Abstract
Purpose
Dedicated gene signatures in small (SD-iCCA) and large (LD-iCCA) duct type intrahepatic cholangiocarcinoma remain unknown. We performed immune profiling in SD- and LD-iCCA to identify novel biomarker candidates for personalized medicine.
Methods
Retrospectively, 19 iCCA patients with either SD-iCCA (n = 10, median age, 63.1 years (45–86); men, 4) or LD-iCCA (n = 9, median age, 69.7 years (62–85); men, 5)) were included. All patients were diagnosed and histologically confirmed between 04/2009 and 01/2021. Tumor tissue samples were processed for differential expression profiling using NanoString nCounter® PanCancer Immune Profiling Panel.
Results
With the exception of complement signatures, immune-related pathways were broadly downregulated in SD-iCCA vs. LD-iCCA. A total of 20 immune-related genes were strongly downregulated in SD-iCCA with DMBT1 (log2fc = -5.39, p = 0.01) and CEACAM6 (log2fc = -6.38, p = 0.01) showing the strongest downregulation. Among 7 strongly (log2fc > 2, p ≤ 0.02) upregulated genes, CRP (log2fc = 5.06, p = 0.02) ranked first, and four others were associated with complement (C5, C4BPA, C8A, C8B). Total tumor-infiltrating lymphocytes (TIL) signature was decreased in SD-iCCA with elevated ratios of exhausted-CD8/TILs, NK/TILs, and cytotoxic cells/TILs while having decreased ratios of B-cells/TILs, mast cells/TILs and dendritic cells/TILs. The immune profiling signatures in SD-iCCA revealed downregulation in chemokine signaling pathways inclulding JAK2/3 and ERK1/2 as well as nearly all cytokine-cytokine receptor interaction pathways with the exception of the CXCL1/CXCR1-axis.
Conclusion
Immune patterns differed in SD-iCCA versus LD-iCCA. We identified potential biomarker candidate genes, including CRP, CEACAM6, DMBT1, and various complement factors that could be explored for augmented diagnostics and treatment decision-making.
Supplementary Information
The online version contains supplementary material available at 10.1007/s00432-024-05888-y.
Keywords: Biomarker; Cholangiocarcinoma; Intrahepatic; Immune profiling; Pathology, molecular; Surgical oncology
Introduction
Cholangiocarcinoma (CCA) is a highly heterogeneous malignancy originating from the intrahepatic biliary epithelium (iCCA) or from extrahepatic bile ducts (eCCA). Patients with CCA have a poor prognosis and the incidence of iCCA is rising globally, accounting for about 10–15% of primary liver cancers (Bertuccio et al. 2013; Bridgewater et al. 2014). With surgical resection being the sole curative treatment option, the prognosis for iCCA patients remains unfavorable (Groot Koerkamp and Fong 2014). The new standard of care in the palliative setting is the combination of chemotherapy with gemcitabine and cisplatin and the immune checkpoint inhibitors durvalumab or pembrolizumab, leading to a median overall survival of 12.8 and 12.7 months, respectively (Oh et al. 2022; Kelley et al. 2023).
In recent years, histopathological characterization of iCCA revealed two distinct subtypes according to the size of the affected bile duct, which led to implementation in the WHO classification (WHO Classification of Tumours 2019, 5th ed. Vol. 1. Digestive System Tumours, 2019. [Online]. Available: https://publications.iarc.fr/ Book-And-Report-Series/ Who-Classification-Of- Tumours/Digestive-System-Tumours-2019.'; Kendall et al. 2019; Aishima and Oda 2015). On the one hand, small duct type iCCA (SD-iCCA) was found to be more peripheral in the liver and resembling a ductular and cholangiolar type (Liau et al. 2014; Chung and Park 2022). On the other hand, the large duct type (LD-iCCA) arises from large intrahepatic ducts closer to the liver hilum and contains mainly mucin-producing columnar tumor cells (Hayashi et al. 2016; Sigel et al. 2018; Chung and Park 2022). Remarkably, both subtypes differ in underlying diseases, survival, response to chemotherapy, and molecular alterations, emphasizing clinically relevant subtype heterogeneity (Kinzler et al. 2022; Aishima and Oda 2015; Chung et al. 2020; Kendall et al. 2019; Gerber et al. 2022).
Immunotherapy emerged in the last decade and revolutionized treatments and outcomes across multiple cancer entities, including CCA (Pan et al. 2020; Greten et al. 2023; Fiste et al. 2021). Although the results of the TOPAZ-1 and KEYNOTE-966 trial have opened new perspectives for palliative CCA patients (Oh et al. 2022; Kelley et al. 2023), immunotherapeutic approaches in the management of iCCA patients remain challenging as the immunosuppressive tumor microenvironment (TME) plays a pivotal role in iCCA progression and, thereby, potential response to immunotherapeutic agents (Greten et al. 2023; Banales et al. 2020). The highly reactive TME comprises a variety of immune cells, including cancer-associated fibroblasts, tumor-associated macrophages, endothelial cells, and lymphocytes (Xia et al. 2022b; Job et al. 2020; Banales et al. 2020), but detailed characteristics are lacking. Here, improved characterization of the immune landscape in iCCA holds substantial clinical potential, both for predicting response to immunotherapy and for identifying novel treatment strategies. A few preliminary studies investigated immune signatures as predictive biomarkers in iCCA (Xia et al. 2022b; Yoon et al. 2021; Konishi et al. 2022; Yugawa et al. 2021; Jing et al. 2019). However, differences in immune signatures between SD- and LD-iCCA remain unknown, and exploratory studies are lacking so far.
We hypothesized that the heterogeneity of iCCA subtypes is reflected in their immune patterns and that these differences could hold a significant potential for diagnostic and therapeutic personalized medicine.
Materials and methods
Patient cohort
All patients treated with surgically resected (R0, R1) intrahepatic cholangiocarcinoma at Frankfurt University Hospital between December 2005 and December 2021 were retrospectively screened. Clinical data (sex, date of birth, tumor stage, tumor size, laboratory parameters, and comorbidities) were collected from electronic medical records. iCCA were staged according to the 8th edition of the classification of the Union for International Cancer Control (UICC). Tissue samples used in this study were provided by the University Cancer Center Frankfurt (UCT). Written informed consent was obtained from all patients at the time of initial surgery, or written informed consent was waived if the patient was deceased. The study was approved by the institutional Review Boards of the UCT and the Ethical Committee at the University Hospital Frankfurt (project-number: SGI-1-2021, SGI-3-2021).
