Context.—

Immunohistochemistry serves as an ancillary diagnostic tool for a wide variety of neoplastic and nonneoplastic disorders, including infections, workup of inflammatory conditions, and subtyping neoplasms of the pancreas/liver/gastrointestinal luminal tract. In addition, immunohistochemistry is also used to detect a variety of prognostic and predictive molecular biomarkers for carcinomas of the pancreas, liver, and gastrointestinal luminal tract.

Objective.—

To highlight an update on the role of immunohistochemistry in the evaluation of pancreatic/liver/gastrointestinal luminal tract disorders.

Data Sources.—

Literature review and authors’ research data and personal practice experience were used.

Conclusions.—

Immunohistochemistry is a valuable tool, assisting in the diagnosis of problematic tumors and benign lesions of the pancreas, liver, and gastrointestinal luminal tract, and also in the prediction of prognosis and therapeutic response for carcinomas of the pancreas, liver, and gastrointestinal luminal tract.

This article will review immunohistochemical (IHC) markers commonly used as adjuncts in diagnostic problems of tumors and benign lesions of the pancreas, liver, and gastrointestinal (GI) luminal tract. IHC of normal and benign and neoplastic change in pancreas, liver, and GI luminal tract is covered, as well as applications of newer diagnostic markers and molecular biomarkers. In addition to the literature review, we have included our own experience and tested numerous antibodies reported in the literature and in our institution.

Geisinger Medical Laboratories (Danville, Pennsylvania) performed tissue microarray sections and routine sections stained with Dako and Ventana Systems on normal pancreatic tissue of 40 cases. The data revealed that both normal pancreatic ductal and acinar epithelial cells are IHC positive for CK7, CAM5.2, MOC-31, Ber-EP4, prostate stem cell antigen (PSCA), ATRX/DAXX, and DPC4/SMAD4. Normal pancreatic ductal epithelial cells also express MUC6, CK19 (focal), claudin 18 (focal), annexin A8 (weak), claudin4 (weak), MUC1 (weak, on luminal side), and mesothelin (weak). Expression of S100A, monoclonal carcinoembryonic antigen (mCEA), and CA19-9 in normal pancreatic ductal epithelial cells is usually absent or weakly positive. The weak positivity of mCEA is on the luminal side of normal pancreatic duct epithelial cells. Normal pancreatic acinar cells express BCL10 and trypsin; they can weakly express CA19-9 as well. Normal and reactive pancreatic ducts are usually negative for CK20, CK17, maspin, IMP3 (KOC), trypsin, MUC2, MUC4, and MUC5AC. Approximately 10% of pancreatic ducts and acini are focally positive for CDX2.

Autoimmune pancreatitis (AIP) is categorized into types 1 and 2. The former is mediated by immunoglobulin (Ig) G4–positive plasma cells, whereas the latter is characterized by neutrophilic destruction of pancreatic ductal cells. Accordingly, type 1 AIP lesions show dense distribution of IgG4-positive plasma cells, measured by tissue IgG4 counts (>50/high-power field for resection specimens and >10/high-power field for biopsy specimens) or IgG4:IgG ratios (>40%).1  Neutrophils present in most AIP type II lesions are almost entirely absent in type I AIP.2  Recently, ductal programmed death ligand-1 (PD-L1) reactivity has been reported to have potential to distinguish type 2 AIP from other forms of pancreatitis and pancreatic ductal adenocarcinoma (PDAC). The sensitivity and specificity of PD-L1 as a marker of type 2 AIP were 70% and 99%, respectively.3 

Differentiation of PDAC cells from reactive pancreatic ductal epithelial cells can sometimes be very challenging. Many markers, such as mCEA, CA19-9, MUC1, MUC5AC, SMAD4 (loss), mesothelin, and p53, have been reported. In our experience, a panel consisting of pathophysiology von Hippel-Lindau (pVHL), maspin, S100P, and IMP3 works very well in the distinction of PDAC from normal/reactive pancreatic ducts. Negativity of pVHL and positivity of the remaining 3 markers support a diagnosis of PDAC, instead of a reactive process.47  Of note, maspin is normally expressed in normal gastric mucosa and duodenal mucosa. When background staining for S100P is present, S100A6 can be a good substitute, although weak nuclear and cytoplasmic staining for S100A6 can be seen in normal/reactive pancreatic ducts.

Pancreatic acinar cell carcinoma (ACC) typically stains positive for CK AE1/3, BCL10, trypsin, chymotrypsin, and PDX-1.8  BCL10 is considered the most sensitive and specific marker to confirm the diagnosis of ACC9  (Figure 1, A through C). Other ACC markers include lipase, amylase, and carboxyl ester lipase.10  ACC tumor cells express α1-antitrypsin, an acute-phase plasma protein that can be secreted from normal hepatocytes, small intestinal enterocytes, monocytes, macrophages, and bronchial epithelial cells. Therefore, it is not a useful marker for ACC. A recent multicenter study demonstrated that carboxypeptidase A1 (CPA1), a zinc metalloprotease produced by pancreatic acinar cells, was strongly expressed only in acinar cells of the pancreas11,12  (Figure 1, A, B, and D). The sensitivity and specificity of CPA1 in diagnosis of ACC were 100% and 95%, respectively.

Figure 1

Application of carboxypeptidase A1 (CPA1) immunostain in the diagnosis of pancreatic acinar cell carcinoma (ACC). ACC grew in a solid pattern, with adjacent normal pancreatic tissue (A). The tumor cells were relatively uniform, with abundant amphophilic cytoplasm and prominent nucleoli (B). Both BCL-10 (C) and CPA1 (D) highlighted tumor cells in a cytoplasm-staining pattern. The upper one-third normal pancreatic tissue served as a positive control (C and D) (original magnifications ×4 [A], ×20 [B], and ×10 [C and D]).

Figure 1

Application of carboxypeptidase A1 (CPA1) immunostain in the diagnosis of pancreatic acinar cell carcinoma (ACC). ACC grew in a solid pattern, with adjacent normal pancreatic tissue (A). The tumor cells were relatively uniform, with abundant amphophilic cytoplasm and prominent nucleoli (B). Both BCL-10 (C) and CPA1 (D) highlighted tumor cells in a cytoplasm-staining pattern. The upper one-third normal pancreatic tissue served as a positive control (C and D) (original magnifications ×4 [A], ×20 [B], and ×10 [C and D]).

