Open Access

Protein‑loss of SWI/SNF‑complex core subunits influences prognosis dependent on histological subtypes of intra‑ and extrahepatic cholangiocarcinoma

  • Authors:
    • Britta Janina Wagner
    • Patrick Sven Plum
    • Katharina Apel
    • Marie Scherer
    • Denise Buchner
    • Sebastian Brinkmann
    • Reinhard Buettner
    • Dirk Stippel
    • Alexander Quaas
    • Uta Drebber
  • View Affiliations

  • Published online on: March 3, 2021     https://doi.org/10.3892/ol.2021.12610
  • Article Number: 349
  • Copyright: © Wagner et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cholangiocarcinoma (CCA) is an aggressive malignancy with a 5‑year‑survival rate of <10%, mainly due to diagnosis in advanced stages and limited therapeutic options in case of progressive disease. Recently, evidence has indicated that alterations in the SWI/SNF‑complex (SWI/SNF) may have an important role in the tumorigenesis of CCA. SWI/SNF‑related chromatin remodeling has been reported to be crucial for differentiation and tumor suppression, and loss‑of‑function mutations of SWI/SNF are present in 20% of human malignancies; however, at present, little is known about its relevance in CCA. In the present study, a cohort of 52 patients with the diagnosis of primary CCA was retrospectively collected. All patients underwent surgery with curative intent. Tissue microarray analysis was performed on each tumor for immunohistochemical loss‑of‑protein analysis of the SWI/SNF core subunits ARID1A, INI‑1, BRG1, PBRM‑1 and BRM, corresponding to the following CCA subtypes: Extrahepatic CCA (ECCA), small duct or large duct intrahepatic CCA (ICCA). Kaplan‑Meier analysis was used to determine survival distribution and survival differences were evaluated by log‑rank test. In total, 14 of 52 patients (~35%) exhibited protein‑loss of any tested SWI/SNF core subunit. Notably, 17% of patients exhibited a loss of ARID1a; this was the protein loss with the highest frequency. Patients with small and large duct ICCA with protein‑loss of any tested SWI/SNF subunit exhibited significantly worse survival compared with the wild‑type cohort with proficient protein expression (P=0.013 and P=0.002), whereas no significant survival difference was detected for patients with ECCA. SWI/SNF and its core subunits may be considered promising predictive and therapeutic targets, and require further investigation in patients with CCA.

Introduction

Cholangiocarcinoma (CCA) are epithelial cell malignancies arising from various locations within the biliary tree, showing markers of cholangiocyte differentiation. Most CCA are adenocarcinomas, which are anatomically classified as either of intrahepatic (ICCA) or extrahepatic (ECCA) origin (1). In general this heterogeneous group of CCA are aggressive malignancies with poor survival rates (5-year-survival of <10%) (2). Resectability is a critical factor associated with a better outcome. However, the majority of cases is diagnosed at the time of progressive disease, without the opportunity of surgical resection in curative intent (3,4). These data emphasize the need of new therapeutic options besides surgery, chemo- (CTX) and radiotherapy (RTX). So far, there are only very limited options of available targeted therapies, which are based on specific biomarkers of individual tumors like e.g. FGFR2-inhibitors (5). For this reason new biomarkers are urgently required, that might contribute to a better understanding of tumor biology in specific subgroups of the different CCA types. ICCAs are subdivided into large duct type and small duct type, according to their occurrence and cell of origin (68). Large duct ICCAs arise in the large intrahepatic bile ducts near the hepatic hilus and resemble ECCAs, whereas small duct ICCAs typically develop in the hepatic periphery (7,9). The ICCA subtypes, which were included into the WHO classification of 2019, go along with different clinical, histomorphological and molecular features, diverse risk factors and prognosis (10,11).

Recently increasing interest has been paid to nucleosome remodeling complexes in many different cancers, e.g. esophageal-, pancreatic-, ovarian-, renal cell and hepatocellular carcinoma and they appear to be promising new opportunities for novel prognostic markers and therapeutic targets (1214). The mammalian SWI/SNF-complex (SWI/SNF) functions as tumor suppressor in many human malignancies and plays an ATP-dependent chromatin-remodeling role, contributing to transcriptional regulation by altering chromatin structure and controlling the accessibility of DNA (15,16).

SWI/SNF are heterogeneous complexes of 12–15 protein subunits with diverse and variable functions required in different cellular and developmental contexts. Mammalian SWI/SNF complexes always contain two mutually exclusive ATPases: BRM (SMARCA2) or BRG1 (SMARCA4). Three additional subunits [INI-1 (SMARCB1), SMARCC1 and SMARCC2] form the core complex (1719). The mutation frequency of the complexes is ~20% in cancer and AT-rich interactive domain-containing protein 1A (ARID1A) is the most frequently mutated gene subunit (12). Moreover, mutations of SWI/SNF might provide a potential target for therapy (13).