Study design
Hematoxylin and eosin (HE) slides and formalin-fixed paraffin-embedded (FFPE) tissue were retrieved from the archive of the Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt. According to the WHO Classification of Tumours, Digestive System Tumours, 5th edition, Volume 1 ('WHO Classification of Tumours 2019, 5th ed. Vol. 1. Digestive System Tumours, 2019. [Online]. Available: https://publications.iarc.fr/ Book-And-Report-Series/ Who-Classification-Of- Tumours/Digestive-System-Tumours-2019.'), all samples examined in the present study were recently analyzed both histomorphologically and immunohistochemically by an expert hepatobiliary pathologist and assigned to the respective subtype in our previously published work (Kinzler et al. 2022). Retrospectively, 20 iCCA-patients (n = 10, SD-iCCA and n = 10, LD-iCCA) who were diagnosed between 04/2009 and 01/2021, were included in the present study. Inclusion criteria: (1) Histologically confirmed treatment-naïve SD-iCCA or LD-iCCA. Exclusion criteria: (1) insufficient tissue sample / RNA -/ or NanoString® quality or quantity. To gain equal distribution of SD- and LD-iCCA in our study, the number of included SD-iCCA samples was adjusted to the less frequent LD-iCCA type. Figure 1 depicts the flowchart of patient inclusion according to Standards for Reporting Diagnostic Accuracy Studies (STARD).
Fig. 1.
STARD Flowchart of patient inclusion into the study. STARD Standards for Reporting Diagnostic Accuracy Studies
Ribonucleic acid (RNA) isolation and immune profiling analysis
Representative tumor material was punched out of FFPE blocks using a 1 mm core needle. RNA was isolated using the truXTRAC FFPE total NA Kit (Covaris, Woburn, MA, USA) based on focused ultrasonification and column purification according to the manufacturer's instructions. NanoString nCounter® Platform and PanCancer Immune Profiling Panel were used to enrich a commercially available function specific panel of 770 genes by hybrid capture technique (NanoString®, Seattle, WA, USA) as previously published (Kinzler et al. 2023). NanoString nSolver™ software v4 and implemented nCounter® Advanced Analysis module v2.0.134 were used for subsequent raw data processing and normalization by internal controls following differential supervised analysis of SD-iCCA (n = 10) versus baseline of LD-iCCA (n = 9). Quality control was done with default settings as previously described (Preusse et al. 2021). One sample (LD-iCCA) was flagged in quality control as the percentage of successfully scanned fields of view in its cartridge lane was below the threshold of 75%, and we excluded this sample for further analysis. Gene expression of LD-iCCA was set as baseline for the comparative analysis. For pathway analysis of differentially expressed genes, Enrichr (Chen et al. 2013; Kuleshov et al. 2016) was used for functional enrichment analysis for Gene Ontology to identify gene-sets for biological processes. For further differential expression analysis, we used the following cut-offs: log2 fold change < -2 or > 2 and p-Value ≤ 0.05 after Benjamini-Hochberg (BH) correction. T-distributed stochastic neighbor embedding (t-SNE) analysis and plots were performed in Python 3.7.6.
Statistical analysis
We compared baseline clinicopathological characteristics between patients with SD- and LD-iCCA. For statistical analysis, two-sided Students t-test was used for continuous variables and Likelihood Ratio for nominal / ordinal data.
Results
Study population
In total, 19 patients with iCCA were included in this study. Ten patients with SD-iCCA (median age, 63.1 years (45–86); men, 4) and nine patients with LD-iCCA (median age, 69.7 years (62–85); men, 5) were analyzed. The groups did not differ in clinicopathological characteristics including tumor size, the occurrence of multiple tumors, UICC stadium, performance status or selected laboratory values like CA-19/9, bilirubin, or lactate dehydrogenase (Table 1). Interestingly, patients with LD-iCCA were more likely to have hepatolithiasis (p = 0.049). However, patients with SD- and LD-iCCA did not differ in any other common risk factors including viral hepatitis, primary sclerosing cholangitis or liver cirrhosis. Further clinical characteristics are depicted in Table 1.
Table 1.
