67%) of ITPNs and IOPNs showed consistent CK7 and MUC6 coexpression, which was less common among other subtypes (81% versus 33%; P = .004). ITPNs were also consistently negative for MUC5AC.

Table 2

Immunohistochemical Staining by Tumor Differentiation (n = 39)

Immunohistochemical Staining by Tumor Differentiation (n = 39)
Immunohistochemical Staining by Tumor Differentiation (n = 39)

Genetic Findings

The results of molecular studies are summarized in Tables 3 and 4, and details regarding each of the individual cases are described in Supplemental Tables 4 and 5.

Table 3

Recurrent Genetic Alterations and Dominant Histologic Subtypes (n = 41)a

Recurrent Genetic Alterations and Dominant Histologic Subtypes (n = 41)a
Recurrent Genetic Alterations and Dominant Histologic Subtypes (n = 41)a
Table 4

Genes Altered by Subtype

Genes Altered by Subtype
Genes Altered by Subtype

No germline alterations were identified in any cases. The overall number of alterations, including both somatic mutations and copy number variations, per case ranged from 0 to 64 (median, 9; Figure 4) with the highest number of alterations occurring in a mismatch repair–deficient tumor. The most altered gene was ELF3, which encodes a transcription factor; 13 of 41 cases (32%) harbored either mutations or copy number variations. Of 41 cases in our cohort, other frequently altered genes included TP53 (n = 9, 22%), APC (n = 9, 22%), KRAS (n = 6, 15%), AXIN1 (n = 6, 15%), SMAD4 (n = 6, 15%), CDKN2A/B (n = 6, 15%), SOX9 (n = 6, 15%), ARID2 (n = 5, 12%), ATM (n = 5, 12%), and HIST2H3C/D (n = 5, 12%).

Figure 4

Median number of alterations, including both somatic mutations and CNVs, is 9 (range, 0–64). The case with the highest number of alterations is a mismatch repair–deficient case. Abbreviation: CNV, copy number variation.

Figure 4

Median number of alterations, including both somatic mutations and CNVs, is 9 (range, 0–64). The case with the highest number of alterations is a mismatch repair–deficient case. Abbreviation: CNV, copy number variation.

Close modal

Molecular abnormalities varied somewhat depending on morphologic subtypes (Tables 3 and 4). Pancreatobiliary-type IPNs (n = 14) more commonly harbored alterations in TP53 (n = 6, 43%), ARID2 (n = 5, 36%), and CDKN2A/B (n = 5, 36%) than other subtypes (P = .04, P = .002, and P = .01, respectively). On the other hand, gastric-type IPNs (n = 9) showed higher rates of APC (n = 5, 56%) and KRAS (n = 4, 44%) mutations than other subtypes (P = .01 for both), and intestinal-type IPNs (n = 5) were more likely to harbor mutations in SMAD4 (n = 3, 60%) than other subtypes (P = .02). Two of 7 biliary IOPNs (29%) harbored mutations in BLM, which were not seen in any other tumor type (P = .03). One of 7 other biliary IOPNs (14%) was found to have a mutation in ASXL1 similar to that previously reported in pancreatic IOPNs.16  Moreover, 3 of 6 IOPNs (50%) tested revealed an ATP1B1-PRKACB fusion, which was previously reported in both pancreatic and biliary IOPNs.19,28,29  The ITPNs (n = 6) exhibited some recurrent alterations in SWI/SNF complex genes, FGF/FGFR, and PI3K, but none of these alterations were preferential for this subtype (P = .15). Although alterations in the KMT (also known as MLL) lysine methyltransferase family are detected in pancreatic ITPNs,20  they were not found in biliary ITPNs in this series. Rather, 6 IPNs of other subtypes had alterations in genes of the KMT family.

When genes altered were categorized into the predominant pathways (Table 5), the 2 categories most affected were Wnt signaling (n = 21) and DNA damage repair (n = 21), followed in frequency by RAS-MAPK (n = 20) and PI3K (n = 18). Alterations affecting RB1 (n = 15), TP53 (n = 14), SWI/SNF (n = 13), and NOTCH (n = 11) were less common. All gastric-type IPNs harbored at least 1 Wnt pathway gene alteration, which was less common in other subtypes (P = .001). Although MAPK pathway–related alterations were relatively more common in gastric-type IPNs (n = 7, 78%), there was no statistically significant difference among subtypes (P = .07). Tumors with an intestinal phenotype showed disproportionate TGF-β alterations (n = 3, 60%, P = .04) and SWI/SNF alterations were common among tumors with a pancreatobiliary phenotype (n = 8, 57%; P = .02). However, there was no statistically significant difference between intraheaptic and extrahepatic IPNs (Table 6).