To date, inactivating mutations of ARID1a and PBRM-1 have been detected in 17–19% of ICCA by exome sequencing and have been correlated with worse survival (20). ARID1a and PBRM-1 gene mutations have even been described as predictors for poor prognosis in ICCA, however histomorphological subtypes were not considered (21). Simbolo et al demonstrated that specific molecular SWI/SNF alterations, including ARID1a, PBRM-1 and INI-1, were associated with different CCA subtypes and that potentially actionable pathways for small molecule inhibitors are evident in 68% of cases (16).

Despite advances in the analysis of SWI/SNF, so far little is known about its relevance in CCA. In our study we concentrated on the analysis of the ATPase-dependent core subunits of SWI/SNF (ARID1a, INI-1, BRG1, PBRM-1 and BRM) which, if mutated, all separately result in loss-of-function of SWI/SNF (22). These mutations can be detected by immunohistochemistry (IHC), demonstrating the loss of the nuclear protein.

Cases of large- and small duct ICCA as well as cases of ECCA were included in order to analyze the frequency of mutations of SWI/SNF in correlation to the subtype of CCA and survival. Simultaneously we assessed FGFR2-translocations, HER2-amplification status and P53 mutations to exclude other underlying alterations, which may bias our survival data.

Due to the abundance of actionable mutations and the absence of effective systemic therapy options against CCA, the aim of our study was, to reveal the influence of SWI/SNF core subunits protein-loss on overall survival of CCA patients corresponding to its subtype and further to identify possible new promising therapeutic targets.

Materials and methods

Case selection

Between 2000 and 2019, a cohort of 52 patients with the diagnosis of primary CCA was collected. All these patients underwent surgical treatment with curative intent within the surgical department of the University Hospital of Cologne (Cologne, Germany). All patients gave their written informed consent for the procedure. The current retrospective study was conducted with the approval of the Ethics Committee of the University of Cologne, utilizing clinical follow-up data that were collected retrospectively according to a standardized follow-up within the oncological outpatient clinic (application 18–269). The following exclusion criteria were defined: i) Administration of systematic therapy prior to surgery to avoid any bias affecting survival analysis. ii) Survival <14 day after surgery, to exclude short-term deaths due to surgical complications. iii) Age <42 or >89 years. Detailed patient cohort information is displayed in (Table I).

Table I.

Specifications of the patient cohort.

Table I.

Specifications of the patient cohort.

CharacteristicsTotalECCAICCAICCA-large ductICCA-small duct
Total patients521735827
Sex
  Male311021417
  Female21615312
Portal vein embolization83514
Postoperative chemotherapy22220515
Postoperative radiotherapy21110
Alive at time of investigation15312210
  pT-statius
  pT112012210
  pT2251213112
  pT392716
  pT462431
  pN1231013211
  M131202
  L1441430525
  V122418315
  Pn1341519316
R-status
  R0381028325
  R1136743
  R210101
Grading
  G111000
  G2301119613
  G320416115
  G410101
Operation
  Hemihepatectomy right80817
  Extended hemihepatectomy right81725
  Hemihepatectomy left91817
  Extended hemihepatectomy left41303
  Liver segment resection50514
  Atypical liver resection20211
  Pancreas operation (Whipple or Traverso-modification)22000
  Trisegmentectomy43101
  Extrahepatic bile duct resection55000
  Hemihepatectomy right + extrahepatic bile duct resection42211
  Hemihepatectomy left + pancreas operation11000

[i] ECCA, extrahepatic cholangiocarcinoma; ICCA, intrahepatic cholangiocarcinoma.

Classification and pathological features

CCAs were divided in ECCA (n=17) and ICCA (n=35) corresponding to their location in the biliary tree. Moreover, ICCA were subdivided into small duct (n=27) and large duct type (n=8) corresponding to the WHO classification of 2019 (10). This subdivision was based on standard histomorphological and cellular criteria (HE and PAS staining), as well as immunohistochemical staining (see below).