Clinical and epidemiological characteristics
| Characteristics | SD-iCCA (n = 10), No. (%) | LD-iCCA (n = 9), No. (%) | p-value |
|---|---|---|---|
| Sex | 0.525 | ||
| Female | 6 (60) | 4 (44.4) | |
| Male | 4 (40) | 5 (55.6) | |
| Age at initial diagnosis | 0.172 | ||
| Mean, years, (range) | 63.1 (45–86) | 69.7 (62–85) | |
| UICC | 0.695 | ||
| 1a | 1 (10) | 2 (22.2) | |
| 1b | 3 (30) | 2 (22.2) | |
| 2 | 3 (30) | 1 (11.1) | |
| 3a | 1 (10) | 0 (0) | |
| 3b | 2 (20) | 3 (33.3) | |
| 4 | 0 (0) | 1 (11.1) | |
| ECOG | 0.72 | ||
| 0 | 7 (70) | 7 (77.8) | |
| 1 | 3 (30) | 2 (22.2) | |
| CA-19/9 (ng/ml) | 1 | ||
| < 37 | 5 (50) | 4 (44.4) | |
| ≥ 37 | 5 (50) | 4 (44.4) | |
| n.a. | 0 (0) | 1 (11.1) | |
| Tumor size (cm) | 0.541 | ||
| ≤ 5 | 3 (30) | 4 (44.4) | |
| > 5 | 7 (70) | 5 (55.6) | |
| Single Tumor | 0.106 | ||
| Yes | 4 (40) | 7 (77.8) | |
| No | 6 (60) | 2 (22.2) | |
| Pathological grade | 0.912 | ||
| Grade 2 | 8 (80) | 7 (77.8) | |
| Grade 3 | 2 (20) | 2 (22.2) | |
| R status | 0.121 | ||
| R0 | 8 (80) | 4 (44.4) | |
| R1 | 2 (20) | 5 (55.6) | |
| L status | 0.183 | ||
| L0 | 9 (90) | 5 (55.6) | |
| L1 | 1 (10) | 3 (33.3) | |
| Lx | 0 (0) | 1 (11.1) | |
| Pn status | 0.104 | ||
| Pn0 | 7 (70) | 3 (33.3) | |
| Pn1 | 2 (20) | 5 (55.6) | |
| Pnx | 1 (10) | 1 (11.1) | |
| Recurrence | 0.285 | ||
| Yes | 3 (30) | 5 (55.6) | |
| No | 7 (70) | 4 (44.4) | |
| Hepatolithiasis | 0.049 | ||
| Yes | 0 (0) | 3 (33.3) | |
| No | 10 (100) | 6 (66.7) | |
| Viral hepatitis | 0.305 | ||
| Yes | 0 (0) | 1 (11.1) | |
| No | 10 (100) | 8 (88.9) | |
| PSC | 0.357 | ||
| Yes | 1 (10) | 0 (0) | |
| No | 9 (90) | 9 (100) | |
| Diabetes | 0.884 | ||
| Yes | 3 (30) | 3 (33.3) | |
| No | 7 (70) | 6 (66.7) | |
| Liver cirrhosis | 0.305 | ||
| Yes | 0 (0) | 1 (11.1) | |
| No | 10 (100) | 8 (88.9) | |
| LDH | |||
| < 248 | 5 (50) | 4 (44.4) | 0.953 |
| ≥ 248 | 4 (40) | 3 (33.3) | |
| n.a. | 1 (10) | 2 (22.2) | |
| Bilirubin | 0.063 | ||
| < 1.4 | 9 (90) | 6 (66.7) | |
| ≥ 1.4 | 0 (0) | 3 (33.3) | |
| n.a. | 1 (10) | 0 (0) |
For statistical analysis, two-sided Students t-test was used for continuous variables and Likelihood Ratio for nominal / ordinal data. Data is shown as absolute numbers (%) or median (min–max)
CA-19/9 carbohydrate antigen 19-9, ECOG Eastern Cooperative Oncology Group, LDH lactate dehydrogenase, LD-iCCA large duct type intrahepatic cholangiocarcinoma, n.a. not available, UICC Union for International Cancer Control, SD-iCCA small duct type intrahepatic cholangiocarcinoma
Immune-pathway scores were downregulated in SD-iCCA with the exception of complement signatures
To explore if patients with SD- and LD-iCCA differed in their immune cell signatures, we first performed unsupervised T-SNE analysis and found a clear separation into two groups (Fig. 2a). Next, we used pathway score analyses of the functionally annotated genes to explore distinct patterns. Here we found that the majority of pathways in patients with SD-iCCA were downregulated, especially pathways associated with regulation, cell function and adhesion, with the exception of complement signatures that were upregulated (Fig. 2b,c).
Fig. 2.
Dominant downregulation in immune pathway scores in small-duct type intrahepatic cholangiocarcinoma. a T-SNE plot of comprehensive log2 normalized mRNA patient data, b trend plot of pathway signatures using NanoString® pathway score analysis tool and c boxplots of the top 4 differentially expressed pathway score signatures
SD- and LD-iCCA revealed strongly differentially regulated candidate genes
Small- and large-duct type iCCA showed strong differences in the expression of immune-related genes (Fig. 3a). In total, 27 genes were strongly differentially expressed (log2fc > 2 or log2fc < -2, BH-p < 0.05) as depicted in Fig. 3b. CRP showed the strongest upregulation (log2fc = 5.06, p = 0.02, 95% CI [2.47–7.66]) in SD-iCCA. 57% (4/7) of the 7 upregulated genes in SD-iCCA were associated with complement, namely C5 (log2fc = 3.80, p = 0.003, 95% CI [2.67–4.93]), C4BPA (log2fc = 3.67, p = 0.01, 95% CI [2.18–5.17]), C8A (log2fc = 3.36, p = 0.02, 95% CI [1.71–5.00]), C8B (log2fc = 3.2, p = 0.01, 95% CI [1.81–4.59]). Most genes were strongly downregulated in SD-iCCA versus LD-iCCA, and the strongest downregulation was seen in carcinoembryonic antigen-related cellular adhesion molecule 6 (CEACAM6) (log2fc = – 6.38, p = 0.01, 95% CI [– 9.14 to – 3.62]), deleted in malignant brain tumor 1 (DMBT1) (log2fc = – 5.39, p = 0.01, 95% CI [– 7.88 to – 2.89]) and CD79A (log2fc = – 3.61, p = 0.01, 95% CI [– 5.18 to – 2.04]). See Table 2.
Fig. 3.
Differentially expressed genes between intrahepatic cholangiocarcinoma subtypes. a Volcano plot of all differentially expressed genes and only b strong and significantly (log2fc < -2 or > 2 and p-value < 0.05 with Benjamini–Hochberg correction)
Table 2.