Close modal

There are some pitfalls in diagnosis of ACC using special or IHC stains.13  About 20% of ACCs show both nuclear and membranous positivity of β-catenin, and 20% of ACCs display loss of expression of SMAD4/DPC4. Periodic acid–Schiff diastase stain is usually positive in ACC. Trypsin immunostain may give a strong background staining. Hepatocellular differentiation markers, including albumin mRNA in situ hybridization (ISH), may be positive in ACC. When more than 30% of tumor cells are positive for neuroendocrine markers, the tumor should be regarded as mixed acinar and neuroendocrine carcinoma.

The 2019 WHO classification of neuroendocrine neoplasms categorizes pancreatic neuroendocrine neoplasms into pancreatic neuroendocrine tumor (PanNET) and pancreatic neuroendocrine carcinoma (PanNEC).14,15  The former is separated into 3 grades (G1 to G3) based on mitotic rate or Ki-67 index, and the latter is further divided into small cell neuroendocrine carcinoma and large cell neuroendocrine carcinoma based on histomorphology. Both PanNETs and PanNECs usually stain positive for cytokeratin, synaptophysin, chromogranin, and insulinoma-associated protein 1 (INSM1).

According to the 2019 WHO classification of tumors of the digestive system,15 Ki-67 is no longer used as a marker to differentiate PanNET G3 from PanNEC.16  A panel consisting of SST2, ATRX, DAXX, p53, and RB1 is usually helpful in differentiating PanNET G3 from PanNEC. The vast majority of NETs show wild-type pattern of p53 expression, intact expression of SST2, and loss of α-thalassemia/mental retardation X-linked (ATRX)/death domain–associated protein (DAXX), whereas most PanNECs display SST2 negativity, mutation pattern of p53, and loss of RB1.17  The molecular signature of PanNET G3 is MEN1, DAXX/ATRX, and mTOR mutations, whereas PanNEC typically shows mutations in TP53, RB1, KRAS, and SMAD4 genes.18  In difficult cases, testing for pancreatic hormones may help, as most PanNETs produce hormones whereas PanNECs do not. Table 1 lists the IHC markers useful in differentiating PanNET G3 from PanNEC.

Table 1

Useful Immunohistochemical Markers in Differentiating Pancreatic Neuroendocrine Tumor (PanNET) Grade 3 (G3) From Pancreatic Neuroendocrine Carcinoma (PanNEC)

Useful Immunohistochemical Markers in Differentiating Pancreatic Neuroendocrine Tumor (PanNET) Grade 3 (G3) From Pancreatic Neuroendocrine Carcinoma (PanNEC)
Useful Immunohistochemical Markers in Differentiating Pancreatic Neuroendocrine Tumor (PanNET) Grade 3 (G3) From Pancreatic Neuroendocrine Carcinoma (PanNEC)

Recent studies demonstrate that status of alternative lengthening of telomeres and loss of ATRX/DAXX are associated not only with adverse prognostic pathologic features but also with shortened 5-year relapse-free survival in nonfunctional PanNETs.19,20 

These 2 populations of cells have different immunoprofiles. Hepatocellular differentiation is determined on 2 levels: (1) At the cytologic level, cells with hepatocytic differentiation express arginase-1, HepPar-1, and keratin 8/18 and show positivity on albumin RNA ISH.2123  (2) At the architectural level, hepatocytic differentiation is evidenced by presence of canalicular structure, which can be highlighted by polyp CEA, villin, CD10, and bile salt export pump (BSEP) antibodies.24  TTF1 antibody clone 8G7G3/1 recognizes hyperplastic cytoplasm.25  Malignant hepatocytic cells also express α-fetoprotein, glypican-3, glutamine synthetase, and heat shock protein 70 (HSP70).26  Cholangiocytes have no specific markers. They express keratins 7 and 19 and display positivity on albumin RNA ISH.

In the diagnosis of liver tumors, the distinction between hepatocellular carcinomas (HCCs) and intrahepatic cholangiocarcinomas (iCCAs) by histomorphology sometimes can be challenging.27,28  A recent study reported that C-reactive protein (CRP) showed a sensitivity of 75.7% and a specificity of 91.1% in the diagnosis of iCCA. Another new marker, hepatocyte nuclear factor 1β (HNF-1β), exhibited improved performance, with a sensitivity of 100% and a specificity of 92.31% (Figure 2, A through I).29  Please note that this application is limited to workup on primary liver tumors, as HNF-1β expression is shown in a variety of other tumors, such as ovarian clear cell carcinomas, endometrial carcinomas, colorectal carcinomas (CRCs), clear cell renal cell carcinomas, yolk sac tumors, PDACs, and urothelial carcinomas. HNF-1β is a more sensitive and specific marker than CRP for the diagnosis of iCCA and to identify the iCCA component in combined hepatocellular-cholangiocarcinoma. Lack of HNF-1β expression may be used to exclude iCCA from consideration in cases of adenocarcinomas of unknown primary.29,30 Table 2 lists the hepatocellular and cholangiocytic markers.

Figure 2

Application of hepatocyte nuclear factor 1β (HNF-1β) immunostain in differentiating hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC). In nonneoplastic liver tissue, very weak and heterogeneous nuclear positivity was present in hepatocytes. In contrast, cholangiocytes in native bile ducts and ductular reaction were strongly and diffusely labeled by HNF-1β immunostain (A). In moderately differentiated HCC, the tumor cells had minimal nuclear immunoreactivity to HNF-1β (B and C). In poorly differentiated HCC, the tumor cells had weak and heterogeneous nuclear immunoreactivity to HNF-1β (D and E). In well-differentiated CC, the tumor cells demonstrated homogeneous, diffuse, and strong positivity for HNF-1β (F and G). In moderately differentiated CC, the tumor cell nuclei stained diffusely and strongly positive for HNF-1β (H and I) (original magnification ×10 [A through I]).

Figure 2

Application of hepatocyte nuclear factor 1β (HNF-1β) immunostain in differentiating hepatocellular carcinoma (HCC) and cholangiocarcinoma (CC). In nonneoplastic liver tissue, very weak and heterogeneous nuclear positivity was present in hepatocytes. In contrast, cholangiocytes in native bile ducts and ductular reaction were strongly and diffusely labeled by HNF-1β immunostain (A). In moderately differentiated HCC, the tumor cells had minimal nuclear immunoreactivity to HNF-1β (B and C). In poorly differentiated HCC, the tumor cells had weak and heterogeneous nuclear immunoreactivity to HNF-1β (D and E). In well-differentiated CC, the tumor cells demonstrated homogeneous, diffuse, and strong positivity for HNF-1β (F and G). In moderately differentiated CC, the tumor cell nuclei stained diffusely and strongly positive for HNF-1β (H and I) (original magnification ×10 [A through I]).