Immunohistochemical study (IHC)

Tissue microarray analysis (TMA) construction was performed as previously described (23,24). In brief, four tissue cylinders from each tumor center with a diameter of 1.2 mm were punched out from the tumor center of selected tumor tissue blocks using a semi-automated precision instrument and embedded in empty recipient paraffin blocks. Four-micrometer sections of the resulting TMA blocks were transferred to an adhesive-coated slide system (Instrumedics Inc.) for further staining. IHC was performed on TMA slides using primary antibodies specific for ARID1A (Abcam, clone EPR13501, 1:1,000 citrate buffer), BRG1 (Abcam, clone EPNCIR111A, 1:300 citrate buffer), BRM (Cell Signaling, Inc., clone D9E8B, 1:50 citrate buffer), INI1 (Dako, clone DO-7, 1:1200, citrate buffer), PBRM-1 (Abcam, BAF180, EPR15860, 1:1000, EDTA buffer) and P53 (Dako, clone DO-7, 1:800, citrate buffer) with a Bond Max automated system (Leica). All these markers showed a nuclear staining pattern. For subdivision of ICCA into small duct- and large duct type the primary antibodies S100P (Cell Marque, 16/f5, 1:1600, enzyme buffer), N-cadherin (Novocastra, IAR06, 1:100, EDTA buffer), CD56 (Thermo Fisher Scientific, Inc., 123C3, 1:500, EDTA buffer) and MUC5ac (Abcam, MUC5AC/917, 1:500, EDTA buffer) were used in the same manner. S100P and MUC5ac showed a cytoplasmatic -, N-cadherin and CD56 a membranous staining pattern. The expression frequency of these markers corresponding to the CC subtypes is depicted in Fig. 1. Lymphoid tissue served as an internal control. Two pathologists (either U.D. or A.Q. and B.J.W.) manually performed IHC analysis. ARID1A-, BRG1-, BRM-, PBRM-1and INI-1 staining was assessed according to a three-tier scoring system (score 0, 1 and 2). A score of 0 associated with the loss of protein and defined as unequivocal clean absent staining in the nuclei of viable tumor cells for the SWI/SNF-complex subunits was interpreted as an underlying mutation, deletion or promotor alteration. Score 1 was determined as nuclear staining of tumor cells and interpreted as an intact, unmuted ARID1-, BRG1-, BRM-, PBRM-1 or INI1 gene with regular protein expression. Score 2 was used in case of heterogeneous expression, defined as loss and intact nuclear staining within different tumor areas of the same tumor sample and interpreted as partial underlying mutation, deletion or promotor alteration. Strong nuclear stainability of the surrounding non-tumor cells served as an internal control. Discrepant results were resolved by consensus between the reviewers. In case of PBRM-1 to proof absent nuclear staining certain tumor samples were reanalyzed on large tumor slides. S100P, N-cadherin, CD56 and MUC5ac staining was classed as positive if ≥10% of tumor cells were positive. P53 was considered as altered in case of nuclear loss or homogeneous nuclear overexpression.

FISH-analysis

Fluorescence in situ hybridization (FISH) for FGFR2-translocation and HER2-amplification was performed on formalin-fixed, paraffin-embedded tissue specimens (TMA). Sections of 1.5 µm tumor material were cut and hybridized overnight. FGFR2-translocation was assessed by using a ZytoLight SPEC, FGFR2 Dual Color Break Apart probe (Z-2169-200), HER2-amplification by using a ZytoLight SPEC ERBB2/CEN 17 Dual Color probe (Z-2015-200). Review of fluorescence signals was performed at ×630 magnification with a Leica CTR 5500 fluorescence microscope. FGFR2 was scored as positive, if separate Spectrum red and/or Spectrum green signals were present in >20% of nuclei throughout the tumor. HER2 analysis was performed according to defined guidelines (25).

Data analysis and statistics

For statistical analysis and graphic presentation of the results, IBM SPSS v26.0 was used. Descriptive analysis included the frequency of nominal parameters, the median with lower (LQ) and upper (UQ) quartiles for numeric variables (ordinal or asymmetric distribution) and the mean for numeric variables with a normal distribution. Prognosis was calculated including all types of mortality beginning 14 days after the date of surgery. In this way, mortality associated to the surgical procedure itself was excluded. Kaplan-Meier univariate analysis was used to describe survival distribution, and log-rank tests were performed to evaluate survival differences. Significant differences between patient cohorts were defined as P<0.05 for all analyses.

Results

IHC analysis of SWI/SNF complex subunits

ARID1a-, BRG1-, BRM-, PBRM-1and INI-1 IHC staining shows a clear nuclear staining pattern, if proficiently expressed (Fig. 2). In case of protein-loss the nuclear staining turned negative. For ARID1a and PBRM-1 only complete protein-loss was detected whereas BRG1-, BRM and INI1 also showed a heterogeneous staining pattern. Assuming that a heterogeneous protein expression at least results in a reduced function of SWI/SNF, we summarized both immunophenotypes as protein-loss for further analysis. This approach was confirmed by the finding that heterogeneous protein expression and protein-loss, separately analyzed, both resulted in reduced overall survival (data not shown).