Strongly differentially expressed genes in SD-iCCA vs. LD-iCCA
| Log2fc | 95% CI | p-value | BH p-value | Gene sets | |
|---|---|---|---|---|---|
| CRP | 5.06 | 2.47/7.66 | 0.001 | 0.024 | Transporter functions |
| C5 | 3.80 | 2.67/4.93 | < 0.001 | 0.003 | Complement |
| C4BPA | 3.67 | 2.18/5.17 | < 0.001 | 0.011 | Complement |
| SAA1 | 3.67 | 2.06/5.28 | < 0.001 | 0.012 | |
| LBP | 3.51 | 2.07/4.95 | < 0.001 | 0.011 | Macrophage functions |
| C8A | 3.36 | 1.71/5.00 | 0.001 | 0.020 | Complement |
| C8B | 3.20 | 1.81/4.59 | < 0.001 | 0.012 | Complement |
| SLC11A1 | – 2.12 | – 3.26/– 0.98 | 0.002 | 0.027 | Macrophage functions |
| DUSP4 | – 2.19 | – 3.12/– 1.26 | < 0.001 | 0.012 | |
| TNFRSF11A | – 2.23 | – 2.95/– 1.50 | < 0.001 | 0.003 | TNF superfamily |
| IL1RN | – 2.23 | – 3.58/– 0.872 | 0.005 | 0.048 | Cytokines, Interleukins |
| POU2AF1 | – 2.26 | – 3.25/– 1.26 | < 0.001 | 0.013 | |
| CD79B | – 2.38 | – 3.62/– 1.14 | 0.002 | 0.024 | B-cell functions |
| TNFRSF13C | – 2.40 | – 3.82/– 0.966 | 0.005 | 0.047 | Regulation, TNF superfamily |
| PTGS2 | – 2.53 | – 3.79/– 1.27 | 0.001 | 0.022 | Cytokines |
| SMPD3 | – 2.55 | – 3.62/– 1.48 | < 0.001 | 0.012 | Cell functions |
| LCN2 | – 2.63 | – 3.88/– 1.38 | 0.001 | 0.017 | |
| CD19 | – 2.65 | – 4.08/– 1.22 | 0.002 | 0.028 | B-cell functions, Regulation |
| CEACAM1 | – 2.76 | – 4.02/– 1.49 | 0.001 | 0.014 | Adhesion |
| OSM | – 2.79 | – 4.01/– 1.58 | < 0.001 | 0.012 | Cell functions |
| PPARG | – 2.89 | – 3.84/– 1.94 | < 0.001 | 0.003 | |
| IL11 | – 2.96 | – 4.7/– 1.23 | 0.004 | 0.040 | B-cell functions, cytokines, interleukins, T-cell functions |
| LTF | – 2.98 | – 4.19/– 1.76 | < 0.001 | 0.011 | |
| IL1B | – 3.01 | – 4.49/– 1.54 | 0.001 | 0.020 | Chemokines, Cytokines, Interleukins, Pathogen Defense, Regulation |
| CD79A | – 3.61 | – 5.18/– 2.04 | < 0.001 | 0.012 | |
| DMBT1 | – 5.39 | – 7.88/– 2.89 | 0.001 | 0.014 | |
| CEACAM6 | -6.38 | -9.14 / -3.62 | < 0.001 | 0.012 | Adhesion |
'Estimated log fold-change' estimates a gene's differential expression. For each gene, a single linear regression was fit with all selected covariates for prediction of expression to eliminate measured confounding and isolate the independent associations. The log2 fold change is presented, along with a p-value, an adjusted p-value or FDR (BH correction) and the 95% CI
BH Benjamini–Hochberg, FDR false discovery rate
Cell type profiling signatures revealed two immune type subsets
Total TILs and the absolute amount of the vast majority of cell type profiles were reduced in SD-iCCA (Fig. 4a). The relative cell type to TIL profile ratios revealed the most prominent decreases in B cells/TILs, mast cells/TILs and dendritic cells(DC)/TILs (Fig. 4b,c). Increased relative cell type profile ratios were revealed for exhausted CD8/TILs, cytotoxic cells/TILs and NK cells/TILs (Fig. 4b, d).
Fig. 4.
Cell type profiling revealed decreased immune infiltrates in SD-iCCA. Cell population abundance was measured based on characteristically expressed genes. The cell type abundance measurements are plotted against the tumor subtypes. a Total cell type scores and b the relative cell type scores. c, d Top differentially expressed cell type scores relative to total TILs c with downregulation in small-duct type iCCA or d upregulation in SD-iCCA
Gene ontology term enrichment analysis
Applying Enrichr gene ontology term enrichment on the whole differentially expressed dataset, the biological processes were mostly related to a downregulation in the subtotal cytokine-cytokine receptor interaction signatures (Suppl. Fig. 1). Only the CX3CL1/CX3CR1-axis of the CX3C subfamily and TGF-beta2 were upregulated in SD-iCCA (Suppl. Fig. 1). Also, chemokine signaling pathways were downregulated in SD-iCCA, a.o. JAK2/3, PI3K, ERK1/2 and PKC (Suppl. Fig. 2).
Discussion
By applying NanoString® technology, we exploratorily identified substantial differences in the local immune patterns of patient with SD- and LD-iCCA, including signatures of inflammation and immune response. By analyzing treatment-naïve tumor samples, our results suggest that the immune signatures are an intrinsic trait of the tumor types. Further, we corroborated our results by performing a multitude of complementary NanoString® technology analysis like pathway scoring, gene set enrichment analysis, differential expression analysis and cell type profiling to create a holistic model of the immune signatures in both iCCA subtypes. Considering the rapid emergence of immune-oncology diagnostics and treatment, our results provide insights and give evidence which can be used for biomarker discovery and to inform future studies with therapeutic intend before clinical translation.
Our data indicate that immune-related pathways are broadly downregulated in SD-iCCA, with the exception of complement signatures. Among 27 strongly differently expressed genes, 20 were downregulated in SD-iCCA with CEACAM6, DMBT1 and CD79A showing the strongest effect. From the 7 upregulated genes, CRP showed the highest differential expression and the complement factors C5, C4BPA, C8A and C8B comprised 57% of the upregulated genes. Remarkably, total TIL signatures were reduced in SD-iCCA. Cell type signatures differed in both iCCA subtypes and chemokine as well as cytokine-cytokine receptor interaction pathways were broadly downregulated in SD-iCCA.
