Context.—

Cholangiocarcinoma (CCA) is a heterogeneous cancer of the bile duct, and its diagnosis is often challenging.

Objective.—

To provide insights into state-of-the-art approaches for the diagnosis of CCA.

Data Sources.—

Literature review via PubMed search and authors’ experiences.

Conclusions.—

CCA can be categorized as intrahepatic or extrahepatic. Intrahepatic CCA is further classified into small-duct–type and large-duct–type, whereas extrahepatic CCA is classified into distal and perihilar according to site of origin within the extrahepatic biliary tree. Tumor growth patterns include mass forming, periductal infiltrating, and intraductal tumors. The clinical diagnosis of CCA is challenging and usually occurs at an advanced tumor stage. Pathologic diagnosis is made difficult by tumor inaccessibility and challenges in distinguishing CCA from metastatic adenocarcinoma to the liver. Immunohistochemical stains can assist in differentiating CCA from other malignancies, such as hepatocellular carcinoma, but no distinctive CCA-specific immunohistochemical profile has been identified. Recent advances in next-generation sequencing–based high-throughput assays have identified distinct genomic profiles of CCA subtypes, including genomic alterations that are susceptible to targeted therapies or immune checkpoint inhibitors. Detailed histopathologic and molecular evaluations of CCA by pathologists are critical for correct diagnosis, subclassification, therapeutic decision-making, and prognostication. The first step toward achieving these goals is to acquire a detailed understanding of the histologic and genetic subtypes of this heterogeneous tumor group. Here, we review state-of-the-art approaches that should be applied to establish a diagnosis of CCA, including clinical presentation, histopathology, staging, and the practical use of genetic testing methodologies.

Cholangiocarcinoma (CCA) is a heterogeneous biliary tract cancer originating from cells lining bile ducts, including cholangiocytes and hepatic stem or progenitor cells.13  Based on their anatomic site of origin, CCAs are divided into intrahepatic CCA (iCCA), perihilar CCA (pCCA,) and distal CCA (dCCA).1,3  iCCA is further subclassified into small-duct–type and large-duct–type, whereas pCCA, also called Klatskin tumor, and dCCA are together referred to as extrahepatic CCA (eCCA) (Figure 1, A).1,4  CCA is usually asymptomatic in early disease stages and is most commonly diagnosed if compression of the extrahepatic bile duct by a tumor results in jaundice or from symptoms related to metastases.1,5  Most patients with iCCA in the Surveillance, Epidemiology, and End Results (SEER) database were diagnosed at an advanced tumor stage, with 20.1% of patients having stage III and 34.9% having stage IV disease at diagnosis, as defined by the American Joint Committee on Cancer (AJCC).6  The prognosis of CCA remains poor because of its high recurrence rate.1,4 

Figure 1

A, Anatomic classification of cholangiocarcinoma and common target genes altered in cholangiocarcinoma subtypes. B, Morphologic classification of cholangiocarcinoma based on tumor growth patterns. C, Gross images of tumor growth patterns. A and B modified from 4Kendall T, Verheij J, Gaudio E, et al. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int. 2019;39(suppl 1):7–18. Copyright © 2019, Liver international, reproduced with permission of John Wiley & Sons Ltd, and from 11Alsaleh M, Leftley Z, Barbera TA, et al. Cholangiocarcinoma: a guide for the nonspecialist. Int J Gen Med. 2019;12:13–23. Copyright © 2019. International Journal of General Medicine, which is licensed under CC BY 4.0 (https://creativecommons.org/licenses/by/4.0/). Abbreviations: CCA, cholangiocarcinoma; eCCA, extrahepatic cholangiocarcinoma; iCCA, intrahepatic cholangiocarcinoma.

Figure 1

A, Anatomic classification of cholangiocarcinoma and common target genes altered in cholangiocarcinoma subtypes. B, Morphologic classification of cholangiocarcinoma based on tumor growth patterns. C, Gross images of tumor growth patterns. A and B modified from 4Kendall T, Verheij J, Gaudio E, et al. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int. 2019;39(suppl 1):7–18. Copyright © 2019, Liver international, reproduced with permission of John Wiley & Sons Ltd, and from 11Alsaleh M, Leftley Z, Barbera TA, et al. Cholangiocarcinoma: a guide for the nonspecialist. Int J Gen Med. 2019;12:13–23. Copyright © 2019. International Journal of General Medicine, which is licensed under CC BY 4.0 (https://creativecommons.org/licenses/by/4.0/). Abbreviations: CCA, cholangiocarcinoma; eCCA, extrahepatic cholangiocarcinoma; iCCA, intrahepatic cholangiocarcinoma.

Close modal

The pathologic diagnosis of CCA is often challenging, particularly in poorly differentiated tumors with atypical radiologic or clinical features.3,712  Once a tumor has been sampled via a tissue core or fine-needle aspiration biopsy, pathologists must differentiate CCA from hepatocellular carcinoma (HCC) and metastatic adenocarcinoma to the liver or biliary tree, as well as from pancreatic ductal adenocarcinoma if the tumor arises in the vicinity of the intrapancreatic portion of the extrahepatic bile duct.3,4,13  An array of immunohistochemical markers and in situ hybridization tests are typically used in this endeavor.14  However, in this era of precision medicine, pathologists must be mindful to use stains judiciously, in a stepwise fashion to preserve tissue for biomarkers and genomic analysis.

Advances in high-throughput molecular analysis, including next-generation sequencing (NGS), have improved characterization of CCA with identification of distinct genomic profiles and targetable genetic alterations in almost half of patients with CCA.15,16  Patterns of genomic alterations and potential oncogenic drivers differ in iCCA and eCCA. For example, alterations in isocitrate dehydrogenase 1 (IDH1), BRCA1-associated protein–1 (ubiquitin carboxyl-terminal hydrolase; BAP1), and fibroblast growth factor receptor 2 (FGFR2) occur more commonly in iCCA, whereas alterations in Kirsten ras oncogene homolog (KRAS) and tumor protein p53 (TP53) occur more commonly in eCCA.1,15,1721  Furthermore, risk factors and etiologies that vary by geographic region (eg, liver flukes and hepatolithiasis in Asia) may be associated with different molecular landscapes in CCA.22,23  Selecting testing methodologies that allow for the identification of all potential oncogenic drivers of CCA will facilitate delineation of CCA subtypes, based on molecular alterations and specific genomic signatures.

Through histopathologic evaluation, exclusion of non-CCA tumors, and recommendations based on appropriate genomic testing, pathologists play a critical role in the multidisciplinary care team for patients with CCA.13  This review provides insights into state-of-the-art approaches for the diagnosis of CCA with respect to the clinical presentation, histopathology, staging, and the practical use of genetic testing methodologies, including their specificities for CCA subtypes.

Multiple risk factors and predisposing conditions are associated with the development of CCA, some shared by all CCA subtypes, and others specific to 1 subtype or geographic region.2,22  To date, these include bile duct cysts, primary sclerosing cholangitis (PSC), Caroli disease, cholelithiasis/choledocholithiasis, cirrhosis, viral hepatitis B and C, liver flukes (Clonorchis sinensis, Opisthorchis viverrini), and nonalcoholic fatty liver disease (NAFLD)/nonalcoholic steatohepatitis (NASH).2 

PSC is an autoimmune disease characterized by narrowing or obstruction of both intrahepatic and extrahepatic bile ducts.2,24  Bile duct obstruction leads to intracellular accumulation of toxic bile acids that contribute to hepatocyte and cholangiocyte damage and inflammation.25  Patients with PSC have an approximately 400-fold (standardized incidence rate, 398; 95% CI, 246–608)26  to 1500-fold (relative risk, 1560; 95% CI, 780–2793; P < .001)27  increased risk of developing CCA as compared with the general population. CCA may present within the first year of the diagnosis of PSC, with a reported range of presentation of 0.8 to 15.0 years.27,28 

Worldwide, choledochal cysts, choledocholithiasis, and cirrhosis (regardless of cause) were identified as the strongest risk factors for CCA in a recent meta-analysis study.29  Of these, choledochal cysts had the greatest contribution to the risk of both iCCA and eCCA, with pooled odds ratios (ORs) of 26.71 (95% CI, 15.80–45.16) and 34.94 (95% CI, 24.36–50.12), respectively.29  Cirrhosis has a greater contribution to the risk of iCCA than eCCA.29  Cholangitis and liver fluke infections are reported as the main risk factors for large-duct–type iCCA, and cirrhosis and viral hepatitis as major risk factors for small-duct–type iCCA.1,30 