Close modal
Table 2

Hepatocellular and Cholangiocytic Markers

Hepatocellular and Cholangiocytic Markers
Hepatocellular and Cholangiocytic Markers

Liver sinusoidal endothelial cells (LSECs) differ from classic vascular endothelial cells in multiple aspects:21,24,31  (1) they lack basement membranes and tight junctions; (2) they are architecturally noncontinuous and fenestrated; and (3) they act mainly as scavengers, eliminating macromolecules and small particulates from the blood, and as immune gatekeepers through their striking endocytotic activity.32  Not surprisingly, LSECs present a unique immunoprofile: They strongly express macrophage markers (CD4, CD14, CD16, CD31, CDw32 [FcRII], ICAM-1) but rarely express CD34 or factor VIII. They stain positive for ERG but negative for vimentin and D2-40.3335 

After vascular remodeling as seen in benign and malignant liver conditions, LSECs undergo capillarization and can switch immunophenotypically and functionally to classic vascular endothelial cells, which can aid in diagnosis.36 

Bile ductular reactions are commonly seen in cholestatic liver diseases. Using a panel of markers such as CD56, EMA, and CD10 may help to differentiate probable etiologies.37  In fulminant hepatic failure and cirrhosis, hepatic ductules stain positive for CD10, CD56, and EMA. In an autoimmune bile duct injury, such as primary biliary cholangitis and primary sclerosing cholangitis, hepatic ductules express CD56 but not EMA or CD10. When the biliary system is obstructed, hepatic ductules express EMA but not CD10 or CD56.38 

The 2019 WHO classification of tumors of the digestive system15  classifies hepatocellular adenoma (HCA) into 4 types: HNF1A mutated, inflammatory, β-catenin activated, and unclassified.39,40  Recent substantial accumulation of histomorphologic and molecular data enables a more accurate subclassification of HCA (Table 3).4143  Please note that in HCC, expression of LFABP can be lost in 16% to 25% of cases.44  Interpretation of serum amyloid A (SAA) and CRP immunostain results requires more caution; particularly, they should not be used as a reliable feature to imply a specific type of adenoma as potential precursor for HCC, as their expression can be detected in HCC.45  The expression of CRP and SAA can also be seen in focal nodular hyperplasia. This is sometimes a more important diagnostic challenge because focal nodular hyperplasia is often the most important morphologic mimicker of inflammatory hepatic adenoma when dealing with small biopsies. In addition, SAA and CRP can be expressed in cirrhotic nodules. As to the sonic hedgehog–activated HCA markers, ASS1 generally is more sensitive than PTGDS.4547 

Table 3

Immunohistochemistry Markers for Classifying Hepatocellular Adenoma

Immunohistochemistry Markers for Classifying Hepatocellular Adenoma
Immunohistochemistry Markers for Classifying Hepatocellular Adenoma

Histologic diagnosis of Barrett esophagus relies on identification of goblet cells, that is, intestinal metaplasia, in the tubular esophagus mucosa that is 1 cm above the gastroesophageal junction (GEJ). Recognition of goblet cells can occasionally be challenging.48  Alcian blue stain at pH 2.5 highlights the acidic mucin in the metaplastic goblet cells. The utility of immunostaining markers for Barrett mucosa is debatable in general, and their use is not recommended for clinical practice. The specialized goblet cells have been reported to stain positive for CK7, villin, SOX-9, HepPar-1, Das1, and CDX2.4952  It is worth noting that gastric cardiac mucosa can sometimes stain positive for villin.

There are currently no sensitive and specific markers for diagnosing dysplasia in Barrett esophagus.53,54  Cytologic and architectural alterations are still the best criteria for this purpose. Some authors believe aberrant p53 expression, P504S positivity, and IMP-3 (KOC) overexpression can help with the distinction.55,56  However, investigations exploring the utility of biomarkers in difficult cases are still waiting for randomized controlled prospective clinical trial data. Nevertheless, the commonly used markers determining cytologic dysplasia include p53 immunostain and the TissueCypher (Castle Biosciences, Pittsburgh, Pennsylvania) assay.57  The latter chooses 9 protein markers for immunofluorescent stain, captures images by an automatic imaging system, and puts the 15 best-performing quantitative image analysis features, such as nuclear morphology and tissue architecture, into an algorithm to generate a risk score (0–10) to classify the risk for progression to high-grade dysplasia/esophageal adenocarcinoma within 5 years into a 3-tier (low, intermediate, or high) or a 2-tier (low, high) risk stratification in patients.5860  Both assays have low sensitivity, with p53 immunostain of 31% to 62% and TissueCypher of 29% to 30.4% (for 3-tier risk classification) and 67.7% (for 2-tier risk classification).

Although one recent study showed a promising role for p53 immunostain in identifying higher risk of progression, regardless of histomorphologic presentation, technical variation in evaluation of p53 expression by this method, such as antibody used, staining method, and definition and interpretation of positivity, also makes results noncomparable between laboratories. Similarly, with the TissueCypher system, the effects of preanalytic and analytic factors on performance of TissueCypher are not fully understood yet. In addition, the data from a large cohort (10 000 patients) study conducted by GMC investigators showed that at an acceptability threshold of $100 000/quality-adjusted life year, TissueCypher is marginally cost-effective in identifying risk of progression within 5 years after diagnosis of Barrett esophagus. The 2022 American Gastroenterological Association guideline does not recommend either. Attempts with p17 and Ki-67 immunostains show some positive results but lack wide validation.