Table 5

Common Pathways With Alterations in Intraductal Papillary Neoplasms of Bile Ducts

Common Pathways With Alterations in Intraductal Papillary Neoplasms of Bile Ducts
Common Pathways With Alterations in Intraductal Papillary Neoplasms of Bile Ducts
Table 6

The Most Commonly Altered Pathways by Tumor Location

The Most Commonly Altered Pathways by Tumor Location
The Most Commonly Altered Pathways by Tumor Location

Hierarchical clustering based on the pathways revealed 2 major tumor groups: gastric-type, noninvasive IPNs located within the intrahepatic bile ducts and pancreatobiliary-type IPNs with invasive carcinoma located within the distal bile ducts. Pancreatobiliary-type IPNs had more genetic alterations than other groups (15 versus 4; P = .002) affecting PI3K, histone modifier, SWI/SNF, p53, p16/RB, and TGF-β pathways (Figures 5 and 6).

Figure 5

The most altered molecular pathways.

Figure 5

The most altered molecular pathways.

Close modal
Figure 6

Hierarchical clustering based on the pathways reveals 2 groups. Group 1 is characterized by more gastric-type, noninvasive intraductal papillary neoplasm of the bile ducts that is located within the intrahepatic bile ducts. Group 2 is characterized by more pancreatobiliary-type intraductal papillary neoplasm of the bile ducts with invasive carcinoma that is located within the distal bile ducts. Group 2 harbors more alterations on average and several pathways, including PI3K, histone modifier, SWI/SNF, p53, p16/RB, and TGF-β and are significantly more common in Group 2. Abbreviations: IOPN, intraductal oncocytic papillary neoplasm; ITPN, intraductal tubulopapillary neoplasm.

Figure 6

Hierarchical clustering based on the pathways reveals 2 groups. Group 1 is characterized by more gastric-type, noninvasive intraductal papillary neoplasm of the bile ducts that is located within the intrahepatic bile ducts. Group 2 is characterized by more pancreatobiliary-type intraductal papillary neoplasm of the bile ducts with invasive carcinoma that is located within the distal bile ducts. Group 2 harbors more alterations on average and several pathways, including PI3K, histone modifier, SWI/SNF, p53, p16/RB, and TGF-β and are significantly more common in Group 2. Abbreviations: IOPN, intraductal oncocytic papillary neoplasm; ITPN, intraductal tubulopapillary neoplasm.

Close modal

Clinical Outcome

Clinical follow-up information was available for 38 of 41 patients (93%) with a median follow-up of 58.5 months (range, 4–143 months). Fifteen of 38 patients (39.5%) died of disease; sites of dissemination included lung (n = 4), and/or brain (n = 2), and/or liver (n = 3). Six of 38 patients (16%) were alive with disease with a median follow-up of 31 months (range, 4–76 months). Six of 38 patients (16%) were alive with no evidence of disease with a median follow-up of 83.5 months (range, 15–129 months).

Overall, the 5-year survival was 80% for patients who had a noninvasive tumoral intraductal neoplasm of the bile ducts, compared with 60% for patients who had a tumoral intraductal neoplasm of the bile ducts associated with invasive carcinoma. Although the ratio of disease-related deaths was higher for patients who had a tumoral intraductal neoplasm of the bile ducts associated with invasive carcinoma (56% versus 10%), since the number of disease-related deaths in the noninvasive group was low, the difference was not found to be statistically significant by Kaplan-Meier log-rank test (P = .1). There was also no statistically significant difference in survival rates between intrahepatic and extrahepatic IPNs (Table 7). Similarly, age, sex, the presence of invasive carcinoma, tumor size, lymphovascular invasion, perineural invasion, incomplete tumor resection, and lymph node metastases were not predictive of overall survival. Moreover, there was no correlation among genetic alterations or pathways affected and clinical outcome.

Table 7

Survival by Tumor Location

Survival by Tumor Location
Survival by Tumor Location

In this study, we explored the molecular characteristics of 41 tumoral intraductal neoplasms of the bile ducts, one of the largest series analyzed to date, by high-depth targeted next-generation sequencing for a large panel of key cancer-associated genes (n = 468), along with a clinicopathologic correlative analysis. Various previously unrecognized characteristics of these tumors and their subtypes were elucidated.