In total, 14 of 52 patients (~35%) showed a protein-loss of any tested SWI/SNF core subunit, 2 of them with heterogeneous expression, and 4 of them with protein-loss of more than one SWI/SNF subunits. The highest frequency showed ARID1a with 17%. The CCA subtypes showed different frequencies of the lost SWI/SNF subunits (Fig. 3). In small duct ICCA, all tested subunits showed protein loss. The most frequent alterations were detected for ARID1a and PBRM-1 with 11% each. In opposite, in large duct type ICCA only protein-loss of ARID1a (25%) and BRM (12.5%) were revealed. With 6 of 17 ECCA patients this CCA subtype showed the highest frequency (35%) of any lost SWI/SNF subunit. ARID1a, BRM and PBRM-1 alterations were found in decreasing frequency. Besides these findings, alterations of HER2, P53 and FGFR2 were observed as follows: HER2 amplification in 13.46%, P53 alterations in 25% and FGFR2 translocation in 5.77% (Fig. 3). Whereas HER2- and P53 alterations were found in each CCA subtype, FGFR2 translocations were only observed in ICCA. Further, 7 of the 14 patients showed protein-loss of SWI/SNF subunits parallel to HER2, FGFR2- or P53 alterations.

Overall survival analysis

Kaplan-Meier analysis (log-rank test) was used for overall survival. We combined IHC proven protein-loss of ARID1A, BRG1, BRM, PBRM-1 or INI-1 as SWI/SNF altered cohort (n=14) and compared it to patients with intact expression pattern (n=38) (Fig. 4A). The median survival of the SWI/SNF subunit protein-loss cohort was 13±4.677 months versus 34±7.200 months in the wild-type CCA patients, which was statistically significant (P=0.013). Further, we analyzed the effect of molecular alterations (HER2-amplification, FGFR2-translocation or P53 alteration) on the overall survival and excluded CCA patients with these alterations from the cohorts (Fig. 4B). Again, the difference in overall survival (P=0.002) was statistically significant: The median survival of SWI/SNF subunit protein loss cohort (n=25) was 10±7.071 months compared to 43±13.835 months for the cohort with intact expression pattern (n=7).

As we did not see an effect of HER2, P53 and FGFR2 alterations on overall survival in ratio SWI/SNF altered patients and patients with intact expression pattern, we analyzed the overall survival corresponding to the histological CCA subtypes in the total patient cohort (Fig. 5). Only for small duct (Fig. 5A) and large duct ICCA (Fig. 5B) a significant worse survival for the SWI/SNF subunit protein-loss cohort was detected (small duct ICCA P=0.031, large duct ICCA P=0.010), but not for ECCA (Fig. 5C). For small duct ICCA the median overall survival of the cohort with intact expression pattern was 43.911±7.356 months, whereas SWI/SNF altered patients only survived 13±7.088 months in average. Similar results were detected for large duct ICC: overall survival of the cohort with intact expression pattern was 46.5±6.759 months compared with 16±8.667 months in SWI/SNF altered patients. General overall survival of small duct ICCA was the longest (39.709±6.849 months), followed by large duct ICCA (33.571±7.480 months). The shortest overall survival was detected for ECCA patients (21.486±6.381 months). Each SWI/SNF subtype protein-loss in relation to overall survival and CCA subtype was analyzed separately (data not shown). For this analysis other underlying SWI/SNF alterations were excluded. ARID1a protein loss correlated with lower overall survival significantly (P=0.025). Median overall survival of the CCA patient cohort with intact expression pattern was 39.4±5.626 months versus 16.125±4.725 months of ARID1a altered patients. Corresponding to the histological subtypes, median survival of ARID1a altered patients was also shorter, whereas only significant for large duct ICCA (P=0.010). For PBRM-1 survival of the cohort with intact expression pattern was longer compared to the altered PRBM-1 patients. However, this finding was non-significant. BRM protein-loss also decreased overall survival in all CCA subtypes, whereas significant results were only detected for the large duct ICCA (P=0.046).

Discussion

In our study we established a cohort of 52 cases of resected CCAs. Consistent with prognostic data given in the literature (26), our patients had a poor median overall survival of 35 months, despite all of them underwent surgery in curative intent. Overall incidence of CCA has increased progressively worldwide over the past four decades (27,28). CCA are heterogeneous tumors with different risk factors and precursor lesions, clinical features and prognosis. Previous studies have documented that their origins form different parts of the biliary tree result in different histopathological and molecular pathological features (6). Within the last few years, new molecular alterations have been discovered in CCA, which are improving the pathological characterization and which might be transformed into personalized targeted therapy algorithms, which are imperatively needed.

We divided our cohort of tumors into the subtypes ECCA, large duct and small duct type ICCA according to a comprehensive analysis of macroscopic and histopathological aspects together with an immunohistochemical and molecular pathological characterization (ECCA, 17; small duct ICCA, 27; large duct ICCA, 8) (10).