In recent years, the nihilistic approach to the treatment of iCCA has been replaced by new therapies in the field of molecular and immunotherapeutic regimens. However, the heterogeneity of both iCCA subtypes and the lack of predictive biomarkers remain a major challenge in determining which patient subgroup will benefit from immunotherapy in order to provide stratified medicine. In addition to the known heterogeneity of SD- and LD-iCCA in terms of survival, response to chemotherapy, and molecular alterations, this study is the first to address differences in immune patterns. Our data demonstrate a downregulation of DMBT1 in SD-iCCA compared to LD-iCCA. DMBT1 is a mucin-like molecule that exerts functions in the regulation of epithelial differentiation and inflammation participating in mucosal immune defense (Mollenhauer et al. 2000; Bathum Nexoe et al. 2020). So far, only two studies investigated the role of DMBT1 in iCCA. Goeppert and colleagues did not observe differences in DMBT1 expression in iCCA compared to normal biliary tissue while they identified a significant decrease of DMBT1 expression in iCCA compared to biliary intraepithelial neoplasia (BilIN) 3 suggesting a tumor suppressing role of DMBT1 in early cholangiocarcinogenesis (Goeppert et al. 2017). In line, Sasaki et al. demonstrate a decreased expression of DMBT1 in tissue of invasive iCCA compared to intraductal papillary neoplasms (Sasaki et al. 2003). Against this background, one would rather suspect DMBT1 downregulation in LD-iCCA as this subtype is commonly associated with impaired survival and a more aggressive tumor biology (Kinzler et al. 2022). However, lack of DMBT1 expression in non-neoplastic biliary tissue of CCA patients was associated with poor survival while no significant impact on outcome was observed when DMBT1 expression was reduced in cancer cells (Goeppert et al. 2017). Interestingly, overexpression of DMBT1 was shown in primary sclerosing cholangitis as well as in hepatolithiasis (Bisgaard et al. 2002; Sasaki et al. 2003), two common risk factors for the occurrence of LD-iCCA (Aishima and Oda 2015). This may explain the upregulation of DMBT1 in LD-iCCA as hepatolithiasis was significantly more present in in this cohort in our study. It should be noted that both studies did not differentiate between iCCA subtypes, which may hinder comparability to our data. Further, we found a downregulation of CEACAM6 in SD-iCCA compared to LD-iCCA. CEACAM6 is a member of the immunoglobulin cell adhesion molecule superfamily, and its overexpression is associated with poor prognosis and invasiveness in vivo and in vitro in iCCA (Ieta et al. 2006; Liu et al. 2022; Kurlinkus et al. 2021). In addition, high levels of CEACAM6 are suggested as a screening parameter especially for eCCA (Rose et al. 2016), which is consistent with our data showing overexpression of CEACAM6 in LD-iCCA, as LD-iCCA are generally mucin-secreting tubular adenocarcinomas resembling perihilar and distal CCA (Kendall et al. 2019). Intriguingly, Ieta et al. could demonstrate that CEACAM6 overexpression is associated with chemoresistance to gemcitabine in vitro (Ieta et al. 2006) while two recently published studies revealed significant shorter progression-free survival for LD-iCCA receiving gemcitabine-based chemotherapy (Kinzler et al. 2022; Yoon et al. 2021). Thus, our data suggest that CEACAM6 could serve as a potential chemoresistant marker to gemcitabine especially in patients suffering from LD-iCCA.
Complement proteins, as a part of tumor microenvironment, can play a pivotal role in local immune response in various cancer entities (Roumenina et al. 2019). A recent proteomic analysis demonstrated that complement factors were significantly increased in CCA patients (Son et al. 2020) while it was shown that the reduced expression of complement factor H-related 3 is associated with poor prognosis and immune regulation in CCA patients (Wang et al. 2022). As such, presence of complement factor H-related 3 negatively correlated with tumor infiltrating lymphocytes like CD8 + T cells (Wang et al. 2022). In line with these findings, our data show an upregulation of various complement factors and a concomitant decrease of TIL in SD- compared to LD-iCCA. As part of the adaptive immune system, TIL can either target tumor cells to prevent carcinogenesis and cancer progression, or cancer cells can adopt strategies to evade the immune responses against the cancer, thus promoting tumor progression (Gooden et al. 2011). TIL comprise of highly heterogeneous immune cells, including CD8 + T cells. In iCCA, two studies have shown that an increase in TIL is associated with favorable outcome (Xia et al. 2022a; Yoon et al. 2021) while Goeppert et al. confirmed the prognostic value of TIL only for eCCA, but not for iCCA (Goeppert et al. 2013). For the first time, we could show increased CD8 + /TIL ratios in SD-iCCA compared to LD-iCCA in our study, which is in line with Xia et al. and Yoon et al. as this subtype is generally associated with better overall survival (Kinzler et al. 2022). However, Yoon et al. investigated TIL solely by determining the CD8 + status by immunohistochemistry in a sub cohort of PD-L1-inhibitor treated patients in recurrent or unresectable CCA that underwent upfront chemotherapy, which likely affected the immune landscape (Yoon et al. 2021), while we used NanoString® technology in treatment naïve tumor tissue. Thereby, our results were not potentially cofounded by prior treatments and thereby indicate a tumor-intrinsic trait.
In general, the presence of TIL- and chemokine-infiltrated TME is associated with better response to immune checkpoint blockade in CCA (Binnewies et al. 2018). So far, both iCCA subtypes are traditionally merged in the clinical context and are treated similarly with regard to chemo- and immunotherapeutic approaches. This long-held hypothesis is challenged by our finding that the immune signatures of LD-iCCA comprise higher levels of total TIL and chemokine signaling. Therefore, our results might suggest that this subtype is potentially more responsive to immunotherapy. However, studies investigating the potentially different response to immunotherapy in SD- and LD-iCCA with a possible link to TIL expression are needed.
Our study has several limitations that warrant discussion. We performed a retrospective analysis and selection bias cannot be ruled out. Our study population was small, and generalizability may not be presumed as this study was exploratory and hypothesis-generating in nature. However, we used treatment naïve tumor tissue of surgically resected specimen which ensured that the immune landscape of our samples was not altered due to prior anti-cancer treatments. Given these considerations, the substantial novelty of our data and the fact that data on this topic are lacking so far, the results of the present study are of high clinical relevance. Our aim was to exploratively analyze different immune patterns using NanoString® technology. We anticipate our results to accelerate and inform future work using confirmatory analysis such as immunofluorescence staining, flow cytometry, qPCR, in vitro, and in vivo experiments in prospective studies to augment and validate our results prior to clinical translation.
In conclusion, our study is the first to demonstrate that SD- and LD-iCCA are associated with dedicated local patterns of immune profiles. The substantial differences hold clinically relevant promise for biomarker discovery and treatment planning, especially using immune checkpoint therapy aimed at subtype-specific, personalized medicine. We anticipate our findings to inform future work which is needed to build upon and corroborate our findings, and thereby improve our understanding of iCCA biology for improved diagnostics, and treatment approaches.