Clinicopathologic Characteristics

Our results confirm that tumoral intraductal neoplasms of the bile ducts are indeed highly analogous to their pancreatic counterparts in their display of a range of histomorphologic differentiation with distinct characteristics. However, IOPNs (n = 7, 17%) and ITPNs (n = 6, 15%), which are not recognized as distinct entities by the current (2019) WHO classification, seem to be represented at a higher percentage among tumoral intraductal neoplasms of the bile ducts than in the pancreas.30,31  Also, intestinal-type IPNs are relatively rare in our cohort (n = 5, 12%) but this could be population related, as previous studies analyzing tumoral intraductal neoplasms of the bile ducts in Eastern cohorts reported a higher proportion (up to 50%) of intestinal-type IPNs.4346  Of note, while other subtypes are found in both intrahepatic and extrahepatic locations, all our intestinal-type IPNs were extrahepatic. Immunohistochemical studies revealed significant overlaps between subtypes (such as CK7 and MUC1 expression is very common regardless of subtype, including 60% of the intestinal-type IPNs). However, there were some staining trends. For example, CK20 and CDX2 expression was identified only in the intestinal-type IPNs, even though the sensitivity was poor (80% and 20%, respectively), whereas MUC6 was not detected in any of the intestinal-type IPNs. Moreover, gastric-type IPNs, biliary IOPNs, and biliary ITPNs were characterized with CK7 and MUC6 coexpression. Gastric-type IPNs may also express MUC5AC but biliary ITPNs consistently lacked MUC5AC, resembling pancreatic intraductal neoplasms (Table 2).18,21  Previous studies analyzing smaller series have also reported similar results.1,5  As such, current data suggest that immunohistochemical staining may not be helpful in subtyping these neoplasms.

Akin to IPMNs, the gastric-type of IPN was most frequently associated with noninvasive disease (n = 6, 67%) and the pancreatobiliary-type had the highest rate of invasion (n = 13, 93%) and the invasion was of tubular type.47,48  Colloid carcinoma was relatively rare but when it occurred, it arose in association with intestinal-type, as is the case in the pancreas. Moreover, biliary IOPNs may also exhibit stromal mucinous changes and mimic colloid carcinoma.

Despite the presence of invasion in the majority of cases (n = 28, 68%), tumoral intraductal neoplasms of the bile ducts followed a protracted clinical course, akin to their pancreatic counterparts.16,20,30,31,36  In a median follow-up of 58.5 months, the 5-year survival was 80% for patients who had a noninvasive tumoral intraductal neoplasm of the bile ducts and 60% for patients who had an invasive tumoral intraductal neoplasm of the bile ducts. These outcomes are similar to prior reports.3,4,8  However, there was no statistical difference in survival between our noninvasive and invasive tumoral intraductal neoplasms of the bile ducts, which is rather unexpected. This might be, in part, due to the relatively low number of noninvasive tumoral intraductal neoplasms of the bile ducts. Initially, we thought the small size of invasive component in invasive tumoral intraductal neoplasms of the bile ducts might be a factor as well (there are 9 cases in our cohort that exhibit minimal [ie, <10% of the tumor] invasion). However, even when these cases were included in the noninvasive cohort, the status of invasion still did not affect survival, raising the possibility of inaccuracies in identifying and quantifying the extent of invasion. In fact, because of their tendency to grow in lobulated configurations, introducing a degree of subjectivity in distinguishing between expansile (pushing-border invasion) versus true intraductal (noninvasive) growth, assessing invasion can be very challenging in these tumors. Of note, there was also 1 patient with gastric-type IPN without identifiable invasive carcinoma who developed a biopsy-proven metastasis in the lung and died of disease. Multifocality and field effect phenomenon49  may be factors in such cases with unexpected progression and emphasize the importance of extensive sampling and thorough examination along with long-term follow-up. While earlier studies showed that a positive surgical margin was also correlated with worse outcomes, only 3 tumors in our cohort had a positive margin, which limits the analysis of this feature.3,46,50 