NAFLD/NASH also have been shown to be risk factors for CCA and may contribute to the increasing incidence of CCA worldwide.31,32  A recent meta-analysis of 7 case-control studies showed a positive association between NAFLD and CCA (pooled OR, 1.95; 95% CI, 1.36–2.79).32  When classified according to CCA subtypes, NAFLD was a stronger risk factor for iCCA (OR, 2.22; 95% CI, 1.52–3.24) than for eCCA (OR, 1.55; 95% CI, 1.03–2.33).32  NASH was also reported as an independent risk factor for iCCA (OR, 3.36; 95% CI, 1.15–10.2).33 

Although the exact molecular mechanisms of tumor induction evoked in these disparate predisposing conditions remain to be resolved, it is speculated that tumorigenesis occurs in a proinflammatory background in the setting of biliary epithelial injury, chronic inflammation, and cytokine and growth factor upregulation, all of which contribute to DNA damage, with resulting mutations in cell cycle genes.3437  The current World Health Organization classification identifies 2 molecular subtypes of iCCA: the inflammation and proliferation subtypes.38  The inflammation subtype shows signal transducer and activator of transcription 3 (STAT3) activation with overexpression of cytokines, and generally is associated with a better prognosis.38  The proliferation subtype is more commonly associated with mutations in IDH, KRAS, B-Raf proto-oncogene serine/threonine-protein kinase (BRAF), and other oncogenic signaling pathways, as well as with chromosomal instability, and is associated with a worse prognosis.38 

The most common clinical presentation of eCCA is jaundice due to bile duct obstruction; however, jaundice is an initial symptom in only 10% to 15% of patients with iCCA.3,39  Presenting symptoms for iCCA are often not site-specific and include abdominal pain, night sweats, asthenia, nausea, and weight loss.39  Altered liver function test results are commonly found at the time of diagnosis in patients with CCA, but are not specific or indicative of malignancy.11,39  Nevertheless, elevated total and direct bilirubin, alkaline phosphatase, gamma-glutamyl transferase, and 5′-nucleotidase suggest the presence of biliary obstruction.40  Reduced albumin and elevated alkaline phosphatase, total bilirubin, direct bilirubin, and gamma-glutamyl transpeptidase have been correlated with shorter overall survival in patients with iCCA.41 

Radiologically, it is challenging to distinguish iCCA from pCCA because of the propensity of pCCA tumors to involve both the large ducts at the liver hilum and the perihilar parenchyma.3,42,43  Moreover, tumors within the biliary tract are usually difficult to access for sampling, leading to insufficient or inadequate tumor tissue for diagnosis and biomarker analysis.3,44 

Gross Features

CCA arises from transformed epithelial cells including hepatic or biliary tree progenitor cells, mature cholangiocytes, and hepatocytes as potential cells of origin.45  The pCCA variant of eCCA is localized to one or both hepatic ducts and/or the junction between the left and right ducts, whereas the dCCA variant is localized in the common bile duct (Figure 1, A).1,4  Both eCCA subtypes generally exhibit a periductal growth pattern that presents as flat or poorly defined nodular sclerosing tumor with diffuse infiltration into adjacent structures, and less frequently show an intraductal growth pattern (intraductal papillary tumor).4,46  iCCA may occur anywhere from the periphery to central and hilar segments of the liver, arising from virtually any ductal structure from large segmental bile ducts to smaller branches of the intrahepatic biliary tree (Figure 1, A).1,4  Three gross iCCA tumor growth patterns are reported: mass forming, periductal infiltrating, and intraductal growing (Figure 1, B and C).1,4  Mass forming is the most common growth pattern in iCCA, present in approximately 60% of patients.47  Small-duct iCCA generally shows a mass-forming growth pattern, whereas large-duct–type iCCA usually shows periductal growth but may also occur as an intraductal tumor.4 

Microscopic Features

The morphology of CCA includes the spectrum of extremely well-differentiated gland-forming tumors to poorly differentiated adenocarcinoma, all of which tend to demonstrate a desmoplastic stromal response and frequent perineural invasion.3,46  Cuboidal or low columnar cells are typical in CCA; however, mucinous, clear cell, micropapillary, and other variants occur rarely.3  Differentiating iCCA from benign glandular proliferations and dCCA from pancreatic ductal adenocarcinoma may be challenging (see Diagnosis and Differential Diagnosis).3,39,48 

The World Health Organization classification of digestive system tumors (5th edition) separates iCCA into 2 main subtypes: a small-duct type, which shares many etiologic, pathogenetic, and imaging characteristics with HCC, and a large-duct type resembling eCCA.49 

Small-duct iCCAs, originating from interlobular and septal bile ducts lined by cuboidal cholangiocytes,4  manifest histologically as adenocarcinomas composed of small tubular or acinar structures with nodular growth invading the liver parenchyma and with no or minimal mucin production (Figure 2).1,45  Large-duct iCCA, originating from tall cylindrical cholangiocytes, often shows papillary architecture with mucin-producing columnar tumor cells.1,4  Large-duct iCCA is commonly preceded by precancerous lesions such as biliary intraepithelial neoplasia or intraductal papillary neoplasia (Figure 2).45 

Figure 2

Representative hematoxylin-eosin–stained histology and respective immunohistochemical staining profiles of CCA subtypes (original magnification ×100). aBased on author experience. Abbreviations: CCA, cholangiocarcinoma; CEA, carcinoembryonic antigen; CK, cytokeratin; CRP, C-reactive protein; dCCA, distal cholangiocarcinoma; eCCA, extrahepatic cholangiocarcinoma; EMA, epithelial membrane antigen; HepPar-1, hepatocyte paraffin 1; HNF-1β, hepatocyte nuclear factor-1beta; iCCA, intrahepatic cholangiocarcinoma; IHC, immunohistochemical; IMP3, insulin-like growth factor-I mRNA binding protein-3; MOC31, anti–epithelial cell adhesion molecule antibody; MUC, mucin; NCAM, neural cell adhesion molecule; pCCA, perihilar CCA; pVHL, von Hippel–Lindau protein; TFF1, trefoil factor 1; TUBB3, tubulin beta-III.

Figure 2

Representative hematoxylin-eosin–stained histology and respective immunohistochemical staining profiles of CCA subtypes (original magnification ×100). aBased on author experience. Abbreviations: CCA, cholangiocarcinoma; CEA, carcinoembryonic antigen; CK, cytokeratin; CRP, C-reactive protein; dCCA, distal cholangiocarcinoma; eCCA, extrahepatic cholangiocarcinoma; EMA, epithelial membrane antigen; HepPar-1, hepatocyte paraffin 1; HNF-1β, hepatocyte nuclear factor-1beta; iCCA, intrahepatic cholangiocarcinoma; IHC, immunohistochemical; IMP3, insulin-like growth factor-I mRNA binding protein-3; MOC31, anti–epithelial cell adhesion molecule antibody; MUC, mucin; NCAM, neural cell adhesion molecule; pCCA, perihilar CCA; pVHL, von Hippel–Lindau protein; TFF1, trefoil factor 1; TUBB3, tubulin beta-III.

Close modal

A novel “solid tubulocystic variant” of iCCA has been described recently (previously referred to as the cholangioblastic variant of iCCA).50,51  This variant is an inhibin-positive carcinoma with most cases reported in younger women (17–44 years of age).50  These rare tumors have distinctive solid, tubular, and cystic growth patterns, with tubules, cysts, and solid/nested pattern interrupted by lumina. Morphologically, these resemble thyroid follicular neoplasms and granulosa cell tumors.50  The cytology is biphasic, with small basophilic cells and large, polygonal eosinophilic cells.51  The pattern of spread and morphologic features differs from iCCA; however, based on its primary hepatic location, strong albumin in situ hybridization staining, and glandular differentiation, it is designated as a variant of CCA.50  Recently, a novel gene fusion, nipped-B–like protein-nucleus accumbens-associated protein 1 (NIPBL-NACC1), was reported in 4 patients with tumors that are morphologically similar to solid tubulocystic iCCA.51 

The use of a consistent staging system is important for all tumors to predict prognosis, develop treatment plans, and evaluate therapeutics. The AJCC TNM classification defines cancer stages according to the extension of primary tumor (T), regional lymph node infiltration (N), and the presence of distant metastases (M).52  The 8th edition of the AJCC staging manual separates staging criteria for CCA subgroups (iCCA, pCCA, and dCCA) and contains significant updates from prior editions.52  In this review, updated criteria for T-stage are discussed below. As for all solid tumors using AJCC staging criteria, final stage designations in CCA encompass lymph node and distal metastasis status.47  The reader is referred to the 8th edition AJCC Cancer Staging Manual for details.53 