The Cancer Genome Atlas project classifies gastric adenocarcinoma into 4 major genomic subtypes: Epstein-Barr virus–infected tumors, tumors with microsatellite instability, genomically stable tumors, and chromosomally unstable tumors.61  This classification greatly helps determine a patient’s treatment plan. The current National Comprehensive Cancer Network guideline (Gastric Carcinoma, v2.2022) lists HER2 overexpression/amplification, PD-L1 expression, microsatellite instability, high tumor mutation burden (≥10 mutations/megabase), Epstein-Barr virus positivity, and NTRK gene fusion as targetable prognostic markers.62 

About 20% of gastric and GEJ cancers have HER2/neu overexpression and/or amplification. Trastuzumab provides clear survival benefit in patients of advanced stage, as demonstrated in the Trastuzumab for Gastric Cancer trial.63  Trastuzumab in combination with chemotherapy is now the standard of care when treating HER2-positive metastatic gastric and GEJ cancers. Of note, response to the tyrosine kinase inhibitor afatinib can be associated with EGFR and HER2 gene amplification.64,65 

Claudin 18.2, a tight junction protein, is another targetable molecule in gastric cancer. It is highly expressed in cancers of the uterine cervix, pancreas, ovary, lung, and esophagus.66  The monoclonal claudin 18.2 antibody zolbetuximab is associated with significantly longer survival in advanced gastric cancer.67 

Although not currently targetable, loss of E-cadherin protein is prognostic in gastric cancer.68  Hereditary diffuse gastric cancer (HDGC) can be caused by germline CDH-1 gene mutation and presents with signet cell features, which confer worse prognosis, but loss of E-cadherin protein is neither a sensitive nor a specific marker for screening for HDGC. Patients with germline detrimental mutation of CDH-1 have up to an 80% risk of developing gastric cancer in both sexes and up to a 60% risk for lobular breast cancer in women. Genetic counseling and testing should be recommended for suspected HDGC cases.

Benign glandular proliferative lesions involving the appendix include those arising from the appendix, such as reactive hyperplasia (secondary to appendicitis and diverticulum), and conditions with other tissue origins, such as endometriosis, endosalpingiosis, and mesothelial cyst.69  In reactive hyperplasia, appendiceal epithelial cells express CK20, CDX2, SATB2, and variable CK7. When Müllerian-origin glandular hyperplasia is present, the epithelial cells express ER and PAX8, as seen in endometriosis and endosalpingiosis. The stromal cells in endometriosis stain positive for CD10. The epithelial cells in endometriosis can undergo mucinous metaplasia, which may display CK20 positivity on immunostain. Endosalpingiosis may demonstrate variable expression of Wilms tumor 1 (WT-1) in the epithelial cells, but calretinin should be negative.70  Benign mesothelial cells can form cystic structures, especially in the mesoappendix. The lining cells of mesothelial cysts stain positive for both WT-1 and calretinin.

The 2019 WHO classification of tumors of the digestive system15  replaces the old terms goblet cell carcinoid and adenocarcinoma ex-goblet cell carcinoid with appendiceal goblet cell adenocarcinoma (GCA), low grade or high grade. The GCA can also be graded by a 3-tiered system (grades 1, 2, and 3) based on the proportion of tumor that consists of low-grade (tubular and clustered growth) and high-grade (loss of tubular or clustered growth) patterns.71  GCA should be differentiated from classic neuroendocrine tumor or conventional adenocarcinoma with tubular and/or signet ring cell features by morphologic features and IHC studies. Molecularly, GCA bears little resemblance to colorectal adenocarcinoma, with rare KRAS, BRAF, APC, and TP53 gene mutations detected.72 

GCA is composed of tubules or clusters of amphicrine neoplasm containing gobletlike mucinous cells, variable numbers of endocrine cells, and Paneth-like cells. The tumor cells can be IHC biphenotypic: they strongly express intestinal epithelial cell markers, including CEA, CK7, CK20, SATBS, MUC2, and CDX2, and also variably express neuroendocrine markers such as INSM1, CD56, synaptophysin, and chromogranin. Appendiceal neuroendocrine tumor cells are the opposite: they express variable amounts of the intestinal epithelial cell markers mentioned above but with consistent strong expression of neuroendocrine markers.73  Conventional appendiceal adenocarcinoma expresses intestine epithelial cell markers but not neuroendocrine markers.

Mucinous tumors in appendix can be de novo mesothelial hyperplasia/neoplasms or may arise from endometriosis. Rarely, ovarian mucinous tumor can spread to the appendix, although we now know that tumor spread with the opposite direction between these 2 organs dominates. On IHC, appendiceal mucinous adenocarcinoma stains positive for intestinal markers including CK7, CK20, CDX2, SATB2, β-catenin (nuclear and membranous), and MUC2.7476  Cadherin 17 (CDH17), a transmembrane protein involved in cellular adhesion, has distinct membranous and cytoplasmic expression in GI glands.77,78  CDH17 is expressed in colorectal (including appendiceal), stomach, and pancreatic cancers and cholangiocarcinomas. Ovarian mucinous carcinoma tumor cells have varied immunoreactivity to intestinal cell marker proteins, but with no expression of SATB2, MUC2, or CDH17.79  CK7 is usually diffusely positive in ovarian mucinous tumors, whereas CK20 and CDX2 are usually focally or patchily positive. In contrast, both CDX2 and CK20 are diffusely positive in most appendiceal carcinomas, with variable CK7 positivity.80  On the other hand, they express PAX8 (20%–30%) and WT-1, which can further help with the distinction.81,82  They are almost always negative for nuclear β-catenin expression, suggesting a different molecular pathogenesis than appendiceal mucinous adenocarcinoma.83 

A small proportion of CRCs overexpress the HER2 oncogene, and the effective targeting of this pathway in other malignancies such as breast and gastric cancer has led to efforts to determine if it can also be exploited as a target in CRC.84,85  Activation of the HER2 pathway as a bypass signaling pathway has been identified as a mechanism of resistance for anti-EGFR therapy in both the first-line and salvage settings. It has also been shown that RAS and BRAF wild-type metastatic CRC enriches for the presence of HER2 amplification. In contrast to gastric and breast cancers, the diagnostic criteria for HER2 positivity in CRC have not been fully standardized and have varied across studies.8688  The HER2 Amplification for Colorectal Cancer Enhanced Stratification (HERACLES) trial was a multicenter, open-label phase II clinical trial in patients with CRC resistant to chemotherapy and anti-EGFR therapy. This trial enrolled metastatic CRC patients with wild-type KRAS and HER2 overexpression, who were then treated with a combination of trastuzumab and lapatinib, with an objective response rate of 30%.89  They defined 3 conditions of HER2 positivity as follows (HERACLES criteria): (1) a HER2 IHC 3+ score in 50% or more of CRC cells; (2) a HER2 IHC 3+ score in 10% to 50% of the CRC cells, and a fluorescence ISH HER2/CEP17 2.0 or higher in 50% or more of CRC cells; (3) more than 50% of CRC cells with a HER2 IHC 2+ score and a fluorescence ISH HER2/CEP17 2.0 or higher. However, to date, HER2 diagnostic criteria for gastroesophageal adenocarcinoma88  or other independent HER2 IHC scoring systems90,91  have also been used in CRC HER2 studies. Moreover, the prognostic role of HER2 in CRC remains controversial. Some studies have shown that HER2 overexpression/amplification, as an adverse prognostic factor, is closely correlated with the tumor stage and survival in CRC patients.92  However, other studies have shown no association between HER2 expression and patient survival.88,93  These controversial results suggest that the role of HER2 in CRC needs to be further explored.