Molecular Characteristics

Our results also illustrate that while tumoral intraductal neoplasms of the bile ducts show some genetic similarities to other pancreatobiliary neoplasms, including aberrations in KRAS, CDKN2A, TP53, and SMAD4, which are commonly seen in pancreatic ductal adenocarcinoma, IPMNs, and conventional bile duct carcinoma,51  the genetic alterations and their corresponding prevalence are not always congruent. For example, ELF3, which encodes a transcription factor with possible interactions with ErbB as well as Notch signaling pathways, was altered in about one-third of tumoral intraductal neoplasms of the bile ducts; however, it has been rarely reported in pancreas (1%) and ampullary (2%–12%) carcinomas.52,53  Similarly, APC mutations were more common in tumoral intraductal neoplasms of the bile ducts (22%) than in conventional bile duct adenocarcinomas (3%–7%).33,54  Emerging studies analyzing smaller numbers of cases and/or a limited panel of genes have also reported similar results.32,33,54  For example, in one of the first studies analyzing the genetic landscape of IPNs, Fujikura et al32  performed whole exome sequencing on 7 cases, followed by validation targeted sequencing on 7 additional cases, and identified mutations in APC or CTNNB1 in 6 cases (43%). Additionally, in contrast with pancreatic IPMNs, especially intestinal-type IPMNs, which show relatively high frequency of RNF43 and GNAS mutations,27 RNF43 was mutated only in 3 cases in our cohort (1 intestinal-type IPN, 1 gastric-type IPN, and 1 IOPN) and another case of intestinal-type IPN harbored GNAS mutation. However, previous studies analyzing RNF43 and/or GNAS mutations in Eastern cohorts that have many more (50%) intestinal-type IPNs reported higher frequencies for both mutations, and the mutations were significantly more common in intestinal-type IPNs.25,44,45,55  Therefore, it is possible that the difference between those studies and our results is due to the relatively low number (n = 5, 12%) of intestinal-type IPNs in our cohort.

Unfortunately, owing to the small size of the invasive carcinoma component in most of our cases, we could not sequence the invasive carcinoma component separately. Therefore, we are unable to compare the spectrum of alterations in invasive tumoral intraductal neoplasms of the bile ducts with conventional bile duct adenocarcinomas.

The molecular findings in this study also support the separation of biliary IOPNs and ITPNs from the other tumoral intraductal neoplasms of the bile ducts subtypes. To the best of our knowledge, our study has the largest cohort to date analyzing molecular features of biliary IOPNs. Similar to their pancreatic counterparts, all 7 biliary IOPNs analyzed in this study lack the mutations commonly found in other subtypes of pancreatic and biliary intraductal neoplasms. Also, of several recurrently altered genes in pancreatic IOPNs,16 ASXL1 is found to be mutated in a case of biliary IOPN. Moreover, 50% of the biliary IOPNs revealed an ATP1B1-PRKACB fusion, which was previously reported in both pancreatic and biliary IOPNs.19,28,29  Of note, compared to pancreatic IOPNs, although the frequency of the fusion is lower in our current study, it is most likely due to the old nature of the negative cases, which were resected before 2005 (ie, suboptimal tissue quality for fusion assay). Similarly, this study confirms that biliary ITPNs do not have KRAS mutations, nor do they have mutations in Wnt pathway genes. Previous studies on biliary ITPNs also revealed a very low prevalence of alterations in common oncogenic signaling pathways.5,34,56  Instead, alterations in chromatin remodelling, SWI/SNF complex, PI3K pathway, and FGFR pathway genes seem to be significantly associated with biliary ITPNs as is the case for the pancreatic ITPNs.20  Overall, these findings demonstrate that biliary IOPN and ITPN are different from the other subtypes of biliary intraductal neoplasms not only based on their distinctive histopathologic features, but also based on the genetic pathways of tumor development.

Some of these specific genes or pathways are of significance because there are targeted therapies being developed, some of which are already in clinical trials. For example, alterations found in several receptor tyrosine kinases in the ERBB, NTRK, FGFR, and VEGFR families, which were discovered in these intraductal tumors, have available therapeutic inhibitors.5760  Similarly, PI3K and mTOR inhibitors may be of value in the tumors that reveal alterations in this pathway. Other potentially actionable targets found in our cohort include MAP kinases, SWI/SNF, MDM2, JAK/STAT, and DNA methyltransferases.

Finally, the groups based on our hierarchical clustering appear to have some similarities with the classification system proposed by the Japan-Korea Cooperative Study Group.61  Gastric-type, noninvasive tumoral intraductal neoplasms that are located within the intrahepatic bile ducts, which are likely to be classified as Type 1 by the Japan-Korea Classification, were significantly more common in our Group 1. In contrast, pancreatobiliary-type, tumoral intraductal neoplasms associated with invasive carcinoma that are located within the distal bile ducts, which are likely to be classified as Type 2, were significantly more common in our Group 2. However, none of our cases in Group 1 had grossly visible mucin in the lumen.

In summary, our comprehensive analysis confirms that tumoral intraductal neoplasms of the bile ducts, similar to their pancreatic counterparts, are diverse tumors, not only morphologically but also genetically. They consist of 5 distinct groups: pancreatobiliary-type IPN, gastric-type IPN, intestinal-type IPN, biliary IOPN, and biliary ITPN. Even the patients with invasive carcinoma appear to have a relatively protracted clinical course. The most common genetic alterations occur in the Wnt, DNA repair, MAP kinase, and PI3 kinase pathways, as well as many receptor tyrosine kinases. Some of these alterations are potentially targetable. Continued investigation of the clinical, histologic, and molecular features of these tumors will likely shed new light on better risk stratification and customized therapy to optimize patient care and outcomes.