In line with literature, our analysis of molecular pathological changes revealed in all CCA subtypes mutations of the tumor suppressor gene P53, with a maximum of 38% in ICCA, large duct type. FISH-analysis for HER2 revealed amplification in all three tumor types with a maximum of 15% in small duct type ICCA. HER2 amplification is a relevant marker, serving as putative therapeutical target with the availability of drugs.

According to other studies, FGFR2 fusions are typically found in ICCA and are present in ~10–16% of patients (29,30). The efficiency of FGFR2 inhibitors is tested to date in several clinical trials in patients with advanced ICCA. Targeted therapies e.g. the pan-selective FGFR kinase inhibitor BGJ398 lately showed promising antitumor activity in a multicenter, open label phase II trial in CCA patients with FGFR2-fusions (26). In our cohort FGFR2-fusions were detected in ICCA with a frequency of 8% in small duct type and 15% in large duct type. As described earlier, FGFR2 translocation were absent in ECCA (29,30).

Recent studies demonstrated somatic mutations of chromatin remodelers in a number of human cancers. Notably, Jiao et al documented mutations in at least one chromatin-remodeling gene in 47% of CCA (20). Mutations in the genes coding for ARID1a-, BRG1-, BRM-, PBRM-1and INI-1 are inactivating and result in a loss of protein expression. Previous studies demonstrated that loss of protein expression correlates strongly with a mutational status in these genes (31).

In our analysis, 35% of the cases showed protein loss in at least one of the complex-proteins, suggesting inactivating mutations in the respective gene. ARID1a was the most frequent lost SWI/SNF protein, followed by BRM. ARID1a- and BRM-negative cases were found in a small percentage of all tumor subtypes. In literature, alterations in the ARID1a gene were detected in 7.2–36% of ICCAs and 5%-12.3% of ECCAs (32). In this aspect, our results are in line with previous studies. The absence of ARID1a in the different subtypes of biliary carcinoma appears to reflect similar mechanisms of carcinogenesis in the different subtypes of CCA.

In our study, BRG1 and INI-1 protein loss was only seen in the group of small duct ICCA, while PRMB-1 protein loss was found in small duct type ICCA and ECCA. Luchini et al analyzed PBRM-1 loss in large duct and small duct ICCA and found a prevalence of 20–30% in both subtypes (33). In our study we did not find cases with PRBM-1 loss in the group of large duct type ICCA, which might be explained by the low number of cases of this subtype (n=8). Within the whole group 7.6% of cases showed protein loss of PBRM-1.

There is still controversy about the loss of proteins of the SWI/SNF complex subunits and its correlation to prognosis of CCA. In the study of Jiao et al, there was no correlation between overall survival and ARID1a alteration (20). This was in line with other studies (32,34). However, they subdivided ECCA and ICCA and did not look at small duct and large duct type ICCA separately, which might be an explanation for the varying results, compared to our data. We identified that median survival of ARID1a altered patients was reduced, but it was only significant for large duct ICCA. Sarcognato et al saw in their cohort of ICCA a correlation with retained protein expression of PBRM-1 and longer overall survival and disease-free survival (35). Other results presented by Misumi et al, demonstrated PBRM1 protein loss in both, small-duct type and large-duct type ICCA. However, it was not associated significantly with any specific characteristics, including prognosis (36).

Our current data give strong evidence, that IHC proven protein-loss of SWI/SNF core subunits (namely ARID1A-, BRG1-, BRM-, PBRM-1and INI1) is associated with a highly significant worse survival of small duct and large duct ICCA, whereas no significant change in survival for ECCA was detected. Furthermore, significant worse overall survival could also be shown for the SWI/SNF-altered cohort after exclusion of HER2-amplificated, FGFR2-translocated or P53 altered patients, which was partly observed in coincidence. These data underline the importance of this chromatin remodeler complex in tumorigenesis of CCA. Our results strengthen preceding data, that the SWI/SNF complex and its core subunits is worth further investigation for targeted therapy options as well as predictive marker for ICCA in future (16,20,21).

Furthermore, several studies suggest a possible increased therapeutic vulnerability of ARID1a-deficient carcinoma. Shen et al were able to show in cell culture analyses and in mouse models that therapeutic inhibition of the enzyme poly-ADP-ribose polymerase (PARP) is effective in ARID1a-deficient ovarian and colon carcinoma (37). ARID1a-deficient carcinoma cells also show activation of the PIk3 pathway. Therapeutic inhibition of this pathway could also be promising in this tumor subgroup. Lee et al successfully inhibited ARID1a-deficient gastric cancer cells with AKT inhibitors (38). Several studies indicate an increased expression of the programmed cell death-ligand 1 (PD-L1) in ARID1a deficient tumor cells. First results show a significantly better response to PD1-/PD-L1 blockade in this setting than in ARID1a intact tumors (39). The vast majority of studies focus on the loss of function of ARID1a. There are no reliable findings as to whether the loss of function of the other SWI/SNF subunits investigated offers comparable therapeutic intervention options. This should be the subject of future clinical studies.