1.
Deshpande
V,
Zen
Y,
Chan
JK,
et al
Consensus statement on the pathology of IgG4-related disease
.
Mod Pathol
.
2012
;
25
(9)
:
1181
1192
.
2.
Ku
Y,
Hong
SM,
Fujikura
K,
et al
IL-8 expression in granulocytic epithelial lesions of idiopathic duct-centric pancreatitis (type 2 autoimmune pancreatitis)
.
Am J Surg Pathol
.
2017
;
41
(8)
:
1129
1138
.
3.
Gupta
R,
Neyaz
A,
Chougule
A,
et al
Autoimmune pancreatitis type 2: diagnostic utility of PD-L1 immunohistochemistry
.
Am J Surg Pathol
.
2019
;
43
(7)
:
898
906
.
4.
Yantiss
RK,
Woda
BA,
Fanger
GR,
et al
KOC (K homology domain containing protein overexpressed in cancer): a novel molecular marker that distinguishes between benign and malignant lesions of the pancreas
.
Am J Surg Pathol
.
2005
;
29
(2)
:
188
195
.
5.
Lin
F,
Shi
J,
Liu
H,
et al
Diagnostic utility of S100P and von Hippel-Lindau gene product (pVHL) in pancreatic adenocarcinoma—with implication of their roles in early tumorigenesis
.
Am J Surg Pathol
.
2008
;
32
(1)
:
78
91
.
6.
Karanjawala
ZE,
Illei
PB,
Ashfaq
R,
et al
New markers of pancreatic cancer identified through differential gene expression analyses: claudin 18 and annexin A8
.
Am J Surg Pathol
.
2008
;
32
(2)
:
188
196
.
7.
Deng
H,
Shi
J,
Wilkerson
M,
Meschter
S,
Dupree
W,
Lin
F.
Usefulness of S100P in diagnosis of adenocarcinoma of pancreas on fine-needle aspiration biopsy specimens
.
Am J Clin Pathol
.
2008
;
129
(1)
:
81
88
.
8.
Al-Hader
A,
Al-Rohil
RN,
Han
H,
Von Hoff
D.
Pancreatic acinar cell carcinoma: a review on molecular profiling of patient tumors
.
World J Gastroenterol
.
2017
;
23
(45)
:
7945
7951
.
9.
Hosoda
W,
Sasaki
E,
Murakami
Y,
Yamao
K,
Shimizu
Y,
Yatabe
Y.
BCL10 as a useful marker for pancreatic acinar cell carcinoma, especially using endoscopic ultrasound cytology specimens
.
Pathol Int
.
2013
;
63
(3)
:
176
182
.
10.
Abraham
SC,
Wu
TT,
Hruban
RH,
et al
Genetic and immunohistochemical analysis of pancreatic acinar cell carcinoma: frequent allelic loss on chromosome 11p and alterations in the APC/beta-catenin pathway
.
Am J Pathol
.
2002
;
160
(3)
:
953
962
.
11.
Said
S,
Kurtin
PJ,
Nasr
SH,
et al
Carboxypeptidase A1 and regenerating islet-derived 1α as new markers for pancreatic acinar cell carcinoma
.
Hum Pathol
.
2020
;
103
:
120
126
.
12.
Uhlig
R,
Contreras
H,
Weidemann
S,
et al
Carboxypeptidase A1 (CPA1) immunohistochemistry is highly sensitive and specific for acinar cell carcinoma (ACC) of the pancreas
.
Am J Surg Pathol
.
2022
;
46
(1)
:
97
104
.
13.
Askan
G,
Deshpande
V,
Klimstra
DS,
et al
Expression of markers of hepatocellular differentiation in pancreatic acinar cell neoplasms: a potential diagnostic pitfall
.
Am J Clin Pathol
.
2016
;
146
(2)
:
163
169
.
14.
Rindi
G,
Mete
O,
Uccella
S,
et al
Overview of the 2022 WHO classification of neuroendocrine neoplasms
.
Endocr Pathol
.
2022
;
33
(1)
:
115
154
.
15.
WHO Classification of Tumours Editorial Board
.
Digestive System Tumours
. 5th ed.
Lyon, France
:
International Agency for Research on Cancer Press
;
2019
.
WHO Classification of Tumours; vol 1
.
16.
Nagtegaal
ID,
Odze
RD,
Klimstra
D,
et al
The 2019 WHO classification of tumours of the digestive system
.
Histopathology
.
2020
;
76
(2)
:
182
188
.
17.
Konukiewitz
B,
Jesinghaus
M,
Kasajima
A,
Kloppel
G.
Neuroendocrine neoplasms of the pancreas: diagnosis and pitfalls
.
Virchows Arch
.
2022
;
480
(2)
:
247
257
.
18.
Tang
LH.
Pancreatic neuroendocrine neoplasms: landscape and horizon
.
Arch Pathol Lab Med
.
2020
;
144
(7)
:
816
828
.
19.
Hackeng
WM,
Brosens
LAA,
Kim
JY,
et al
Non-functional pancreatic neuroendocrine tumours: ATRX/DAXX and alternative lengthening of telomeres (ALT) are prognostically independent from ARX/PDX1 expression and tumour size
.
Gut
.
2022
;
71
(5)
:
961
973
.
20.
Singhi
AD,
Liu
TC,
Roncaioli
JL,
et al
Alternative lengthening of telomeres and loss of DAXX/ATRX expression predicts metastatic disease and poor survival in patients with pancreatic neuroendocrine tumors
.
Clin Cancer Res
.
2017
;
23
(2)
:
600
609
.
21.
Chu
PG,
Chu
P,
Weiss
L.
Modern Immunohistochemistry
.
Cambridge, UK
:
Cambridge University Press
;
2009
.
22.
Lin
F,
Shi
J,
Wang
HL,
et al
Detection of albumin expression by RNA in situ hybridization is a sensitive and specific method for identification of hepatocellular carcinomas and intrahepatic cholangiocarcinomas
.
Am J Clin Pathol
.
2018
;
150
(1)
:
58
64
.
23.
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
:
149
.
24.
Dabbs
DJ.
Immunohistology of the pancreas and hepatobiliary tract
. In:
Dabbs
DJ,
ed.