The authors thank Marina Asher for her assistance with immunohistochemical stains and Bruce Crilly for his assistance with the figures.

1.
Katabi
N,
Torres
J,
Klimstra
DS.
Intraductal tubular neoplasms of the bile ducts
.
Am J Surg Pathol
.
2012
;
36
(11)
:
1647
1655
.
2.
Nakanuma
Y,
Basturk
O,
Esposito
I,
Klimstra
DS,
Komuta
M,
Zen
Y.
Intraductal papillary neoplasm of the bile ducts
. In:
WHO Classification of Tumours Editorial Board
, ed.
Digestive System Tumours
.
International Agency for Research on Cancer
;
2019
:
279
282
.
3.
Kim
WJ,
Hwang
S,
Lee
YJ,
et al
Clinicopathological features and long-term outcomes of intraductal papillary neoplasms of the intrahepatic bile duct
.
J Gastrointest Surg
.
2016
;
20
(7)
:
1368
1375
.
4.
Jung
G,
Park
KM,
Lee
SS,
Yu
E,
Hong
SM,
Kim
J.
Long-term clinical outcome of the surgically resected intraductal papillary neoplasm of the bile duct
.
J Hepatol
.
2012
;
57
(4)
:
787
793
.
5.
Schlitter
AM,
Jang
KT,
Klöppel
G,
et al
Intraductal tubulopapillary neoplasms of the bile ducts: clinicopathologic, immunohistochemical, and molecular analysis of 20 cases
.
Mod Pathol
.
2015
;
28
(9)
:
1249
1264
.
6.
Harada
F,
Matsuyama
R,
Mori
R,
et al
Outcomes of surgery for 2010 WHO classification-based intraductal papillary neoplasm of the bile duct: case-control study of a single Japanese institution’s experience with special attention to mucin expression patterns
.
Eur J Surg Oncol
.
2019
;
45
(5)
:
761
768
.
7.
Kim
JR,
Jang
KT,
Jang
JY,
et al
Clinicopathologic analysis of intraductal papillary neoplasm of bile duct: Korean multicenter cohort study
.
HPB (Oxford)
.
2020
;
22
(8)
:
1139
1148
.
8.
Youn
JM,
Hwang
S,
Ahn
CS,
et al
Clinicopathological features and long-term outcomes of intraductal papillary neoplasms of the bile duct of the liver: single-institution experience with 146 patients
.
J Gastrointest Surg
.
2022
;
26
(7)
:
1394
1405
.
9.
Matthaei
H,
Wu
J,
Dal Molin
M,
et al
GNAS codon 201 mutations are uncommon in intraductal papillary neoplasms of the bile duct
.
HPB (Oxford)
.
2012
;
14
(10)
:
677
683
.
10.
Rocha
FG,
Lee
H,
Katabi
N,
et al
Intraductal papillary neoplasm of the bile duct: a biliary equivalent to intraductal papillary mucinous neoplasm of the pancreas?
Hepatology
.
2012
;
56
(4)
:
1352
1360
.
11.
Fukumura
Y,
Nakanuma
Y,
Kakuda
Y,
Takase
M,
Yao
T.
Clinicopathological features of intraductal papillary neoplasms of the bile duct: a comparison with intraductal papillary mucinous neoplasm of the pancreas with reference to subtypes
.
Virchows Arch
.
2017
;
471
(1)
:
65
76
.
12.
Ozcan
K,
Klimstra
DS.
A review of mucinous cystic and intraductal neoplasms of the pancreatobiliary tract
.
Arch Pathol Lab Med
.
2022
;
146
(3)
:
298
311
.
13.
Adsay
V,
Mino-Kenudson
M,
Furukawa
T,
et al
Pathologic evaluation and reporting of intraductal papillary mucinous neoplasms of the pancreas and other tumoral intraepithelial neoplasms of pancreatobiliary tract: recommendations of Verona Consensus Meeting
.
Ann Surg
.
2016
;
263
(1)
:
162
177
.
14.
Askan
G,
Bagci
P,
Memis
B,
Basturk
O.
Intraductal neoplasms of the pancreas: an update
.
Turk Patoloji Derg
.
2017
;
33
(2)
:
87
102
.
15.
Adsay
NV,
Adair
CF,
Heffess
CS,
Klimstra
DS.
Intraductal oncocytic papillary neoplasms of the pancreas
.
Am J Surg Pathol
.
1996
;
20
(8)
:
980
994
.
16.
Basturk
O,
Tan
M,
Bhanot
U,
et al
The oncocytic subtype is genetically distinct from other pancreatic intraductal papillary mucinous neoplasm subtypes
.
Mod Pathol
.
2016
;
29
(9)
:
1058
1069
.
17.
Basturk
O,
Chung
SM,
Hruban
RH,
et al