However, our study has some limitations. As a monocentric study, the case number is limited. The statistical analysis gives interesting results and should be confirmed by the analysis of a larger, separate and prospective cohort.

In conclusion, our data prove that CCAs are heterogeneous tumors with different immunohistochemical and molecular marker profiles. The proteins of the SWI/SNF complex are lost in 35% of cases with an impact on prognosis. The core subunits of SWI/SNF, with ARID1A and PBRM-1 for small duct ICCA and ARID1a and BRM for large duct ICCA leading the way, may be possible promising predictive and therapeutic targets and worth further investigation in CCA patients.

Acknowledgements

Mrs. Wiebke Jeske, medical technical assistant, and Ms. Elke Binot, biological- technical assistant (Institute of Pathology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany) produced the TMA samples and performed the hybridization steps for FISH analysis, respectively.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

BJW, RB, AQ and UD designed the study, planned the study and were the primary writers. PSP, KA, MS, DS, DB and SB contributed to the study design. BJW, AQ and UD conducted and evaluated histopathological and immunohistochemical analyses. BJW conducted the FISH analysis. PSP performed statistical analysis. PSP, MS, DS, DB, and SB chose, collected and assembled the clinical data, including patient follow-up from 2000 to 2019. BJW, UD and KA chose and prepared the samples, and collected and assembled pathological data. BJW, AQ and UD performed clinical-pathological data analysis and interpretated the results related to the CCA subtype and clinical implications. BJW and UD confirm the authenticity of all raw data. All authors participated in writing, and read and approved the final manuscript.

Ethics approval and consent to participate

The present retrospective study was conducted with the approval of the Ethics Committee of the University of Cologne, utilizing clinical follow-up data that was collected retrospectively according to a standardized protocol (application 18–269). All patients gave their appropriate informed consent to the procedure.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

CCA

cholangiocarcinoma

CTX

chemotherapy

ECCA

extrahepatic cholangiocarcinoma

ICCA

intrahepatic cholangiocarcinoma

IHC

immunohistochemistry/immunohistochemical

n

number of patients

RTX

radiotherapy

SWI/SNF

SWI/SNF-complex

References

1 

Blechacz B, Komuta M, Roskams T and Gores GJ: Clinical diagnosis and staging of cholangiocarcinoma. Nat Rev Gastroenterol Hepatol. 8:512–522. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Everhart JE and Ruhl CE: Burden of digestive diseases in the United States Part III: Liver, biliary tract, and pancreas. Gastroenterology. 136:1134–1144. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Bridgewater J, Galle PR, Khan SA, Llovet JM, Park JW, Patel T, Pawlik TM and Gores GJ: Guidelines for the diagnosis and management of intrahepatic cholangiocarcinoma. J Hepatol. 60:1268–1289. 2014. View Article : Google Scholar : PubMed/NCBI

4 

DeOliveira ML, Cunningham SC, Cameron JL, Kamangar F, Winter JM, Lillemoe KD, Choti MA, Yeo CJ and Schulick RD: Cholangiocarcinoma: Thirty-one-year experience with 564 patients at a single institution. Ann Surg. 245:755–762. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Rizvi S, Khan SA, Hallemeier CL, Kelley RK and Gores GJ: Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol. 15:95–111. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Komuta M, Govaere O, Vandecaveye V, Akiba J, Van Steenbergen W, Verslype C, Laleman W, Pirenne J, Aerts R, Yano H, et al: Histological diversity in cholangiocellular carcinoma reflects the different cholangiocyte phenotypes. Hepatology. 55:1876–1888. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Akita M, Fujikura K, Ajiki T, Fukumoto T, Otani K, Azuma T, Itoh T, Ku Y and Zen Y: Dichotomy in intrahepatic cholangiocarcinomas based on histologic similarities to hilar cholangiocarcinomas. Mod Pathol. 30:986–997. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Liau JY, Tsai JH, Yuan RH, Chang CN, Lee HJ and Jeng YM: Morphological subclassification of intrahepatic cholangiocarcinoma: Etiological, clinicopathological, and molecular features. Mod Pathol. 27:1163–1173. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Aishima S and Oda Y: Pathogenesis and classification of intrahepatic cholangiocarcinoma: Different characters of perihilar large duct type versus peripheral small duct type. J Hepatobiliary Pancreat Sci. 22:94–100. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Nagtegaal ID, Odze RD, Klimstra D, Paradis V, Rugge M, Schirmacher P, Washington KM, Carneiro F and Cree IA; WHO Classification of Tumours Editorial Board, : The 2019 WHO classification of tumours of the digestive system. Histopathology. 76:182–188. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Gupta A and Dixon E: Epidemiology and risk factors: Intrahepatic cholangiocarcinoma. Hepatobiliary Surg Nutr. 6:101–104. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Shain AH and Pollack JR: The spectrum of SWI/SNF mutations, ubiquitous in human cancers. PLoS One. 8:e551192013. View Article : Google Scholar : PubMed/NCBI