Diagnostic Immunohistochemistry: Theranostic and Genomic Applications
. 4th ed.
Amsterdam, Netherlands
:
Elsevier Saunders
;
2014
:
508
539
.
25.
Pan
CC,
Chen
PC,
Tsay
SH,
Chiang
H.
Cytoplasmic immunoreactivity for thyroid transcription factor-1 in hepatocellular carcinoma: a comparative immunohistochemical analysis of four commercial antibodies using a tissue array technique
.
Am J Clin Pathol
.
2004
;
121
(3)
:
343
349
.
26.
Wang
HL,
Anatelli
F,
Zhai
QJ,
Adley
B,
Chuang
ST,
Yang
XJ.
Glypican-3 as a useful diagnostic marker that distinguishes hepatocellular carcinoma from benign hepatocellular mass lesions
.
Arch Pathol Lab Med
.
2008
;
132
(11)
:
1723
1728
.
27.
Geller
SA,
Dhall
D,
Alsabeh
R.
Application of immunohistochemistry to liver and gastrointestinal neoplasms: liver, stomach, colon, and pancreas
.
Arch Pathol Lab Med
.
2008
;
132
(3)
:
490
499
.
28.
Lagana
SM,
Salomao
M,
Remotti
HE,
Knisely
AS,
Moreira
RK.
Bile salt export pump: a sensitive and specific immunohistochemical marker of hepatocellular carcinoma
.
Histopathology
.
2015
;
66
(4)
:
598
602
.
29.
Patil
PA,
Taddei
T,
Jain
D,
Zhang
X.
HNF-1beta 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
.
30.
Yeh
YC,
Lei
HJ,
Chen
MH,
et al
C-reactive protein (CRP) is a promising diagnostic immunohistochemical marker for intrahepatic cholangiocarcinoma and is associated with better prognosis
.
Am J Surg Pathol
.
2017
;
41
(12)
:
1630
1641
.
31.
DeLeve
LD,
Maretti-Mira
AC.
Liver sinusoidal endothelial cell: an update
.
Semin Liver Dis
.
2017
;
37
(4)
:
377
387
.
32.
Sorensen
KK,
McCourt
P,
Berg
T,
et al
The scavenger endothelial cell: a new player in homeostasis and immunity
.
Am J Physiol Regul Integr Comp Physiol
.
2012
;
303
(12)
:
R1217
R1230
.
33.
Mills
SE.
Histology for Pathologists
. 3rd ed.
Philadelphia, PA
:
Lippincott Williams & Wilkins
;
2007
.
34.
Misdraji
J,
Centeno
BA,
Pitman
MB.
Ancillary tests in the diagnosis of liver and pancreatic neoplasms
.
Cancer Cytopathol
.
2018
;
126
(suppl 8)
:
672
690
.
35.
Taylor
CR,
Cote
RJ.
Immunomicroscopy: A Diagnostic Tool for the Surgical Pathologist
. 3rd ed.
Philadelphia, PA
:
Saunders
;
2006
.
36.
Burt
AD,
Ferrell
LD,
Hübscher
SG.
MacSween’s Pathology of the Liver
. 5th ed.
London, UK
:
Churchill Livingstone
;
2007
.
37.
Turanyi
E,
Dezso
K,
Csomor
J,
Schaff
Z,
Paku
S,
Nagy
P.
Immunohistochemical classification of ductular reactions in human liver
.
Histopathology
.
2010
;
57
(4)
:
607
614
.
38.
Bateman
AC,
Hubscher
SG.
Cytokeratin expression as an aid to diagnosis in medical liver biopsies
.
Histopathology
.
2010
;
56
(4)
:
415
425
.
39.
Bioulac-Sage
P,
Gouw
ASH,
Balabaud
C,
Sempoux
C.
Hepatocellular adenoma: what we know, what we do not know, and why it matters
.
Histopathology
.
2022
;
80
(6)
:
878
897
.
40.
Wang
X,
Zhang
X.
Hepatocellular adenoma: where are we now?
World J Gastroenterol
.
2022
;
28
(14)
:
1384
1393
.
41.
Hytiroglou
P,
Bioulac-Sage
P,
Theise
ND,
Sempoux
C.
Etiology, pathogenesis, diagnosis, and practical implications of hepatocellular neoplasms
.
Cancers (Basel)
.
2022
;
14
(15)
42.
Rowan
DJ,
Yasir
S,
Chen
ZE,
et al
Morphologic and molecular findings in myxoid hepatic adenomas
.
Am J Surg Pathol
.
2021
;
45
(8)
:
1098
1107
.
43.
Torbenson
M.
Hepatic adenomas: classification, controversies, and consensus
.
Surg Pathol Clin
.
2018
;
11
(2)
:
351
366
.
44.
Kim
H,
Lee
H,
Park
YN.
Loss of liver fatty acid binding protein expression in hepatocellular carcinomas is associated with a decreased recurrence-free survival
.
J Liver Cancer
.
2015
;
15
(1)
:
6
.
45.
Sasaki
M,
Yoneda
N,
Sawai
Y,
et al
Clinicopathological characteristics of serum amyloid A-positive hepatocellular neoplasms/nodules arising in alcoholic cirrhosis
.
Histopathology
.
2015
;
66
(6)
:
836
845
.
46.
Nault
JC,
Couchy
G,
Caruso
S,
et al
Argininosuccinate synthase 1 and periportal gene expression in sonic hedgehog hepatocellular adenomas
.
Hepatology
.
2018
;
68
(3)
:
964
976
.
47.
Sala
M,
Gonzales
D,
Leste-Lasserre
T,
et al
ASS1 overexpression: a hallmark of sonic hedgehog hepatocellular adenomas; recommendations for clinical practice
.
Hepatol Commun
.
2020
;
4
(6)
:
809
824
.
48.
Sarbia
M,
Donner
A,
Franke
C,
Gabbert
HE.
Distinction between intestinal metaplasia in the cardia and in Barrett’s esophagus: the role of histology and immunohistochemistry
.
Hum Pathol
.
2004
;
35
(3)
:
371
376
.
49.
Flucke
U,
Steinborn
E,
Dries
V,
et al
Immunoreactivity of cytokeratins (CK7, CK20) and mucin peptide core antigens (MUC1, MUC2, MUC5AC) in adenocarcinomas, normal and metaplastic tissues of the distal oesophagus, oesophago-gastric junction and proximal stomach