Distinct pathways of pathogenesis of intraductal oncocytic papillary neoplasms and intraductal papillary mucinous neoplasms of the pancreas
.
Virchows Arch
.
2016
;
469
(5)
:
523
532
.
18.
Wang
T,
Askan
G,
Adsay
V,
et al
Intraductal oncocytic papillary neoplasms: clinical-pathologic characterization of 24 cases, with an emphasis on associated invasive carcinomas
.
Am J Surg Pathol
.
2019
;
43
(5)
:
656
661
.
19.
Vyas
M,
Hechtman
JF,
Zhang
Y,
et al
DNAJB1-PRKACA fusions occur in oncocytic pancreatic and biliary neoplasms and are not specific for fibrolamellar hepatocellular carcinoma
.
Mod Pathol
.
2020
;
33
(4)
:
648
656
.
20.
Basturk
O,
Berger
MF,
Yamaguchi
H,
et al
Pancreatic intraductal tubulopapillary neoplasm is genetically distinct from intraductal papillary mucinous neoplasm and ductal adenocarcinoma
.
Mod Pathol
.
2017
;
30
(12)
:
1760
1772
.
21.
Basturk
O,
Adsay
V,
Askan
G,
et al
Intraductal tubulopapillary neoplasm of the pancreas: a clinicopathologic and immunohistochemical analysis of 33 cases
.
Am J Surg Pathol
.
2017
;
41
(3)
:
313
325
.
22.
Chadwick
B,
Willmore-Payne
C,
Tripp
S,
Layfield
LJ,
Hirschowitz
S,
Holden
J.
Histologic, immunohistochemical, and molecular classification of 52 IPMNs of the pancreas
.
Appl Immunohistochem Mol Morphol
.
2009
;
17
(1)
:
31
39
.
23.
Furukawa
T,
Kuboki
Y,
Tanji
E,
et al
Whole-exome sequencing uncovers frequent GNAS mutations in intraductal papillary mucinous neoplasms of the pancreas
.
Sci Rep
.
2011
;
1
:
161
.
24.
Wu
J,
Jiao
Y,
Dal Molin
M,
et al
Whole-exome sequencing of neoplastic cysts of the pancreas reveals recurrent mutations in components of ubiquitin-dependent pathways
.
Proc Natl Acad Sci U S A
.
2011
;
108
(52)
:
21188
21193
.
25.
Sasaki
M,
Matsubara
T,
Nitta
T,
Sato
Y,
Nakanuma
Y.
GNAS and KRAS mutations are common in intraductal papillary neoplasms of the bile duct
.
PLoS One
.
2013
;
8
(12)
:
e81706
.
26.
Amato
E,
Molin
MD,
Mafficini
A,
et al
Targeted next-generation sequencing of cancer genes dissects the molecular profiles of intraductal papillary neoplasms of the pancreas
.
J Pathol
.
2014
;
233
(3)
:
217
227
.
27.
Tan
MC,
Basturk
O,
Brannon
AR,
et al
GNAS and KRAS mutations define separate progression pathways in intraductal papillary mucinous neoplasm-associated carcinoma
.
J Am Coll Surg
.
2015
;
220
(5)
:
845
854.e1
.
28.
Singhi
AD,
Wood
LD,
Parks
E,
et al
Recurrent rearrangements in PRKACA and PRKACB in intraductal oncocytic papillary neoplasms of the pancreas and bile duct
.
Gastroenterology
.
2020
;
158
(3)
:
573
582.e2
.
29.
Maimaitiaili
Y,
Fukumura
Y,
Hirabayashi
K,
et al
Investigation of -PRKACA/-PRKACB fusion genes in oncocytic tumors of the pancreatobiliary and other systems [published online September 24, 2022]
.
Virchows Arch
.
30.
Basturk
O,
Esposito
I,
Fukushima
N,
et al
Pancreatic intraductal tubulopapillary neoplasm
. In:
WHO Classification of Tumours Editorial Board
, ed.
Digestive System Tumours
.
International Agency for Research on Cancer
;
2019
:
317
318
.
31.
Basturk
O,
Esposito
I,
Fukushima
N,
et al
Pancreatic intraductal oncocytic papillary neoplasm
. In:
WHO Classification of Tumours Editorial Board
, ed.
Digestive System Tumours
.
International Agency for Research on Cancer
;
2019
:
315
316
.
32.
Fujikura
K,
Akita
M,
Ajiki
T,
Fukumoto
T,
Itoh
T,
Zen
Y.
Recurrent mutations in APC and CTNNB1 and activated Wnt/β-catenin signaling in intraductal papillary neoplasms of the bile duct: a whole exome sequencing study
.
Am J Surg Pathol
.
2018
;
42
(12)
:
1674
1685
.
33.
Yang
CY,
Huang
WJ,
Tsai
JH,
et al