13 

Schallenberg S, Bork J, Essakly A, Alakus H, Buettner R, Hillmer AM, Bruns C, Schroeder W, Zander T, Loeser H, et al: Loss of the SWI/SNF-ATPase subunit members SMARCF1 (ARID1A), SMARCA2 (BRM), SMARCA4 (BRG1) and SMARCB1 (INI1) in oesophageal adenocarcinoma. BMC Cancer. 20:122020. View Article : Google Scholar : PubMed/NCBI

14 

Numata M, Morinaga S, Watanabe T, Tamagawa H, Yamamoto N, Shiozawa M, Nakamura Y, Kameda Y, Okawa S, Rino Y, et al: The clinical significance of SWI/SNF complex in pancreatic cancer. Int J Oncol. 42:403–410. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Hodges C, Kirkland JG and Crabtree GR: The Many Roles of BAF (mSWI/SNF) and PBAF Complexes in Cancer. Cold Spring Harb Perspect Med. 6:62016. View Article : Google Scholar

16 

Simbolo M, Fassan M, Ruzzenente A, Mafficini A, Wood LD, Corbo V, Melisi D, Malleo G, Vicentini C, Malpeli G, et al: Multigene mutational profiling of cholangiocarcinomas identifies actionable molecular subgroups. Oncotarget. 5:2839–2852. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Reisman D, Glaros S and Thompson EA: The SWI/SNF complex and cancer. Oncogene. 28:1653–1668. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Phelan ML, Sif S, Narlikar GJ and Kingston RE: Reconstitution of a core chromatin remodeling complex from SWI/SNF subunits. Mol Cell. 3:247–253. 1999. View Article : Google Scholar : PubMed/NCBI

19 

Pulice JL and Kadoch C: Composition and Function of Mammalian SWI/SNF Chromatin Remodeling Complexes in Human Disease. Cold Spring Harb Symp Quant Biol. 81:53–60. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Jiao Y, Pawlik TM, Anders RA, Selaru FM, Streppel MM, Lucas DJ, Niknafs N, Guthrie VB, Maitra A, Argani P, et al: Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nat Genet. 45:1470–1473. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Simbolo M, Vicentini C, Ruzzenente A, Brunelli M, Conci S, Fassan M, Mafficini A, Rusev B, Corbo V, Capelli P, et al: Genetic alterations analysis in prognostic stratified groups identified TP53 and ARID1A as poor clinical performance markers in intrahepatic cholangiocarcinoma. Sci Rep. 8:71192018. View Article : Google Scholar : PubMed/NCBI

22 

Wilson BG and Roberts CW: SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer. 11:481–492. 2011. View Article : Google Scholar : PubMed/NCBI

23 

Helbig D, Ihle MA, Pütz K, Tantcheva-Poor I, Mauch C, Büttner R and Quaas A: Oncogene and therapeutic target analyses in atypical fibroxanthomas and pleomorphic dermal sarcomas. Oncotarget. 7:21763–21774. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Simon R, Mirlacher M and Sauter G: Tissue microarrays. Methods Mol Med. 114:257–268. 2005.PubMed/NCBI

25 

Wolff AC, Hammond ME, Hicks DG, Dowsett M, McShane LM, Allison KH, Allred DC, Bartlett JM, Bilous M, Fitzgibbons P, et al American Society of Clinical Oncology; College of American Pathologists, : Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update. J Clin Oncol. 31:3997–4013. 2013. View Article : Google Scholar : PubMed/NCBI

26 

Javle M, Lowery M, Shroff RT, Weiss KH, Springfeld C, Borad MJ, Ramanathan RK, Goyal L, Sadeghi S, Macarulla T, et al: Phase II Study of BGJ398 in Patients With FGFR-Altered Advanced Cholangiocarcinoma. J Clin Oncol. 36:276–282. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Khan SA, Taylor-Robinson SD, Toledano MB, Beck A, Elliott P and Thomas HC: Changing international trends in mortality rates for liver, biliary and pancreatic tumours. J Hepatol. 37:806–813. 2002. View Article : Google Scholar : PubMed/NCBI