According to the AJCC 8th edition iCCA criteria, the T1 category comprises solitary tumors without vascular invasion and is divided into 2 subcategories: 1a (≤5 cm) and 1b (>5 cm), to account for the prognostic impact of tumor size.47,52  The T2 category encompasses solitary tumors with vascular invasion or multiple tumors with or without vascular invasion.47,52  Tumors perforating the visceral peritoneum are T3 category and tumors directly invading local extrahepatic structure are T4.47,52 

Considering pCCA T-stage criteria, high-grade biliary intraepithelial neoplasia is classified as carcinoma in situ (Tis). The T1 category applies to tumors confined to the bile duct, with extension up to the fibrous tissue or muscle layer.52,54  T2 tumors invade beyond the wall of the bile duct to surrounding adipose tissue (T2a) or invade adjacent hepatic parenchyma (T2b). T3 tumors invade unilateral branches of the hepatic artery or portal vein.52,54  Tumors meeting criteria for T4 invade the common hepatic artery, or the main portal vein or its branches bilaterally, or unilateral second-order biliary radicals with contralateral portal vein or hepatic artery involvement.52,54 

T-stage criteria for dCCA distinguish T1, T2, and T3 category tumors by the depth of invasion into the bile duct wall (<5 mm, 5–12 mm, and >12 mm, respectively).52,55  The T4 category applies to tumors involving the celiac axis, superior mesenteric artery, and/or common hepatic artery.52,55 

The classic morphology and immunohistochemical staining profiles for CCA subtypes are shown in Figure 2. Cytokeratin (CK) stains of various molecular weights (eg, CK7, CK19, and CK20) and other markers usually expressed in adenocarcinomas of gastrointestinal and pancreatobiliary origin (eg, BerEP4, cytoplasmic epithelial membrane antigen, carcinoembryonic antigen, and CA19-9) are reliably positive in most CCAs, although entirely nonspecific.5658  Compared with small-duct–type iCCA, the large-duct–type iCCA is more likely to be positive for CA19-9 (57.1% versus 8.4%) and exhibit the CK7+, CK19+, CD56 phenotype (71.4% versus 26.5%).59  The immunohistochemistry profile for eCCA also includes keratins, mucins, and tumor-associated epithelial markers (Figure 2).60  However, because of the possibility of false positives or false negatives associated with these markers, a diagnosis of CCA should not be made on the basis of the presence or absence of immunohistochemical markers alone.61 

Distinguishing CCA from metastatic adenocarcinoma to the hepatobiliary tree or from benign biliary lesions, such as bile duct adenomas and reactive ductular proliferations, can be challenging, largely because of similarities in clinical presentation, overlapping histopathologic features, and the paucity of iCCA-specific tissue markers.4,14,43,58,61  For these reasons, iCCA is frequently a diagnosis of exclusion.43  Immunohistochemical and in situ hybridization markers commonly used to differentiate iCCA and metastatic adenocarcinoma are shown in Table 1.

Table 1

Common Immunohistochemical Markers for Differentiating Intrahepatic and Potential Metastatic Carcinomas to the Liver Based on Their Cellular Origina

Common Immunohistochemical Markers for Differentiating Intrahepatic and Potential Metastatic Carcinomas to the Liver Based on Their Cellular Origina
Common Immunohistochemical Markers for Differentiating Intrahepatic and Potential Metastatic Carcinomas to the Liver Based on Their Cellular Origina

Differentiating CCA from HCC is usually accomplished on hematoxylin-eosin sections. In histologically challenging samples, CCA may be differentiated from HCC by the absence of hepatocytic markers including hepatocyte paraffin 1 antigen and arginase 1, and by the absence of a canalicular staining pattern with CD10 or polyclonal carcinoembryonic antigen stains (Figure 2).3,14,58,62  Hepatocyte nuclear factor-1β (HNF-1β), positive in virtually all iCCAs as well as other adenocarcinomas (pancreatic, gallbladder, and ovarian carcinomas), is positive in only 9.2% of HCC tumors.63,64  Conversely, a lack of HNF-1β expression may be used to exclude iCCA in the setting of adenocarcinomas of unknown primary in the liver.63  Albumin mRNA, detected by in situ hybridization, is a marker of hepatocellular differentiation and is positive in most iCCAs; therefore, it cannot be used to distinguish iCCA from HCC.12,65  Characterization of albumin expression across multiple tumor types reveals positive albumin in situ hybridization in 100% of HCCs, 81% of iCCAs, 39% of gallbladder cancer, and 20% of lung cancer samples.65  Expression of albumin mRNA is also reported in hepatoid pancreatic adenocarcinoma (33%) and acinar cell carcinoma (29%).65  Of note, most eCCAs are negative for albumin.12  In situ hybridization for α-fetoprotein can be useful to distinguish between CCA (low sensitivity and specificity) and HCC (high specificity).55,66  In addition, MOC31 (anti–epithelial cellular adhesion molecule antibody) is positive in most cases of CCA, while typically negative in HCC.12  However, MOC31 may be positive in 10% to 20% of cases of poorly differentiated HCC.12 

In the era of precision medicine, when confronted with a liver tumor biopsy, the key factor in the approach to diagnosis is to select the minimal number of stains necessary to render the diagnosis, working in a stepwise fashion that takes the clinical history and radiologic findings into account in what can be termed a personalized approach to diagnosis.67 

Serum biomarkers specific for CCA have not yet been conclusively identified.18  CA19-9, although not specific for CCA, has been used to support a CCA diagnosis in the proper clinical setting, with levels greater than 1000 U/mL associated with the presence of unresectable disease.18,68  However, sensitivity for detecting disease is low, with CA19-9 elevations found in only 50% of patients with iCCA in some studies.68,69  A recent study reported that doublecortin-like kinase 1 (DCLK1) was present in higher concentration in serum samples from patients with iCCA or pCCA than in serum samples from patients with HCC, PSC, or cirrhosis (P < .05), and was undetectable in samples from healthy individuals.70  Although confirmatory research is needed, this suggests DCLK1 is a potential serum biomarker for the early CCA diagnosis.

Comprehensive gene profiling studies based on NGS methodologies are defining the genomic and transcriptomic landscape in CCA and may guide the selection and optimal use of targeted therapies.15,71  Advancements in NGS technology have led to the development of numerous molecular diagnostic assays profiling large gene panels that further confirm histopathologic diagnosis within a short time frame (Table 2).71,72  These assays detect a wide spectrum of genomic alterations including mutations, amplifications, substitutions, deletions, insertions, copy number alterations, fusions or other rearrangements, and other genomic signatures such as tumor mutational burden and microsatellite instability.72 

Table 2

Next-Generation Sequencing–Based Genomic Profiling Assaysa

Next-Generation Sequencing–Based Genomic Profiling Assaysa
Next-Generation Sequencing–Based Genomic Profiling Assaysa

Commonly altered genes in iCCA are IDH1, isocitrate dehydrogenase 2 (IDH2), FGFR2, BAP1, BRAF, AT-rich interactive domain-containing protein 1A (ARID1A), TP53, SMAD family member 4 (SMAD4), KRAS, neuroblastoma RAS viral oncogene homolog (NRAS), mouse double minute 2 homolog (E3 ubiquitin-protein ligase Mdm2; MDM2), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA), protein polybromo-1 (PBRM1), and erythroblastic oncogene B b2 receptor tyrosine kinase 2 (ERBB2), whereas the commonly altered genes in eCCA are KRAS, TP53, SMAD4, ARID1A, and ERBB2 (Figure 1, A).1,4,15,1720,7379  Alterations in BAP1 and FGFR2 have been reported to be almost exclusively in iCCA.15,17  Notably, large-duct–type iCCA demonstrates a high mutation frequency of KRAS, TP53, SMAD4, and MDM2, whereas IDH1/2 mutations, FGFR2 fusions, and BAP1 loss are more frequently observed in small-duct–type iCCA (Figure 1, A).1,4,20,7478 IDH1 and IDH2 mutations are associated with poor prognosis, whereas FGFR2 fusions are associated with better prognosis in small-duct–type iCCA.3 