Targeted next-generation sequencing identifies distinct clinicopathologic and molecular entities of intraductal papillary neoplasms of the bile duct
.
Mod Pathol
.
2019
;
32
(11)
:
1637
1645
.
34.
Goeppert
B,
Stichel
D,
Toth
R,
et al
Integrative analysis reveals early and distinct genetic and epigenetic changes in intraductal papillary and tubulopapillary cholangiocarcinogenesis
.
Gut
.
2022
;
71
(2)
:
391
401
.
35.
Basturk
O,
Aishima
S,
Esposito
I.
Biliary intraepithelial neoplasia
. In:
WHO Classification of Tumours Editorial Board
, ed.
Digestive System Tumours
.
International Agency for Research on Cancer
;
2019
:
273
275
.
36.
Basturk
O,
Esposito
I,
Fukushima
N,
et al
Pancreatic intraductal papillary mucinous neoplasm
. In:
WHO Classification of Tumours Editorial Board
, ed.
Digestive System Tumours
.
International Agency for Research on Cancer
;
2019
:
310
314
.
37.
DePristo
MA,
Banks
E,
Poplin
R,
et al
A framework for variation discovery and genotyping using next-generation DNA sequencing data
.
Nat Genet
.
2011
;
43
(5)
:
491
498
.
38.
Cibulskis
K,
Lawrence
MS,
Carter
SL,
et al
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
.
2013
;
31
(3)
:
213
219
.
39.
Robinson
JT,
Thorvaldsdóttir
H,
Winckler
W,
et al
Integrative genomics viewer
.
Nat Biotechnol
.
2011
;
29
(1)
:
24
26
.
40.
Benayed
R,
Offin
M,
Mullaney
K,
et al
High yield of RNA sequencing for targetable kinase fusions in lung adenocarcinomas with no mitogenic driver alteration detected by DNA sequencing and low tumor mutation burden
.
Clin Cancer Res
.
2019
;
25
(15)
:
4712
4722
.
41.
Zehir
A,
Benayed
R,
Shah
RH,
et al
Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients
.
Nat Med
.
2017
;
23
(6)
:
703
713
.
42.
Zheng
Z,
Liebers
M,
Zhelyazkova
B,
et al
Anchored multiplex PCR for targeted next-generation sequencing
.
Nat Med
.
2014
;
20
(12)
:
1479
1484
.
43.
Shibahara
H,
Tamada
S,
Goto
M,
et al
Pathologic features of mucin-producing bile duct tumors: two histopathologic categories as counterparts of pancreatic intraductal papillary-mucinous neoplasms
.
Am J Surg Pathol
.
2004
;
28
(3)
:
327
338
.
44.
Tsai
JH,
Yuan
RH,
Chen
YL,
Liau
JY,
Jeng
YM.
GNAS Is frequently mutated in a specific subgroup of intraductal papillary neoplasms of the bile duct
.
Am J Surg Pathol
.
2013
;
37
(12)
:
1862
1870
.
45.
Tsai
JH,
Liau
JY,
Yuan
CT,
Cheng
ML,
Yuan
RH,
Jeng
YM.
RNF43 mutation frequently occurs with GNAS mutation and mucin hypersecretion in intraductal papillary neoplasms of the bile duct
.
Histopathology
.
2017
;
70
(5)
:
756
765
.
46.
Kim
JR,
Lee
KB,
Kwon
W,
Kim
E,
Kim
SW,
Jang
JY.
Comparison of the clinicopathologic characteristics of intraductal papillary neoplasm of the bile duct according to morphological and anatomical classifications
.
J Korean Med Sci
.
2018
;
33
(42)
:
e266
.
47.
Kim
J,
Jang
KT,
Mo Park
S,
et al
Prognostic relevance of pathologic subtypes and minimal invasion in intraductal papillary mucinous neoplasms of the pancreas
.
Tumour Biol
.
2011
;
32
(3)
:
535
542
.
48.
Rong
Y,
Wang
D,
Xu
C,
et al
Prognostic value of histological subtype in intraductal papillary mucinous neoplasm of the pancreas: a retrospective analysis of outcome from one single center
.
Medicine (Baltimore)
.
2017
;
96
(15)
:
e6599
.
49.
Reid
M,
Losada
H,
Muraki
T,
et al
Field risk (“field-effect”/“field-defect”) in the gallbladder and biliary tree: an under-recognized phenomenon with major implications for management and carcinogenesis
.
Mod Pathol
.
2019
;
32
(S2)
:
1714A
.
50.
Uemura
S,
Higuchi
R,
Yazawa
T,
et al
Prognostic factors for surgically resected intraductal papillary neoplasm of the bile duct: a retrospective cohort study