28 

Saha SK, Zhu AX, Fuchs CS and Brooks GA: Forty-Year Trends in Cholangiocarcinoma Incidence in the U.S.: Intrahepatic Disease on the Rise. Oncologist. 21:594–599. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Graham RP, Barr Fritcher EG, Pestova E, Schulz J, Sitailo LA, Vasmatzis G, Murphy SJ, McWilliams RR, Hart SN, Halling KC, et al: Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma. Hum Pathol. 45:1630–1638. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Wu YM, Su F, Kalyana-Sundaram S, Khazanov N, Ateeq B, Cao X, Lonigro RJ, Vats P, Wang R, Lin SF, et al: Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discov. 3:636–647. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Rao Q, Xia QY, Wang ZY, Li L, Shen Q, Shi SS, Wang X, Liu B, Wang YF, Shi QL, et al: Frequent co-inactivation of the SWI/SNF subunits SMARCB1, SMARCA2 and PBRM1 in malignant rhabdoid tumours. Histopathology. 67:121–129. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Sasaki M, Nitta T, Sato Y and Nakanuma Y: Loss of ARID1A Expression Presents a Novel Pathway of Carcinogenesis in Biliary Carcinomas. Am J Clin Pathol. 145:815–825. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Luchini C, Robertson SA, Hong SM, Felsenstein M, Anders RA, Pea A, Nottegar A, Veronese N, He J, Weiss MJ, et al: PBRM1 loss is a late event during the development of cholangiocarcinoma. Histopathology. 71:375–382. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, Zuo M, Zinner R, Hong D, Meric-Bernstam F, et al: Mutation profiling in cholangiocarcinoma: Prognostic and therapeutic implications. PLoS One. 9:e1153832014. View Article : Google Scholar : PubMed/NCBI

35 

Sarcognato S, Gringeri E, Fassan M, Di Giunta M, Maffeis V, Guzzardo V, Cillo U and Guido M: Prognostic role of BAP-1 and PBRM-1 expression in intrahepatic cholangiocarcinoma. Virchows Arch. 474:29–37. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Misumi K, Hayashi A, Shibahara J, Arita J, Sakamoto Y, Hasegawa K, Kokudo N and Fukayama M: Intrahepatic cholangiocarcinoma frequently shows loss of BAP1 and PBRM1 expression, and demonstrates specific clinicopathological and genetic characteristics with BAP1 loss. Histopathology. 70:766–774. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Shen J, Peng Y, Wei L, Zhang W, Yang L, Lan L, Kapoor P, Ju Z, Mo Q, Shih IeM, et al: ARID1A Deficiency Impairs the DNA Damage Checkpoint and Sensitizes Cells to PARP Inhibitors. Cancer Discov. 5:752–767. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Lee D, Yu EJ, Ham IH, Hur H and Kim YS: AKT inhibition is an effective treatment strategy in ARID1A-deficient gastric cancer cells. OncoTargets Ther. 10:4153–4159. 2017. View Article : Google Scholar

39 

Shen J, Ju Z, Zhao W, Wang L, Peng Y, Ge Z, Nagel ZD, Zou J, Wang C, Kapoor P, et al: ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat Med. 24:556–562. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wagner BJ, Plum PS, Apel K, Scherer M, Buchner D, Brinkmann S, Buettner R, Stippel D, Quaas A, Drebber U, Drebber U, et al: Protein‑loss of SWI/SNF‑complex core subunits influences prognosis dependent on histological subtypes of intra‑ and extrahepatic cholangiocarcinoma. Oncol Lett 21: 349, 2021
APA
Wagner, B.J., Plum, P.S., Apel, K., Scherer, M., Buchner, D., Brinkmann, S. ... Drebber, U. (2021). Protein‑loss of SWI/SNF‑complex core subunits influences prognosis dependent on histological subtypes of intra‑ and extrahepatic cholangiocarcinoma. Oncology Letters, 21, 349. https://doi.org/10.3892/ol.2021.12610
MLA
Wagner, B. J., Plum, P. S., Apel, K., Scherer, M., Buchner, D., Brinkmann, S., Buettner, R., Stippel, D., Quaas, A., Drebber, U."Protein‑loss of SWI/SNF‑complex core subunits influences prognosis dependent on histological subtypes of intra‑ and extrahepatic cholangiocarcinoma". Oncology Letters 21.5 (2021): 349.
Chicago
Wagner, B. J., Plum, P. S., Apel, K., Scherer, M., Buchner, D., Brinkmann, S., Buettner, R., Stippel, D., Quaas, A., Drebber, U."Protein‑loss of SWI/SNF‑complex core subunits influences prognosis dependent on histological subtypes of intra‑ and extrahepatic cholangiocarcinoma". Oncology Letters 21, no. 5 (2021): 349. https://doi.org/10.3892/ol.2021.12610