The specific types of alterations that are targeted for FGFR2 include single-nucleotide variants, gene fusions or other rearrangements or translocations, and copy number amplifications.80,81  These alterations can lead to dimerization, kinase activation, increased ligand-binding affinity, or altered cellular localization, and in turn, upregulated and/or aberrant FGFR signaling and subsequently increased tumor growth and survival.80,81  Resistance to FGFR2 targeted therapy can be primary (lack of initial response to the treatment) or secondary. Secondary resistance can occur through occurrence of additional alterations at specific “gatekeeper” residues in the FGFR protein, hyperactivation of a non-FGFR receptor tyrosine kinase “bypass” signaling pathway, and gene fusion.80,82  A study of 1863 CCA cases using NGS revealed a high prevalence of ERBB family genetic alterations in iCCA (7.6%) and eCCA (15.3%).83  In patients with iCCA and eCCA, amplifications in ERBB2 (HER2) were observed in 2.8% and 4.0% of the cases, and ERBB3 (HER3) amplifications were observed in 0.5% and 0.8%, respectively.83 PIK3CA mutations lead to dysregulation of the PI3K/AKT pathway and have been reported in 6% to 8% of CCA tumors.84,85  Mutation frequencies of PIK3CA in pCCA were reported to be similar to those in iCCA, but notably higher than in dCCA.86 

Subclasses of CCAs have distinct multilevel molecular characteristics. The Cancer Genome Atlas consortium identified a distinct subtype of CCA (predominantly intrahepatic and fluke- and hepatitis-negative) enriched for IDH mutants with distinct molecular features including low chromatin modifier and high mitochondrial gene expression, increased mitochondrial DNA copy number, and potential epigenetic silencing of ARID1A.87 

Recently, multiplatform molecular characterization of eCCA in a cohort of 189 patients identified 4 novel transcriptome-based molecular classes and identified 25% of tumors with actionable genomic alterations having potential prognostic and therapeutic implications.46  Among these molecular classes, the proliferation class is suggested as a strong candidate for ERBB2 inhibitor treatment. Programmed death receptor-1 (PD-1)/programmed death ligand-1 (PD-L1) blockade is recommended for the immune class.46  According to the National Comprehensive Cancer Network guidelines on biliary tract cancers, molecular testing should be considered before initiation of primary therapy in patients with CCA with deficient mismatch repair/microsatellite instability-high tumors or a family history of breast cancer gene 1/2 (BRCA1/2) mutations, and may include tumor mutational burden and neurotrophic tyrosine receptor kinase (NTRK) fusion analysis.19 

In addition to identifying potential therapeutic targets, genomic testing is essential to understand treatment responses to chemotherapy and immunotherapy.16,88,89  A study using targeted sequencing and immunohistochemical staining to identify biomarkers associated with treatment response in biliary tract cancers found that responses to chemotherapy were more favorable in small-duct–type iCCA with mutations in BAP1 and IDH1 than in large-duct–type iCCA with KRAS and SMAD4 mutations.89  Significantly longer progression-free survival (6.4 versus 3.2 months; hazard ratio, 3.70; 95% CI, 1.72–8.33; P < .001) and overall survival (21.9 versus 13.9 months; hazard ratio, 2.22; 95% CI, 1.03–4.76; P = .037) were reported in small-duct–type versus large-duct–type iCCA.89  Tumors with alterations in DNA damage response pathway genes (BAP1, ATM serine/threonine kinase [ATM], IDH1/2) had a higher overall response rate than those with alterations in transforming growth factor-β pathway genes (SMAD4, transforming growth factor beta receptor 2 [TGFBR2], F-box and WD repeat domain-containing 7 [FBXW7], MYC proto-oncogene, BHLH transcription factor [MYC]; 50.0% versus 9.5%, P = .015).89  These results suggest that the distinct molecular alterations in CCA subtypes may help predict response to chemotherapy.89 

Molecular profiling in patients treated with anti–PD-1/PD-L1 antibodies identified KRAS alteration and chromosomal instability in tumors as independent treatment-resistant factors.89  Patients having at least 1 of these resistance factors had shorter median progression-free survival with rapid tumor growth and poor clinical benefit to immunotherapy than those with no resistance factors. A high density of tumor-infiltrating lymphocytes in the tumor center is associated with favorable response to immunotherapy in patients with CCA.89  Of note, CCA with either KRAS alteration or chromosomal instability showed low density of tumor-infiltrating lymphocytes in the tumor center, indicating immune-suppressive tumor microenvironments in these tumors.89 

Targeted therapies that are approved by the US Food and Drug Administration or are in development for the treatment of advanced CCA include fibroblast growth factor receptor inhibitors (pemigatinib,90  infigratinib91 [distribution of infigratinib will be discontinued March 2023],92  and futibatinib93), isocitrate dehydrogenase inhibitors (ivosidenib and dasatinib),94  and NTRK inhibitors (entrectinib and larotrectinib)95  (Table 3).

Table 3

Current Targeted Therapies for Cholangiocarcinoma (CCA)

Current Targeted Therapies for Cholangiocarcinoma (CCA)
Current Targeted Therapies for Cholangiocarcinoma (CCA)

In a retrospective review of treatment outcomes in patients receiving matched therapy for actionable molecular alterations based on the European Society for Medical Oncology Scale for Clinical Actionability of Molecular Target, median overall survival was significantly improved in patients with CCA receiving matched therapy (22.6 months), compared with patients without actionable alterations (14.3 months).96  Actionable alterations included mutations in IDH, FGFR2, and BRAF genes.96 

NGS-based assays can be performed by using tumor tissue samples or whole blood as sources for nucleic acids.72  Liquid biopsy profiling (eg, circulating tumor cells, circulating tumor DNA, RNAs or exosomes from peripheral blood samples) has also been suggested as an alternative to traditional tissue biopsies, since blood sampling is a minimally invasive procedure that can be easily repeated to examine the change in tumor-specific diagnostic, prognostic, or predictive biomarkers over time.97,98  In a recent study on circulating tumor DNA screening in patients with advanced gastric cancer, FGFR2 amplifications were more frequently detected by circulating tumor DNA sequencing (7.7%) than by tissue analysis alone (2.6%–4.4%).99  Despite these advantages, a lack of standardized methods for collection and analysis of liquid biopsy samples has led to variability and lack of reproducibility across studies,97  such that liquid biopsy is not currently used in standard clinical practice.98 

As genomic alterations differ between iCCA and eCCA, and between the histologic subtypes within iCCA,79  genomic screening tests may complement and provide further confirmation of the histopathologic diagnosis. Molecular characterization of CCAs using NGS-based assays can help identify CCA subtypes and shed light on tumor prognosis and treatment response.18,79  Targeted therapies have become an important part of the treatment of CCA based on genomic findings,18,79  supporting the practice of performing genetic testing early in the care of the patient, to identify patients who may benefit from these therapies.18,79  A proposed diagnostic algorithm from clinical presentation to therapeutic intervention in patients with CCA is provided in Figure 3.

Figure 3

Proposed diagnostic algorithm from clinical presentation to therapeutic intervention in patients with cholangiocarcinoma. Abbreviations: BRAF, B-Raf proto-oncogene serine/threonine-protein kinase; CCA, cholangiocarcinoma; dMMR, deficient mismatch repair; eCCA, extrahepatic cholangiocarcinoma; ERBB2, Erb-B2 receptor tyrosine kinase 2; FGFR, fibroblast growth factor receptor; HCC, hepatocellular carcinoma; HER2, human epidermal growth factor receptor 2; iCCA, intrahepatic cholangiocarcinoma; IDH, isocitrate dehydrogenase; NTRK, neurotrophic tyrosine receptor kinase; PD-1, programmed death receptor-1; PD-L1, programmed death ligand-1; TMB, tumor mutational burden.

Figure 3

Proposed diagnostic algorithm from clinical presentation to therapeutic intervention in patients with cholangiocarcinoma. Abbreviations: BRAF, B-Raf proto-oncogene serine/threonine-protein kinase; CCA, cholangiocarcinoma; dMMR, deficient mismatch repair; eCCA, extrahepatic cholangiocarcinoma; ERBB2, Erb-B2 receptor tyrosine kinase 2; FGFR, fibroblast growth factor receptor; HCC, hepatocellular carcinoma; HER2, human epidermal growth factor receptor 2; iCCA, intrahepatic cholangiocarcinoma; IDH, isocitrate dehydrogenase; NTRK, neurotrophic tyrosine receptor kinase; PD-1, programmed death receptor-1; PD-L1, programmed death ligand-1; TMB, tumor mutational burden.