Supplementary Information
Below is the link to the electronic supplementary material.
Acknowledgements
S.B., T.J.V., P.J.W. and M.N.K. would like to thank the Frankfurt Cancer Institute (FCI) and the University Cancer Center (UCT) Frankfurt for the support.
Abbreviations
- BiIIN
Biliary intraepithelial neoplasia
- CCA
Cholangiocarcinoma
- CEACAM6
Carcinoembryonic antigen-related cellular adhesion molecule 6
- DMBT1
Deleted in malignant brain tumor 1
- FFPE
Formalin-fixed paraffin-embedded
- HE
Hematoxylin and eosin
- iCCA
Intrahepatic cholangiocarcinoma
- LD-iCCA
Large duct type intrahepatic cholangiocarcinoma
- UCT
University Cancer Center Frankfurt
- UICC
Union for International Cancer Control
- SD-iCCA
Small duct type intrahepatic cholangiocarcinoma
- TIL
Tumor-infiltrating lymphocytes
- TME
Tumor microenvironment
Author Contributions
Conceptualization, S.B., K.B., P.J.W. and M.N.K.; methodology, S.B., F.S., J.B., K.B., A.S. and M.N.K.; data curation, S.B., J.B., K.B., A.S. and M.N.K.; analysis, S.B., F.S., J.B., S.M., L.D.G., V.K. and M.N.K.; interpretation, S.B., S.M., L.D.G., V.K., A.S., D.W., F.F. and M.N.K.; project administration, S.B. and M.N.K.; acquisition, A.A.S; writing–original draft preparation, S.B. and M.N.K.; writing–review and editing, F.S., J.B., K.B., S.M., L.D.G., V.K., A.S., A.A.S, D.W., F.F., S.Z., T.J.V. and P.J.W.; resources, S.B., S.Z., T.J.V. and P.J.W. All authors have read and agreed to the published version of the manuscript.
Funding
Open Access funding enabled and organized by Projekt DEAL. A.S. and M.N.K. were supported by a rotation grant for medical scientists in the Frankfurt Research Promotion Program (FFF) of the Faculty of Medicine of the Goethe University. S.B. and this work were supported in part by the Trusts of the Faculty of Medicine of the Goethe University (“Stiftungen und Vereine—Stiftungsmittel des Fachbereichs”).
Data availability
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Declarations
Conflict of Interest
F. F. has received travel support from Ipsen, Abbvie, Astrazeneca and speaker’s fees from AbbVie, MSD, Ipsen, Astra. S.Z. has received speaker fees and/or honoraria for consultancy from Abbvie, BioMarin, Gilead, Intercept, Janssen, Madrigal, MSD/Merck, NovoNordisk, SoBi and Theratechnologies. P.J.W. has received consulting fees and honoraria for lectures by Bayer, Janssen-Cilag, Novartis, Roche, MSD, Astellas Pharma, Bristol-Myers Squibb, Thermo Fisher Scientific, Molecular Health, Guardant Health, Sophia Genetics, Qiagen, Eli Lilly, Myriad, Hedera Dx, and Astra Zeneca; research support was provided by Astra Zeneca. The authors declare that there is no relationship relevant to the manuscripts’ subject. All other authors declare no conflicts of interest.
Ethics approval
The study was performed in accordance with the Declaration of Helsinki. The study protocol was approved by the local ethics committee of the University of Frankfurt (project-number: SGI-1–2021, SGI-3–2021).
Consent to participate
Patients provided informed written consent and patient data was provided after approval by the local ethics committee.
Footnotes
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References
- Aishima S, Oda Y (2015) 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 [DOI] [PubMed] [Google Scholar]
- Banales JM, Marin JJG, Lamarca A, Rodrigues PM, Khan SA, Roberts LR, Cardinale V, Carpino G, Andersen JB, Braconi C, Calvisi DF, Perugorria MJ, Fabris L, Boulter L, Macias RIR, Gaudio E, Alvaro D, Gradilone SA, Strazzabosco M, Marzioni M, Coulouarn C, Fouassier L, Raggi C, Invernizzi P, Mertens JC, Moncsek A, Rizvi S, Heimbach J, Koerkamp BG, Bruix J, Forner A, Bridgewater J, Valle JW, Gores GJ (2020) Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol 17:557–588 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bathum Nexoe A, Pedersen AA, von Huth S, Detlefsen S, Hansen PL, Holmskov U (2020) Immunohistochemical Localization of Deleted in Malignant Brain Tumors 1 in Normal Human Tissues. J Histochem Cytochem 68:377–387 [DOI] [PubMed] [Google Scholar]
- Bertuccio P, Bosetti C, Levi F, Decarli A, Negri E, La Vecchia C (2013) A comparison of trends in mortality from primary liver cancer and intrahepatic cholangiocarcinoma in Europe. Ann Oncol 24:1667–1674 [DOI] [PubMed] [Google Scholar]
- Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF (2018) Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med 24:541–550 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bisgaard HC, Holmskov U, Santoni-Rugiu E, Nagy P, Nielsen O, Ott P, Hage E, Dalhoff K, Rasmussen LJ, Tygstrup N (2002) Heterogeneity of ductular reactions in adult rat and human liver revealed by novel expression of deleted in malignant brain tumor 1. Am J Pathol 161:1187–1198 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bridgewater J, Galle PR, Khan SA, Llovet JM, Park JW, Patel T, Pawlik TM, Gores GJ (2014) Guidelines for the diagnosis and management of intrahepatic cholangiocarcinoma. J Hepatol 60:1268–1289 [DOI] [PubMed] [Google Scholar]
- Chen EY, Tan CM, Kou Y, Duan Q, Wang Z, Meirelles GV, Clark NR, Ma’ayan A (2013) Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinform 14:128 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung T, Park YN (2022) Up-to-date pathologic classification and molecular characteristics of intrahepatic cholangiocarcinoma. Front Med (lausanne) 9:857140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chung T, Rhee H, Nahm JH, Jeon Y, Yoo JE, Kim Y-J, Han DH, Park YN (2020) Clinicopathological characteristics of intrahepatic cholangiocarcinoma according to gross morphologic type: cholangiolocellular differentiation traits and inflammation- and proliferation-phenotypes. HPB 22:864–873 [DOI] [PubMed] [Google Scholar]