.
Histopathology
.
2003
;
43
(2)
:
127
134
.
50.
Glickman
JN,
Wang
H,
Das
KM,
et al
Phenotype of Barrett’s esophagus and intestinal metaplasia of the distal esophagus and gastroesophageal junction: an immunohistochemical study of cytokeratins 7 and 20, Das-1 and 45 MI
.
Am J Surg Pathol
.
2001
;
25
(1)
:
87
94
.
51.
Shearer
C,
Going
J,
Neilson
L,
Mackay
C,
Stuart
RC.
Cytokeratin 7 and 20 expression in intestinal metaplasia of the distal oesophagus: relationship to gastro-oesophageal reflux disease
.
Histopathology
.
2005
;
47
(3)
:
268
275
.
52.
Shi
XY,
Bhagwandeen
B,
Leong
AS.
CDX2 and villin are useful markers of intestinal metaplasia in the diagnosis of Barrett esophagus
.
Am J Clin Pathol
.
2008
;
129
(4)
:
571
577
.
53.
Hao
J,
Critchley-Thorne
R,
Diehl
DL,
Snyder
SR.
A cost-effectiveness analysis of an adenocarcinoma risk prediction multi-biomarker assay for patients with Barrett’s esophagus
.
Clinicoecon Outcomes Res
.
2019
;
11
:
623
635
.
54.
Shaheen
NJ,
Falk
GW,
Iyer
PG,
et al
Diagnosis and management of Barrett’s esophagus: an updated ACG guideline
.
Am J Gastroenterol
.
2022
;
117
(4)
:
559
587
.
55.
Lu
D,
Vohra
P,
Chu
PG,
Woda
B,
Rock
KL,
Jiang
Z.
An oncofetal protein IMP3: a new molecular marker for the detection of esophageal adenocarcinoma and high-grade dysplasia
.
Am J Surg Pathol
.
2009
;
33
(4)
:
521
525
.
56.
Redston
M,
Noffsinger
A,
Kim
A,
et al
Abnormal TP53 predicts risk of progression in patients with Barrett’s esophagus regardless of a diagnosis of dysplasia
.
Gastroenterology
.
2022
;
162
(2)
:
468
481
.
57.
Ireland
AP,
Clark
GW,
DeMeester
TR.
Barrett’s esophagus: the significance of p53 in clinical practice
.
Ann Surg
.
1997
;
225
(1)
:
17
30
.
58.
Hanas
JS,
Lerner
MR,
Lightfoot
SA,
et al
Expression of the cyclin-dependent kinase inhibitor p21(WAF1/CIP1) and p53 tumor suppressor in dysplastic progression and adenocarcinoma in Barrett esophagus
.
Cancer
.
1999
;
86
(5)
:
756
763
.
59.
Moskaluk
CA,
Heitmiller
R,
Zahurak
M,
Schwab
D,
Sidransky
D,
Hamilton
SR.
p53 and p21(WAF1/CIP1/SDI1) gene products in Barrett esophagus and adenocarcinoma of the esophagus and esophagogastric junction
.
Hum Pathol
.
1996
;
27
(11)
:
1211
1220
.
60.
Shi
XY,
Bhagwandeen
B,
Leong
AS.
p16, cyclin D1, Ki–67, and AMACR as markers for dysplasia in Barrett esophagus
.
Appl Immunohistochem Mol Morphol
.
2008
;
16
(5)
:
447
452
.
61.
Cancer Genome Atlas Research Network
.
Comprehensive molecular characterization of gastric adenocarcinoma
.
Nature
.
2014
;
513
(7517)
:
202
209
.
62.
Niclauss
N,
Gutgemann
I,
Dohmen
J,
Kalff
JC,
Lingohr
P.
Novel biomarkers of gastric adenocarcinoma: current research and future perspectives
.
Cancers (Basel)
.
2021
;
13
(22)
:
5660
.
63.
Bang
YJ,
Van Cutsem
E,
Feyereislova
A,
et al
Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial
.
Lancet
.
2010
;
376
(9742)
:
687
697
.
64.
Matsubara
J,
Yamada
Y,
Hirashima
Y,
et al
Impact of insulin-like growth factor type 1 receptor, epidermal growth factor receptor, and HER2 expressions on outcomes of patients with gastric cancer
.
Clin Cancer Res
.
2008
;
14
(10)
:
3022
3029
.
65.
Sanchez-Vega
F,
Hechtman
JF,
Castel
P,
et al
EGFR and MET amplifications determine response to HER2 inhibition in ERBB2-amplified esophagogastric cancer
.
Cancer Discov
.
2019
;
9
(2)
:
199
209
.
66.
Sahin
U,
Koslowski
M,
Dhaene
K,
et al
Claudin-18 splice variant 2 is a pan-cancer target suitable for therapeutic antibody development
.
Clin Cancer Res
.
2008
;
14
(23)
:
7624
7634
.
67.
Sahin
U,
Tureci
O,
Manikhas
G,
et al
FAST: a randomised phase II study of zolbetuximab (IMAB362) plus EOX versus EOX alone for first-line treatment of advanced CLDN18.2-positive gastric and gastro-oesophageal adenocarcinoma
.
Ann Oncol
.
2021
;
32
(5)
:
609
619
.
68.
Guilford
P,
Hopkins
J,
Harraway
J,
et al
E-cadherin germline mutations in familial gastric cancer
.
Nature
.
1998
;
392
(6674)
:
402
405
.
69.
Mai
KT,
Burns
BF.
Development of dysplastic mucinous epithelium from endometriosis of the appendix
.
Histopathology
.
1999
;
35
(4)
:
368
372
.
70.
McCluggage
WG,
Clements
WD.
Endosalpingiosis of the colon and appendix
.
Histopathology
.
2001
;
39
(6)
:
645
646
.
71.
Bell
PD,
Pai
RK.
Goblet cell adenocarcinoma of the appendix: an update and practical approach to diagnosis and grading [published online June 9, 2022]
.
Hum Pathol
.
2022
.
72.
Zhang
K,
Meyerson
C,
Kassardjian
A,
Westbrook
LM,
Zheng
W,
Wang
HL.
Goblet cell carcinoid/carcinoma: an update