Close modal

CCA is a group of clinically, pathologically, and genetically heterogeneous tumors that are increasing in incidence and are associated with limited survival because of frequent detection in late disease stage. Correct diagnosis of CCA requires high clinical suspicion and the astute application of histopathology and immunohistochemical/in situ hybridization testing that allows the distinction of CCA from other tumor types. Advances in knowledge of the CCA mutational landscape now place this tumor squarely within the growing group of neoplasms for which patient outcome may be improved by routine molecular profiling. Since almost 50% of patients with CCA have potentially targetable genomic alterations,15,16  tumor genomic analysis should be considered in patients with CCA when feasible. A collaborative approach between oncologists and pathologists will optimize best practices in the use of tumor samples for patient care.

Medical writing assistance was provided by Ciara Duffy, PhD, CMPP (Evidence Scientific Solutions, Sydney, Australia), and Madeeha Aqil, PhD, MWC (Envision Pharma Group, Philadelphia, Pennsylvania), funded by Incyte Corporation.

1.
Banales
JM,
Marin
JJG,
Lamarca
A,
et al
Cholangiocarcinoma 2020: the next horizon in mechanisms and management
.
Nat Rev Gastroenterol Hepatol
.
2020
;
17
(9)
:
557
588
.
2.
Khan
SA,
Tavolari
S,
Brandi
G.
Cholangiocarcinoma: epidemiology and risk factors
.
Liver Int
.
2019
;
39
(suppl 1)
:
19
31
.
3.
Sarcognato
S,
Sacchi
D,
Fassan
M,
et al
Cholangiocarcinoma
.
Pathologica
.
2021
;
113
(3)
:
158
169
.
4.
Kendall
T,
Verheij
J,
Gaudio
E,
et al
Anatomical, histomorphological and molecular classification of cholangiocarcinoma
.
Liver Int
.
2019
;
39
(suppl 1)
:
7
18
.
5.
Huguet
JM,
Lobo
M,
Labrador
JM,
et al
Diagnostic-therapeutic management of bile duct cancer
.
World J Clin Cases
.
2019
;
7
(14)
:
1732
1752
.
6.
Meng
ZW,
Pan
W,
Hong
HJ,
Chen
JZ,
Chen
YL.
Macroscopic types of intrahepatic cholangiocarcinoma and the eighth edition of AJCC/UICC TNM staging system
.
Oncotarget
.
2017
;
8
(60)
:
101165
101174
.
7.
Esnaola
NF,
Meyer
JE,
Karachristos
A,
Maranki
JL,
Camp
ER,
Denlinger
CS.
Evaluation and management of intrahepatic and extrahepatic cholangiocarcinoma
.
Cancer
.
2016
;
122
(9)
:
1349
1369
.
8.
Joo
I,
Lee
JM,
Yoon
JH.
Imaging diagnosis of intrahepatic and perihilar cholangiocarcinoma: recent advances and challenges
.
Radiology
.
2018
;
288
(1)
:
7
13
.
9.
Kawamura
R,
Harada
Y,
Shimizu
T.
Missed diagnosis of cholangiocarcinoma presenting with atypical symptoms
.
Eur J Case Rep Intern Med
.
2021
;
8
(1)
:
002207
.
10.
Tasch
JJ,
Dube
N.
An unusual presentation of advanced intrahepatic cholangiocarcinoma: when biopsy results fail
.
Am J Case Rep
.
2018
;
19
:
35
40
.
11.
Alsaleh
M,
Leftley
Z,
Barbera
TA,
et al
Cholangiocarcinoma: a guide for the nonspecialist
.
Int J Gen Med
.
2019
;
12
:
13
23
.
12.
Balitzer
D,
Kakar
S.
Challenges in diagnosis of hepatocellular carcinoma in cirrhotic liver: a pathologist’s perspective
.
Clin Liver Dis (Hoboken)
.
2021
;
17
(4)
:
249
254
.
13.
Cho
MT,
Gholami
S,
Gui
D,
et al
Optimizing the diagnosis and biomarker testing for patients with intrahepatic cholangiocarcinoma: a multidisciplinary approach
.
Cancers (Basel)
.
2022
;
14
(2)
:
392
.
14.
Wang
HL,
Kim
CJ,
Koo
J,
et al
Practical immunohistochemistry in neoplastic pathology of the gastrointestinal tract, liver, biliary tract, and pancreas
.
Arch Pathol Lab Med
.
2017
;
141
(9)
:
1155
1180
.
15.
Lowery
MA,
Ptashkin
R,
Jordan
E,
et al
Comprehensive molecular profiling of intrahepatic and extrahepatic cholangiocarcinomas: potential targets for intervention
.
Clin Cancer Res
.
2018
;
24
(17)
:
4154
4161
.
16.
Silverman
IM,
Hollebecque
A,
Friboulet
L,
et al
Clinicogenomic analysis of FGFR2-rearranged cholangiocarcinoma identifies correlates of response and mechanisms of resistance to pemigatinib
.
Cancer Discov
.
2021
;
11
(2)
:
326
339
.
17.
Nakamura
H,
Arai
Y,
Totoki
Y,
et al
Genomic spectra of biliary tract cancer
.
Nat Genet
.
2015
;
47
(9)
:
1003
1010
.
18.
Kankeu Fonkoua
LA,
Serrano Uson Junior
PL,
Mody
K,
Mahipal
A,
Borad
MJ,
Roberts
LR.
Novel and emerging targets for cholangiocarcinoma progression: therapeutic implications
.
Expert Opin Ther Targets
.
2022
;
26
(1)
:
79
92
.
19.
National Comprehensive Cancer Network
.
NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines): hepatobiliary cancers
. .
20.
Sirica
AE,
Strazzabosco
M,
Cadamuro
M.
Intrahepatic cholangiocarcinoma: morpho-molecular pathology, tumor reactive microenvironment, and malignant progression
.
Adv Cancer Res
.
2021
;
149
:
321
387
.
21.
Jain
A,
Borad
MJ,
Kelley
RK,
et al
Cholangiocarcinoma with FGFR genetic aberrations: a unique clinical phenotype
.
JCO Precis Oncol
.
2018
;
2
:
1
12
.
22.
Florio
AA,
Ferlay
J,
Znaor
A,
et al
Global trends in intrahepatic and extrahepatic cholangiocarcinoma incidence from 1993 to 2012
.
Cancer
.
2020
;
126
(11)
:
2666
2678
.
23.
Cao
J,
Hu
J,
Liu
S,
et al
Intrahepatic cholangiocarcinoma: genomic heterogeneity between eastern and western patients
.
JCO Precis Oncol
.
2020
;
4
:
PO.18.00414. doi:10.1200/PO.18.00414
24.
Sirpal
S,
Chandok
N.
Primary sclerosing cholangitis: diagnostic and management challenges
.
Clin Exp Gastroenterol
.
2017
;
10
:
265
273
.
25.
Prokopic
M,
Beuers
U.
Management of primary sclerosing cholangitis and its complications: an algorithmic approach
.
Hepatol Int
.
2021
;
15
(1)
:
6
20
.
26.
Boonstra
K,
Weersma
RK,
van Erpecum
KJ,
et al
Population-based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis
.
Hepatology
.
2013
;
58
(6)
:
2045
2055
.
27.
Burak
K,
Angulo
P,
Pasha
TM,
Egan
K,
Petz
J,
Lindor
KD.
Incidence and risk factors for cholangiocarcinoma in primary sclerosing cholangitis
.
Am J Gastroenterol
.
2004
;
99
(3)
:
523
526
.
28.
Tyson
GL,
El-Serag
HB.
Risk factors for cholangiocarcinoma
.
Hepatology
.
2011
;
54
(1)
:
173
184
.
29.
Clements
O,
Eliahoo
J,
Kim
JU,
Taylor-Robinson
SD,
Khan
SA.
Risk factors for intrahepatic and extrahepatic cholangiocarcinoma: a systematic review and meta-analysis
.
J Hepatol
.
2020
;
72
(1)
:
95
103
.
30.
Ahn
KS,
Kang
KJ.
Molecular heterogeneity in intrahepatic cholangiocarcinoma
.
World J Hepatol
.
2020
;
12
(12)