.
Ann Surg Oncol
.
2021
;
28
(2)
:
826
834
.
51.
Schlitter
AM,
Born
D,
Bettstetter
M,
et al
Intraductal papillary neoplasms of the bile duct: stepwise progression to carcinoma involves common molecular pathways
.
Mod Pathol
.
2014
;
27
(1)
:
73
86
.
52.
Kong
L,
Liu
P,
Zheng
M,
et al
The miR-1224-5p/ELF3 axis regulates malignant behaviors of pancreatic cancer via PI3K/AKT/Notch signaling pathways
.
Onco Targets Ther
.
2020
;
13
:
3449
3466
.
53.
Yachida
S,
Wood
LD,
Suzuki
M,
et al
Genomic sequencing identifies ELF3 as a driver of ampullary carcinoma
.
Cancer Cell
.
2016
;
29
(2)
:
229
240
.
54.
Aoki
Y,
Mizuma
M,
Hata
T,
et al
Intraductal papillary neoplasms of the bile duct consist of two distinct types specifically associated with clinicopathological features and molecular phenotypes
.
J Pathol
.
2020
;
251
(1)
:
38
48
.
55.
Nakanuma
Y,
Kakuda
Y,
Fukumura
Y,
et al
The pathologic and genetic characteristics of the intestinal subtype of intraductal papillary neoplasms of the bile duct
.
Am J Surg Pathol
.
2019
;
43
(9)
:
1212
1220
.
56.
Gross
C,
Engleitner
T,
Lange
S,
et al
Whole exome sequencing of biliary tubulopapillary neoplasms reveals common mutations in chromatin remodeling genes
.
Cancers (Basel)
.
2021
;
13
(11)
:
2742
.
57.
Voss
JS,
Holtegaard
LM,
Kerr
SE,
et al
Molecular profiling of cholangiocarcinoma shows potential for targeted therapy treatment decisions
.
Hum Pathol
.
2013
;
44
(7)
:
1216
1222
.
58.
Borad
MJ,
Champion
MD,
Egan
JB,
et al
Integrated genomic characterization reveals novel, therapeutically relevant drug targets in FGFR and EGFR pathways in sporadic intrahepatic cholangiocarcinoma
.
PLoS Genet
.
2014
;
10
(2)
:
e1004135
.
59.
Chong
DQ,
Zhu
AX.
The landscape of targeted therapies for cholangiocarcinoma: current status and emerging targets
.
Oncotarget
.
2016
;
7
(29)
:
46750
46767
.
60.
Aitcheson
G,
Mahipal
A,
John
BV.
Targeting FGFR in intrahepatic cholangiocarcinoma [iCCA]: leading the way for precision medicine in biliary tract cancer [BTC]?
Expert Opin Investig Drugs
.
2021
;
30
(4)
:
463
477
.
61.
Nakanuma
Y,
Jang
KT,
Fukushima
N,
et al
A statement by the Japan-Korea expert pathologists for future clinicopathological and molecular analyses toward consensus building of intraductal papillary neoplasm of the bile duct through several opinions at the present stage
.
J Hepatobiliary Pancreat Sci
.
2018
;
25
(3)
:
181
187
.

Author notes

Supplemental digital content is available for this article at https://meridian.allenpress.com/aplm in the December 2023 table of contents.

This work has been supported by a gift from the Melamed Family Foundation, and by the Marie-Josée and Henry R. Kravis Center for Molecular Oncology, the Cancer Center Support Grant of the National Institutes of Health/National Cancer Institute under award number P30CA008748.

The authors have no relevant financial interest in the products or companies described in this article.

This study was presented in part at the annual meeting of the United States and Canadian Academy of Pathology; March 2018; Vancouver, British Columbia, Canada.

Wang is currently located in the Department of Pathology and Molecular Medicine, Queen’s University, Kingston, Ontario, Canada. Askan is currently located in the Department of Pathology, Istanbul University-Cerrahpasa, Cerrahpasa Faculty of Medicine, Istanbul, Turkey. Rao is currently located in the Department of Pathology and Anatomical Sciences, University of Missouri-Columbia, Columbia. Klimstra is currently located at Paige.AI, New York, New York.

Supplementary data