- Fiste O, Ntanasis-Stathopoulos I, Gavriatopoulou M, Liontos M, Koutsoukos K, Dimopoulos MA, Zagouri F (2021) The emerging role of immunotherapy in intrahepatic cholangiocarcinoma. Vaccines (basel) 9:422 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerber TS, Müller L, Bartsch F, Gröger LK, Schindeldecker M, Ridder DA, Goeppert B, Möhler M, Dueber C, Lang H, Roth W, Kloeckner R, Straub BK (2022) Integrative analysis of intrahepatic cholangiocarcinoma subtypes for improved patient stratification: clinical, pathological, and radiological considerations. Cancers (basel) 14:3156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goeppert B, Frauenschuh L, Zucknick M, Stenzinger A, Andrulis M, Klauschen F, Joehrens K, Warth A, Renner M, Mehrabi A, Hafezi M, Thelen A, Schirmacher P, Weichert W (2013) Prognostic impact of tumour-infiltrating immune cells on biliary tract cancer. Br J Cancer 109:2665–2674 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goeppert B, Roessler S, Becker N, Zucknick M, Vogel MN, Warth A, Pathil-Warth A, Mehrabi A, Schirmacher P, Mollenhauer J, Renner M (2017) DMBT1 expression in biliary carcinogenesis with correlation of clinicopathological data. Histopathology 70:1064–1071 [DOI] [PubMed] [Google Scholar]
- Gooden MJ, de Bock GH, Leffers N, Daemen T, Nijman HW (2011) The prognostic influence of tumour-infiltrating lymphocytes in cancer: a systematic review with meta-analysis. Br J Cancer 105:93–103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greten TF, Schwabe R, Bardeesy N, Ma L, Goyal L, Kelley RK, Wang XW (2023) Immunology and immunotherapy of cholangiocarcinoma. Nat Rev Gastroenterol Hepatol 20:349–365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Groot Koerkamp B, Fong Y (2014) Outcomes in biliary malignancy. J Surg Oncol 110:585–591 [DOI] [PubMed] [Google Scholar]
- Hayashi A, Misumi K, Shibahara J, Arita J, Sakamoto Y, Hasegawa K, Kokudo N, Fukayama M (2016) Distinct Clinicopathologic and Genetic Features of 2 Histologic Subtypes of Intrahepatic Cholangiocarcinoma. Am J Surg Pathol 40:1021–1030 [DOI] [PubMed] [Google Scholar]
- Ieta K, Tanaka F, Utsunomiya T, Kuwano H, Mori M (2006) CEACAM6 gene expression in intrahepatic cholangiocarcinoma. Br J Cancer 95:532–540 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jing C-Y, Yi-Peng Fu, Yi Y, Mei-Xia Zhang S-S, Zheng J-L, Gan W, Xin Xu, Lin J-J, Zhang J, Qiu S-J, Zhang B-H (2019) HHLA2 in intrahepatic cholangiocarcinoma: an immune checkpoint with prognostic significance and wider expression compared with PD-L1. J Immunother Cancer 7:77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Job S, Rapoud D, Dos Santos A, Gonzalez P, Desterke C, Pascal G, Elarouci N, Ayadi M, Adam R, Azoulay D, Castaing D, Vibert E, Cherqui D, Samuel D, Sa Cuhna A, Marchio A, Pineau P, Guettier C, de Reyniès A, Faivre J (2020) Identification of four immune subtypes characterized by distinct composition and functions of tumor microenvironment in intrahepatic cholangiocarcinoma. Hepatology 72:965–981 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelley RK, Ueno M, Yoo C, Finn RS, Furuse J, Ren Z, Yau T, Klümpen HJ, Chan SL, Ozaka M, Verslype C, Bouattour M, Park JO, Barajas O, Pelzer U, Valle JW, Yu L, Malhotra U, Siegel AB, Edeline J, Vogel A (2023) Pembrolizumab in combination with gemcitabine and cisplatin compared with gemcitabine and cisplatin alone for patients with advanced biliary tract cancer (KEYNOTE-966): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 401:1853–1865 [DOI] [PubMed] [Google Scholar]
- Kendall T, Verheij J, Gaudio E, Evert M, Guido M, Goeppert B, Carpino G (2019) Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int 39(Suppl 1):7–18 [DOI] [PubMed] [Google Scholar]
- Kinzler MN, Schulze F, Bankov K, Gretser S, Becker N, Leichner R, Stehle A, Abedin N, Trojan J, Zeuzem S, Schnitzbauer AA, Wild PJ, Walter D (2022) Impact of small duct- and large duct type on survival in patients with intrahepatic cholangiocarcinoma: results from a German tertiary center. Pathol Res Pract 238:154126 [DOI] [PubMed] [Google Scholar]
- Kinzler MN, Bankov K, Bein J, Döring C, Schulze F, Reis H, Mahmoudi S, Koch V, Grünewald LD, Stehle A, Walter D, Finkelmeier F, Zeuzem S, Wild PJ, Vogl TJ, Bernatz S (2023) CXCL1 and CXCL6 are potential predictors for HCC response to TACE. Curr Oncol 30:3516–3528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Konishi D, Umeda Y, Yoshida K, Shigeyasu K, Yano S, Toji T, Takeda S, Yoshida R, Yasui K, Fuji T, Matsumoto K, Kishimoto H, Michiue H, Teraishi F, Kato H, Tazawa H, Yanai H, Yagi T, Goel A, Fujiwara T (2022) Regulatory T cells induce a suppressive immune milieu and promote lymph node metastasis in intrahepatic cholangiocarcinoma. Br J Cancer 127:757–765 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, McDermott MG, Monteiro CD, Gundersen GW, Ma’ayan A (2016) Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res 44:W90–W97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kurlinkus B, Ger M, Kaupinis A, Jasiunas E, Valius M, Sileikis A (2021) CEACAM6’s role as a chemoresistance and prognostic biomarker for pancreatic cancer: a comparison of CEACAM6’s diagnostic and prognostic capabilities with those of CA19–9 and CEA. Life (basel) 11:542 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liau J-Y, Tsai J-H, Yuan R-H, Chang C-N, Lee H-J, Jeng Y-M (2014) Morphological subclassification of intrahepatic cholangiocarcinoma: etiological, clinicopathological, and molecular features. Mod Pathol 27:1163–1173 [DOI] [PubMed] [Google Scholar]