:
1148
1157
.
31.
De Lorenzo
S,
Tovoli
F,
Mazzotta
A,
et al
Non-alcoholic steatohepatitis as a risk factor for intrahepatic cholangiocarcinoma and its prognostic role
.
Cancers (Basel)
.
2020
;
12
(11)
:
3182
.
32.
Wongjarupong
N,
Assavapongpaiboon
B,
Susantitaphong
P,
et al
Non-alcoholic fatty liver disease as a risk factor for cholangiocarcinoma: a systematic review and meta-analysis
.
BMC Gastroenterol
.
2017
;
17
(1)
:
149
.
33.
Kinoshita
M,
Kubo
S,
Tanaka
S,
et al
The association between non-alcoholic steatohepatitis and intrahepatic cholangiocarcinoma: a hospital based case-control study
.
J Surg Oncol
.
2016
;
113
(7)
:
779
783
.
34.
Labib
PL,
Goodchild
G,
Pereira
SP.
Molecular pathogenesis of cholangiocarcinoma
.
BMC Cancer
.
2019
;
19
(1)
:
185
.
35.
Komori
J,
Marusawa
H,
Machimoto
T,
et al
Activation-induced cytidine deaminase links bile duct inflammation to human cholangiocarcinoma
.
Hepatology
.
2008
;
47
(3)
:
888
896
.
36.
Andersen
JB.
Molecular pathogenesis of intrahepatic cholangiocarcinoma
.
J Hepatobiliary Pancreat Sci
.
2015
;
22
(2)
:
101
113
.
37.
Sripa
B,
Kaewkes
S,
Sithithaworn
P,
et al
Liver fluke induces cholangiocarcinoma
.
PLoS Med
.
2007
;
4
(7)
:
e201
.
38.
Gonzalez
RS,
Raza
A,
Propst
R,
et al
Recent advances in digestive tract tumors: updates from the 5th edition of the World Health Organization “Blue Book”
.
Arch Pathol Lab Med
.
2021
;
145
(5)
:
607
626
.
39.
Forner
A,
Vidili
G,
Rengo
M,
Bujanda
L,
Ponz-Sarvise
M,
Lamarca
A.
Clinical presentation, diagnosis and staging of cholangiocarcinoma
.
Liver Int
.
2019
;
39
(suppl 1)
:
98
107
.
40.
Van Beers
BE.
Diagnosis of cholangiocarcinoma
.
HPB (Oxford)
.
2008
;
10
(2)
:
87
93
.
41.
Zhang
C,
Wang
H,
Ning
Z,
et al
Serum liver enzymes serve as prognostic factors in patients with intrahepatic cholangiocarcinoma
.
Onco Targets Ther
.
2017
;
10
:
1441
1449
.
42.
Bragazzi
MC,
Ridola
L,
Safarikia
S,
et al
New insights into cholangiocarcinoma: multiple stems and related cell lineages of origin
.
Ann Gastroenterol
.
2018
;
31
(1)
:
42
55
.
43.
Vijgen
S,
Terris
B,
Rubbia-Brandt
L.
Pathology of intrahepatic cholangiocarcinoma
.
Hepatobiliary Surg Nutr
.
2017
;
6
(1)
:
22
34
.
44.
Kamp
E,
Dinjens
WNM,
Doukas
M,
et al
Optimal tissue sampling during ERCP and emerging molecular techniques for the differentiation of benign and malignant biliary strictures
.
Therap Adv Gastroenterol
.
2021
;
14
:
17562848211002023
.
45.
Moeini
A,
Haber
PK,
Sia
D.
Cell of origin in biliary tract cancers and clinical implications
.
JHEP Rep
.
2021
;
3
(2)
:
100226
.
46.
Montal
R,
Sia
D,
Montironi
C,
et al
Molecular classification and therapeutic targets in extrahepatic cholangiocarcinoma
.
J Hepatol
.
2020
;
73
(2)
:
315
327
.
47.
Aloia
T,
Pawlik
TM,
Taouli
B,
Rubbia-Brandt
L,
Vauthey
J-N.
Intrahepatic bile ducts
.
In
:
Amin
MB,
Edge
SB,
Greene
FL,
et al
, eds.
AJCC Cancer Staging Manual
. 8th ed.
Springer International Publishing
;
2017
:
XVII
,
1032
.
48.
Muttillo
EM,
Ciardi
A,
Troiano
R,
et al
Pancreatic ductal adenocarcinoma and distal cholangiocarcinoma: a proposal of preoperative diagnostic score for differential diagnosis
.
World J Surg Oncol
.
2021
;
19
(1)
:
10
.
49.
Nagtegaal
ID,
Odze
RD,
Klimstra
D,
et al
The 2019 WHO classification of tumours of the digestive system
.
Histopathology
.
2020
;
76
(2)
:
182
188
.
50.
Wen
KW,
Joseph
NM,
Srivastava
A,
et al
Inhibin-positive hepatic carcinoma: proposal for a solid-tubulocystic variant of intrahepatic cholangiocarcinoma
.
Hum Pathol
.
2021
;
116
:
82
93
.
51.
Argani
P,
Palsgrove
DN,
Anders
RA,
et al
A novel NIPBL-NACC1 gene fusion is characteristic of the cholangioblastic variant of intrahepatic cholangiocarcinoma
.
Am J Surg Pathol
.
2021
;
45
(11)
:
1550
1560
.
52.
Liao
X,
Zhang
D.
The 8th edition American Joint Committee on Cancer Staging for Hepato-pancreato-biliary Cancer: a review and update
.
Arch Pathol Lab Med
.
2021
;
145
(5)
:
543
553
.
53.
Amin
MB,
Edge
SB,
Greene
FL,
et al
AJCC Cancer Staging Manual
. 8th ed.
Springer
;
2017
.
54.
Nagorney
DM,
Pawlik
TM,
Chun
YS,
Ebata
T,
Vauthey
JN.
Perihilar bile ducts
.
In
:
Amin
MB,
Edge
SB,
Greene
FL,
et al
, eds.
AJCC Cancer Staging Manual
. 8th ed.
Springer International Publishing
;
2017
:
XVII
,
1032
.
55.
Krasinskas
A,
Pawlik
TM,
Mino-Kenudson
M,
Vauthey
JN.
Distal bile duct
.
In
:
Amin
MB,
Edge
SB,
Greene
FL,
et al
, eds.
AJCC Cancer Staging Manual
. 8th ed.
Springer International Publishing
;
2017
:
XVII
,
1032
.
56.
Selves
J,
Long-Mira
E,
Mathieu
MC,
Rochaix
P,
Ilie
M.
Immunohistochemistry for diagnosis of metastatic carcinomas of unknown primary site
.
Cancers (Basel)
.
2018
;
10
(4)
:
108
.
57.
Lee
T,
Teng
TZJ,
Shelat
VG.
Carbohydrate antigen 19-9—tumor marker: past, present, and future
.
World J Gastrointest Surg
.
2020
;
12
(12)
:
468
490
.
58.
Takahashi
Y,
Dungubat
E,
Kusano
H,
et al
Application of immunohistochemistry in the pathological diagnosis of liver tumors
.
Int J Mol Sci
.
2021
;
22
(11)
:
5780
.
59.
Sigel
CS,
Drill
E,
Zhou
Y,
et al
Intrahepatic cholangiocarcinomas have histologically and immunophenotypically distinct small and large duct patterns
.
Am J Surg Pathol
.
2018
;
42
(10)
:
1334
1345
.
60.
Fernandez Moro
C,
Fernandez-Woodbridge
A,
Alistair D’souza
M,
et al
Immunohistochemical typing of adenocarcinomas of the pancreatobiliary system improves diagnosis and prognostic stratification
.
PLoS One
.
2016
;
11
(11)
:
e0166067
.
61.
Vyas
M,
Jain
D.
A practical diagnostic approach to hepatic masses
.
Indian J Pathol Microbiol
.
2018
;
61
(1)
:
2
17
.
62.
Radwan
NA,
Ahmed
NS.
The diagnostic value of arginase-1 immunostaining in differentiating hepatocellular carcinoma from metastatic carcinoma and cholangiocarcinoma as compared to HepPar-1
.
Diagn Pathol
.
2012
;
7
(1)
:
149
.
63.
Patil
PA,
Taddei
T,
Jain
D,
Zhang
X.
HNF-1β is a more sensitive and specific marker than C-reactive protein for identifying biliary differentiation in primary hepatic carcinomas
.
Arch Pathol Lab Med
.
2022
;
146
(2)
:
220
226
.
64.
Yang
MX,
Coates
RF,
Ambaye
A,
et al
Investigation of HNF-1B as a diagnostic biomarker for pancreatic ductal adenocarcinoma
.
Biomark Res
.
2018
;
6
:
25
.
65.
Nasir
A,
Lehrke
HD,
Mounajjed
T,
et al
Albumin in situ hybridization can be positive in adenocarcinomas and other tumors from diverse sites