.
Adv Anat Pathol
.
2019
;
26
(2)
:
75
83
.
73.
Yozu
M,
Johncilla
ME,
Srivastava
A,
et al
Histologic and outcome study supports reclassifying appendiceal goblet cell carcinoids as goblet cell adenocarcinomas, and grading and staging similarly to colonic adenocarcinomas
.
Am J Surg Pathol
.
2018
;
42
(7)
:
898
910
.
74.
Li
Z,
Roth
R,
Rock
JB,
et al
Dual immunostain with SATB2 and CK20 differentiates appendiceal mucinous neoplasms from ovarian mucinous neoplasms
.
Am J Clin Pathol
.
2017
;
147
(5)
:
484
491
.
75.
Moh
M,
Krings
G,
Ates
D,
Aysal
A,
Kim
GE,
Rabban
JT.
SATB2 expression distinguishes ovarian metastases of colorectal and appendiceal origin from primary ovarian tumors of mucinous or endometrioid type
.
Am J Surg Pathol
.
2016
;
40
(3)
:
419
432
.
76.
Vang
R,
Gown
AM,
Wu
LS,
et al
Immunohistochemical expression of CDX2 in primary ovarian mucinous tumors and metastatic mucinous carcinomas involving the ovary: comparison with CK20 and correlation with coordinate expression of CK7
.
Mod Pathol
.
2006
;
19
(11)
:
1421
1428
.
77.
Panarelli
NC,
Yantiss
RK,
Yeh
MM,
Liu
Y,
Chen
YT.
Tissue-specific cadherin CDH17 is a useful marker of gastrointestinal adenocarcinomas with higher sensitivity than CDX2
.
Am J Clin Pathol
.
2012
;
138
(2)
:
211
222
.
78.
Su
MC,
Yuan
RH,
Lin
CY,
Jeng
YM.
Cadherin-17 is a useful diagnostic marker for adenocarcinomas of the digestive system
.
Mod Pathol
.
2008
;
21
(11)
:
1379
1386
.
79.
Lin
F,
Shi
J,
Zhu
S,
et al
Cadherin-17 and SATB2 are sensitive and specific immunomarkers for medullary carcinoma of the large intestine
.
Arch Pathol Lab Med
.
2014
;
138
(8)
:
1015
1026
.
80.
Vang
R,
Gown
AM,
Barry
TS,
et al
Cytokeratins 7 and 20 in primary and secondary mucinous tumors of the ovary: analysis of coordinate immunohistochemical expression profiles and staining distribution in 179 cases
.
Am J Surg Pathol
.
2006
;
30
(9)
:
1130
1139
.
81.
Aldaoud
N,
Erashdi
M,
AlKhatib
S,
Abdo
N,
Al-Mohtaseb
A,
Graboski-Bauer
A.
The utility of PAX8 and SATB2 immunohistochemical stains in distinguishing ovarian mucinous neoplasms from colonic and appendiceal mucinous neoplasm
.
BMC Res Notes
.
2019
;
12
(1)
:
770
.
82.
Goldstein
NS,
Bassi
D,
Uzieblo
A.
WT1 is an integral component of an antibody panel to distinguish pancreaticobiliary and some ovarian epithelial neoplasms
.
Am J Clin Pathol
.
2001
;
116
(2)
:
246
252
.
83.
Elias
KM,
Labidi-Galy
SI,
Vitonis
AF,
et al
Prior appendectomy does not protect against subsequent development of malignant or borderline mucinous ovarian neoplasms
.
Gynecol Oncol
.
2014
;
132
(2)
:
328
333
.
84.
Greally
M,
Kelly
CM,
Cercek
A.
HER2: an emerging target in colorectal cancer
.
Curr Probl Cancer
.
2018
;
42
(6)
:
560
571
.
85.
La Salvia
A,
Lopez-Gomez
V,
Garcia-Carbonero
R.
HER2-targeted therapy: an emerging strategy in advanced colorectal cancer
.
Expert Opin Investig Drugs
.
2019
;
28
(1)
:
29
38
.
86.
Conradi
LC,
Styczen
H,
Sprenger
T,
et al
Frequency of HER-2 positivity in rectal cancer and prognosis
.
Am J Surg Pathol
.
2013
;
37
(4)
:
522
531
.
87.
Park
JS,
Yoon
G,
Kim
HJ,
et al
HER2 status in patients with residual rectal cancer after preoperative chemoradiotherapy: the relationship with molecular results and clinicopathologic features
.
Virchows Arch
.
2018
;
473
(4)
:
413
423
.
88.
Richman
SD,
Southward
K,
Chambers
P,
et al
HER2 overexpression and amplification as a potential therapeutic target in colorectal cancer: analysis of 3256 patients enrolled in the QUASAR, FOCUS and PICCOLO colorectal cancer trials
.
J Pathol
.
2016
;
238
(4)
:
562
570
.
89.
Valtorta
E,
Martino
C,
Sartore-Bianchi
A,
et al
Assessment of a HER2 scoring system for colorectal cancer: results from a validation study
.
Mod Pathol
.
2015
;
28
(11)
:
1481
1491
.
90.
Moussa
M,
Badawy
A,
Helal
N,
et al
Differential expression of HER2 and SKP2 in benign and malignant colorectal lesions
.
Asian Pac J Cancer Prev
.
2020
;
21
(8)
:
2357
2366
.
91.
Shabbir
A,
Mirza
T,
Khalid
AB,
Qureshi
MA,
Asim
SA.
Frequency of Her2/neu expression in colorectal adenocarcinoma: a study from developing South Asian country
.
BMC Cancer
.
2016
;
16
(1)
:
855
.
92.
Ingold Heppner
B,
Behrens
HM,
Balschun
K,
et al
HER2/neu testing in primary colorectal carcinoma
.
Br J Cancer
.
2014
;
111
(10)
:
1977
1984
.
93.
Marx
AH,
Burandt
EC,
Choschzick
M,
et al
Heterogenous high-level HER-2 amplification in a small subset of colorectal cancers
.
Hum Pathol
.
2010
;
41
(11)
:
1577
1585
.

Author notes

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