- Liu C, Wang M, Lv H, Liu B, Ya X, Zhao W, Wang W (2022) CEACAM6 promotes cholangiocarcinoma migration and invasion by inducing epithelial-mesenchymal transition through inhibition of the SRC/PI3K/AKT signaling pathway. Oncol Lett 23:39 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mollenhauer J, Herbertz S, Holmskov U, Tolnay M, Krebs I, Merlo A, Schrøder HD, Maier D, Breitling F, Wiemann S, Gröne HJ, Poustka A (2000) DMBT1 encodes a protein involved in the immune defense and in epithelial differentiation and is highly unstable in cancer. Cancer Res 60:1704–1710 [PubMed] [Google Scholar]
- Oh D-Y, He AR, Qin S, Chen L-T, Okusaka T, Vogel A, Kim JW, Suksombooncharoen T, Lee MA, Kitano M, Burris III HA, Bouattour M, Tanasanvimon S, Zaucha R, Avallone A, Cundom J, Rokutanda N, Xiong J, Cohen G, Valle JW (2022) A phase 3 randomized, double-blind, placebo-controlled study of durvalumab in combination with gemcitabine plus cisplatin (GemCis) in patients (pts) with advanced biliary tract cancer (BTC): TOPAZ-1. J Clin Oncol 40:378–478 [Google Scholar]
- Pan C, Liu H, Robins E, Song W, Liu D, Li Z, Zheng L (2020) Next-generation immuno-oncology agents: current momentum shifts in cancer immunotherapy. J Hematol Oncol 13:29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Preusse C, Eede P, Heinzeling L, Freitag K, Koll R, Froehlich W, Schneider U, Allenbach Y, Benveniste O, Schänzer A, Goebel HH, Stenzel W, Radke J (2021) NanoString technology distinguishes anti-TIF-1γ(+) from anti-Mi-2(+) dermatomyositis patients. Brain Pathol 31:e12957 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rose JB, Correa-Gallego C, Li Y, Nelson J, Alseidi A, Helton WS, Allen PJ, D’Angelica MI, DeMatteo RP, Fong Y, Kingham TP, Kowdley KV, Jarnagin WR, Rocha FG (2016) The role of biliary carcinoembryonic antigen-related cellular adhesion molecule 6 (CEACAM6) as a biomarker in cholangiocarcinoma. PLoS ONE 11:e0150195 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Roumenina LT, Daugan MV, Petitprez F, Sautès-Fridman C, Fridman WH (2019) Context-dependent roles of complement in cancer. Nat Rev Cancer 19:698–715 [DOI] [PubMed] [Google Scholar]
- Sasaki M, Huang SF, Chen MF, Jan YY, Yeh TS, Ishikawa A, Mollenhauer J, Poustka A, Tsuneyama K, Nimura Y, Oda K, Nakanuma Y (2003) Expression of deleted in malignant brain tumor-1 (DMBT1) molecule in biliary epithelium is augmented in hepatolithiasis: possible participation in lithogenesis. Dig Dis Sci 48:1234–1240 [DOI] [PubMed] [Google Scholar]
- Sigel CS, Drill E, Zhou Yi, Basturk O, Askan G, Pak LM, Vakiani E, Wang T, Boerner T, Do RKG, Simpson AL, Jarnagin W, Klimstra DS (2018) Intrahepatic cholangiocarcinomas have histologically and immunophenotypically distinct small and large duct patterns. Am J Surg Pathol 42:1334–1345 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Son KH, Ahn CB, Kim HJ, Kim JS (2020) Quantitative proteomic analysis of bile in extrahepatic cholangiocarcinoma patients. J Cancer 11:4073–4080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang H, He M, Zhang Z, Yin W, Ren B, Lin Y (2022) Low complement Factor h-related 3 (CFHR3) expression indicates poor prognosis and immune regulation in cholangiocarcinoma. J Oncol 2022:1752827 [DOI] [PMC free article] [PubMed] [Google Scholar]
- WHO Classification of Tumours (2019) 5th ed. Vol. 1. Digestive System Tumours, 2019. [Online]. Available: https://publications.iarc.fr/Book-And-Report-Series/Who-Classification-Of-Tumours/Digestive-System-Tumours-2019. Accessed 20 May 2024
- Xia T, Li K, Niu N, Shao Y, Ding D, Thomas DL, Jing H, Fujiwara K, Hu H, Osipov A, Yuan C, Wolfgang CL, Thompson ED, Anders RA, He J, Mou Y, Murphy AG, Zheng L (2022a) Immune cell atlas of cholangiocarcinomas reveals distinct tumor microenvironments and associated prognoses. J Hematol Oncol 15:37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xia T, Li K, Niu N, Shao Y, Ding D, Thomas DL, Jing H, Fujiwara K, Haijie Hu, Osipov A, Yuan C, Wolfgang CL, Thompson ED, Anders RA, He J, Mou Y, Murphy AG, Zheng L (2022b) Immune cell atlas of cholangiocarcinomas reveals distinct tumor microenvironments and associated prognoses. J Hematol Oncol 15:37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoon JiG, Kim MH, Jang M, Kim H, Hwang HK, Kang CM, Lee WJ, Kang B, Lee C-k, Lee MG, Chung HC, Choi HJ, Park YN (2021) Molecular characterization of biliary tract cancer predicts chemotherapy and programmed death 1/programmed death-ligand 1 blockade responses. Hepatology 74:1914–1931 [DOI] [PubMed] [Google Scholar]
- Yugawa K, Itoh S, Yoshizumi T, Iseda N, Tomiyama T, Toshima T, Harada N, Kohashi K, Oda Y, Mori M (2021) Prognostic impact of tumor microvessels in intrahepatic cholangiocarcinoma: association with tumor-infiltrating lymphocytes. Mod Pathol 34:798–807 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
The data that support the findings of this study are available from the corresponding author upon reasonable request.