.
Am J Clin Pathol
.
2019
;
152
(2)
:
190
199
.
66.
Lu
S-X,
Huang
Y-H,
Liu
L-L,
et al
α-Fetoprotein mRNA in situ hybridisation is a highly specific marker of hepatocellular carcinoma: a multi-centre study
.
Br J Cancer
.
2021
;
124
(12)
:
1988
1996
.
67.
Wang
JD,
Sebastian
C,
Walther
Z,
et al
An appraisal of immunohistochemical stain use in hepatic metastasis highlights the effectiveness of the individualized, case-based approach
.
Arch Pathol Lab Med
.
2023
;
147
(2)
:
185
192
.
68.
Patel
AH,
Harnois
DM,
Klee
GG,
LaRusso
NF,
Gores
GJ.
The utility of CA 19-9 in the diagnoses of cholangiocarcinoma in patients without primary sclerosing cholangitis
.
Am J Gastroenterol
.
2000
;
95
(1)
:
204
207
.
69.
Guro
H,
Kim
JW,
Choi
Y,
Cho
JY,
Yoon
YS,
Han
HS.
Multidisciplinary management of intrahepatic cholangiocarcinoma: current approaches
.
Surg Oncol
.
2017
;
26
(2)
:
146
152
.
70.
Nevi
L,
Di Matteo
S,
Carpino
G,
et al
DCLK1, a putative stem cell marker in human cholangiocarcinoma
.
Hepatology
.
2021
;
73
(1)
:
144
159
.
71.
Rizvi
S,
Khan
SA,
Hallemeier
CL,
Kelley
RK,
Gores
GJ.
Cholangiocarcinoma—evolving concepts and therapeutic strategies
.
Nat Rev Clin Oncol
.
2018
;
15
(2)
:
95
111
.
72.
Bekaii-Saab
TS,
Bridgewater
J,
Normanno
N.
Practical considerations in screening for genetic alterations in cholangiocarcinoma
.
Ann Oncol
.
2021
;
32
(9)
:
1111
1126
.
73.
Bourien
H,
Lamarca
A,
McNamara
MG,
Hubner
RA,
Valle
JW,
Edeline
J.
Druggable molecular alterations in bile duct cancer: potential and current therapeutic applications in clinical trials
.
Expert Opin Investig Drugs
.
2021
;
30
(9)
:
975
983
.
74.
Misumi
K,
Hayashi
A,
Shibahara
J,
et al
Intrahepatic cholangiocarcinoma frequently shows loss of BAP1 and PBRM1 expression, and demonstrates specific clinicopathological and genetic characteristics with BAP1 loss
.
Histopathology
.
2017
;
70
(5)
:
766
774
.
75.
Kim
SJ,
Akita
M,
Sung
YN,
et al
MDM2 amplification in intrahepatic cholangiocarcinomas: its relationship with large-duct type morphology and uncommon KRAS mutations
.
Am J Surg Pathol
.
2018
;
42
(4)
:
512
521
.
76.
Chung
T,
Park
YN.
Up-to-date pathologic classification and molecular characteristics of intrahepatic cholangiocarcinoma
.
Front Med (Lausanne)
.
2022
;
9
:
857140
.
77.
Jeon
Y,
Kwon
SM,
Rhee
H,
et al
Molecular and radiopathologic spectrum between HCC and intrahepatic cholangiocarcinoma
.
Hepatology
.
2023
;
77
(1)
:
92
108
.
78.
Akita
M,
Sofue
K,
Fujikura
K,
et al
Histological and molecular characterization of intrahepatic bile duct cancers suggests an expanded definition of perihilar cholangiocarcinoma
.
HPB (Oxford)
.
2019
;
21
(2)
:
226
234
.
79.
Sinniah
RS,
Shapses
MS,
Ahmed
MU,
Babiker
H,
Chandana
SR.
Novel biomarkers for cholangiocarcinoma: how can it enhance diagnosis, prognostication, and investigational drugs—Part-1
.
Expert Opin Investig Drugs
.
2021
;
30
(10)
:
1047
1056
.
80.
Krook
MA,
Reeser
JW,
Ernst
G,
et al
Fibroblast growth factor receptors in cancer: genetic alterations, diagnostics, therapeutic targets and mechanisms of resistance
.
Br J Cancer
.
2021
;
124
(5)
:
880
892
.
81.
Liu
G,
Chen
T,
Ding
Z,
Wang
Y,
Wei
Y,
Wei
X.
Inhibition of FGF-FGFR and VEGF-VEGFR signalling in cancer treatment
.
Cell Prolif
.
2021
;
54
(4)
:
e13009
.
82.
Yue
S,
Li
Y,
Chen
X,
et al
FGFR-TKI resistance in cancer: current status and perspectives
.
J Hematol Oncol
.
2021
;
14
(1)
:
23
.
83.
Jacobi
O,
Ross
JS,
Goshen-Lago
T,
et al
ERBB2 pathway in biliary tract carcinoma: clinical implications of a targetable pathway
.
Oncol Res Treat
.
2021
;
44
(1–2)
:
20
27
.
84.
Thornblade
LW,
Wong
P,
Li
D,
et al
Patterns of whole exome sequencing in resected cholangiocarcinoma
.
Cancers (Basel)
.
2021
;
13
(16)
:
4062
.
85.
Tessiri
S,
Techasen
A,
Kongpetch
S,
et al
Therapeutic targeting of ARID1A and PI3K/AKT pathway alterations in cholangiocarcinoma
.
PeerJ
.
2022
;
10
:
e12750
.
86.
Zheng
Y,
Qin
Y,
Gong
W,
et al
Specific genomic alterations and prognostic analysis of perihilar cholangiocarcinoma and distal cholangiocarcinoma
.
J Gastrointest Oncol
.
2021
;
12
(6)
:
2631
2642
.
87.
Farshidfar
F,
Zheng
S,
Gingras
MC,
et al
Integrative genomic analysis of cholangiocarcinoma identifies distinct IDH-mutant molecular profiles
.
Cell Rep
.
2017
;
18
(11)
:
2780
2794
.
88.
Bibeau
K,
Féliz
L,
Lihou
CF,
Ren
H,
Abou-Alfa
GK.
Progression-free survival in patients with cholangiocarcinoma with or without FGF/FGFR alterations: a FIGHT-202 post hoc analysis of prior systemic therapy response
.
JCO Precis Oncol
.
2022
;
6
:
e2100414
.
89.
Yoon
JG,
Kim
MH,
Jang
M,
et al
Molecular characterization of biliary tract cancer predicts chemotherapy and programmed death 1/programmed death-ligand 1 blockade responses
.
Hepatology
.
2021
;
74
(4)
:
1914
1931
.
90.
Hoy
SM.
Pemigatinib: first approval
.
Drugs
.
2020
;
80
(9)
:
923
929
.
91.
Kang
C.
Infigratinib: first approval
.
Drugs
.
2021
;
81
(11)
:
1355
1360
.
92.
News
CCA.
October 10, 2022: Important Information About Truseltiq
. .
93.
Taiho Pharmaceutical Co Ltd Japan
.
LYTGOBI (futibatinib) United States prescribing information
.
US Food and Drug Administration. https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/214801s000lbl.pdf. Updated September 2022. Accessed October 10, 2022
.
94.
Adeva
J.
Current development and future perspective of IDH1 inhibitors in cholangiocarcinoma
.
Liver Cancer Int
.
2021
;
3
(1)
:
17
31
.
95.
Rizzo
A,
Ricci
AD,
Detecting
Brandi G.
and targeting NTRK gene fusions in cholangiocarcinoma: news and perspectives
.
Expert Rev Precis Med Drug Dev
.
2021
;
6
(4)
:
225
227
.
96.
Verdaguer
H,
Sauri
T,
Acosta
DA,
et al
ESMO scale for clinical actionability of molecular targets driving targeted treatment in patients with cholangiocarcinoma
.
Clin Cancer Res
.
2022
;
28
(8)
:
1662
1671
.
97.
Malone
ER,
Oliva
M,
Sabatini
PJB,
Stockley
TL,
Siu
LL.
Molecular profiling for precision cancer therapies
.
Genome Med
.
2020
;
12
(1)
:
8
.
98.
El-Deiry
WS,
Goldberg
RM,
Lenz
HJ,
et al
The current state of molecular testing in the treatment of patients with solid tumors, 2019
.
CA Cancer J Clin
.
2019
;
69
(4)
:
305
343
.
99.
Jogo
T,
Nakamura
Y,
Shitara
K,
et al
Circulating tumor DNA analysis detects FGFR2 amplification and concurrent genomic alterations associated with FGFR inhibitor efficacy in advanced gastric cancer
.
Clin Cancer Res
.
2021
;
27
(20)
:
5619
5627
.
100.
Sigel
C,
Wang
T.
Pathological diagnosis of cholangiocarcinoma
.
In
:
Tabibian
JH,
ed.
Diagnosis and Management of Cholangiocarcinoma: A Multidisciplinary Approach
.
Springer Nature Switzerland AG
;
2021
:
203
228
.
101.
Akita
M,
Sawada
R,
Komatsu
M,
et al
An immunostaining panel of C-reactive protein, N-cadherin, and S100 calcium binding protein P is useful for intrahepatic cholangiocarcinoma subtyping
.
Hum Pathol
.
2021
;
109
:
45
52
.
102.
Mall
AS,
Tyler
MG,
Ho
SB,
et al
The expression of MUC mucin in cholangiocarcinoma
.
Pathol Res Pract
.
2010
;
206
(12)
:
805
809
.
103.
Park
SY,
Roh
SJ,
Kim
YN,
et al
Expression of MUC1, MUC2, MUC5AC and MUC6 in cholangiocarcinoma: prognostic impact
.
Oncol Rep
.
2009
;
22
(3)
:
649
657
.
104.
Lok
T,
Chen
L,
Lin
F,
Wang
HL.
Immunohistochemical distinction between intrahepatic cholangiocarcinoma and pancreatic ductal adenocarcinoma
.
Hum Pathol
.
2014
;
45
(2)
:
394
400
.
105.
Sasaki
M,
Sato
Y.
An immunohistochemical panel of insulin-like growth factor II mRNA-binding protein 3 (IMP3), enhancer of zeste homolog 2 (EZH2), and p53 is useful for a diagnosis in bile duct biopsy
.
Virchows Arch
.
2021
;
479
(4)
:
697
703
.
106.
Misdraji
J,
Centeno
BA,
Pitman
MB.
Ancillary tests in the diagnosis of liver and pancreatic neoplasms
.
Cancer Cytopathol
.
2018
;
126
(suppl 8)
:
672
690
.
107.
Porcell
L AI,
De Young
BR,
Proca
DM,
Frankel
WL.
Immunohistochemical analysis of hepatocellular and adenocarcinoma in the liver: MOC31 compares favorably with other putative markers
.
Mod Pathol
.
2000
;
13
(7)
:
773
778
.
108.
Kakar
S,
Muir
T,
Murphy
LM,
Lloyd
RV,
Burgart
LJ.
Immunoreactivity of Hep Par 1 in hepatic and extrahepatic tumors and its correlation with albumin in situ hybridization in hepatocellular carcinoma
.
Am J Clin Pathol
.
2003
;
119
(3)
:
361
366
.
109.
Ferrone
CR,
Ting
DT,
Shahid
M,
et al
The ability to diagnose intrahepatic cholangiocarcinoma definitively using novel branched DNA-enhanced albumin RNA in situ hybridization technology
.
Ann Surg Oncol
.
2016
;
23
(1)
:
290
296
.
110.
Aldyab
M,
El Jabbour
T,
Parilla
M,
Lee
H.
Benign vs malignant pancreatic lesions: molecular insights to an ongoing debate
.
World J Gastrointest Surg
.
2021
;
13
(5)
:
406
418
.
111.
Ai
D,
Yao
J,
Yang
F,
et al
TRPS1: a highly sensitive and specific marker for breast carcinoma, especially for triple-negative breast cancer
.
Mod Pathol
.
2021
;
34
(4)
:
710
719
.
112.
Gurel
B,
Ali
TZ,
Montgomery
EA,
et al
NKX3.1 as a marker of prostatic origin in metastatic tumors
.
Am J Surg Pathol
.
2010
;
34
(8)
:
1097
1105
.
113.
Beach
R,
Gown
AM,
De Peralta-Venturina
MN,
et al
P504S immunohistochemical detection in 405 prostatic specimens including 376 18-gauge needle biopsies
.
Am J Surg Pathol
.
2002
;
26
(12)
:
1588
1596
.
114.
Queisser
A,
Hagedorn
SA,
Braun
M,
Vogel
W,
Duensing
S,
Perner
S.
Comparison of different prostatic markers in lymph node and distant metastases of prostate cancer
.
Mod Pathol
.
2015
;
28
(1)
:
138
145
.
115.
Serag Eldien
MM,
Abdou
AG,
Elghrabawy
GRA,
Alhanafy
AM,
Mahmoud
SF.
Stratification of urothelial bladder carcinoma depending on immunohistochemical expression of GATA3 and CK5/6
.
J Immunoassay Immunochem
.
2021
;
42
(6)
:
662
678
.
116.
Smith
SC,
Mohanty
SK,
Kunju
LP,
et al
Uroplakin II outperforms uroplakin III in diagnostically challenging settings
.
Histopathology
.
2014
;
65
(1)
:
132
138
.
117.
Kaufmann
O,
Volmerig
J,
Dietel
M.
Uroplakin III is a highly specific and moderately sensitive immunohistochemical marker for primary and metastatic urothelial carcinomas
.
Am J Clin Pathol
.
2000
;
113
(5)
:
683
687
.
118.
Maleki
Z,
Nadella
A,
Nadella
M,
Patel
G,
Patel
S,
Kholova
I.
INSM1, a novel biomarker for detection of neuroendocrine neoplasms: cytopathologists’ view
.
Diagnostics (Basel)
.
2021
;
11
(12)
:
2172
.
119.
Damjanov
I.
Testicular germ cell tumors: serological and immunohistochemical diagnosis
.
Acta Med Acad
.
2021
;
50
(1)
:
58
70
.
120.
Fassnacht
M,
Assie
G,
Baudin
E,
et al
Adrenocortical carcinomas and malignant phaeochromocytomas: ESMO-EURACAN Clinical Practice Guidelines for diagnosis, treatment and follow-up
.
Ann Oncol
.
2020
;
31
(11)
:
1476
1490
.
121.
Loy
TS,
Phillips
RW,
Linder
CL.
A103 immunostaining in the diagnosis of adrenal cortical tumors: an immunohistochemical study of 316 cases
.
Arch Pathol Lab Med
.
2002
;
126
(2)
:
170
172
.
122.
Heidarpour
M,
Tavanafar
Z.
Diagnostic utility of PAX8 in differentiation of mullerian from non-mullerian tumors
.
Adv Biomed Res
.
2014
;
3
:
96
.
123.
Li
H,
Jing
X,
Yu
J,
et al
A combination of cytokeratin 5/6, p63, p40 and MUC5AC are useful for distinguishing squamous cell carcinoma from adenocarcinoma of the cervix
.
Diagn Pathol
.
2020
;
15
(1)
:
104
.
124.
Chapel
DB,
Schulte
JJ,
Husain
AN,
Krausz
T.
Application of immunohistochemistry in diagnosis and management of malignant mesothelioma
.
Transl Lung Cancer Res
.
2020
;
9
(suppl 1)
:
S3
S27
.
125.
Wang
HL.
Ancillary tests for hepatobiliary neoplasms: frequently asked questions
.
Presented at: USCAP 111th Annual Meeting
;
March 19–24, 2022
;
Los Angeles, CA
.
126.
Memorial Sloan Kettering Cancer Center
.
MSK-IMPACT: a targeted test for mutations in both rare and common cancers
.
https://www.mskcc.org/msk-impact. Accessed June 29, 2022
.
127.
Foundation Medicine
.
FoundationOne CDx
. .
128.
Foundation Medicine
.
FoundationOne Liquid CDx
. .
129.
Foundation Medicine
.
FoundationOne Heme
. .
130.
Guardant Health
.
Lead with liquid: lead with Guardant360 CDx
. .
131.
Integrated DNA Technologies (previously Archer DX)
.
FusionPlex Pan Solid Tumor v2
. .
132.
Tempus
.
Genomic profiling
. .
135.
136.
NeoGenomics
.
NeoTYPE Discovery Profile for Solid Tumors
. .
137.
KEW Inc
.
CANCERPLEX
. .
138.
Caris Life Sciences
.
Comprehensive molecular profiling
. .
140.
OmniSeq Corporation
.
Introducing OmniSeq INSIGHT
. .
141.
PathGroup
.
SmartGenomics
. .

Author notes

This review was funded by Incyte Corporation (Wilmington, Delaware).

Competing Interests

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