Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 225
Filter
1.
Biosci Trends ; 18(3): 263-276, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38853000

ABSTRACT

This study aims to determine the predictive role of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) derived radiomic model in tumor immune profiling and immunotherapy for cholangiocarcinoma. To perform radiomic analysis, immune related subgroup clustering was first performed by single sample gene set enrichment analysis (ssGSEA). Second, a total of 806 radiomic features for each phase of DCE-MRI were extracted by utilizing the Python package Pyradiomics. Then, a predictive radiomic signature model was constructed after a three-step features reduction and selection, and receiver operating characteristic (ROC) curve was employed to evaluate the performance of this model. In the end, an independent testing cohort involving cholangiocarcinoma patients with anti-PD-1 Sintilimab treatment after surgery was used to verify the potential application of the established radiomic model in immunotherapy for cholangiocarcinoma. Two distinct immune related subgroups were classified using ssGSEA based on transcriptome sequencing. For radiomic analysis, a total of 10 predictive radiomic features were finally identified to establish a radiomic signature model for immune landscape classification. Regarding to the predictive performance, the mean AUC of ROC curves was 0.80 in the training/validation cohort. For the independent testing cohort, the individual predictive probability by radiomic model and the corresponding immune score derived from ssGSEA was significantly correlated. In conclusion, radiomic signature model based on DCE-MRI was capable of predicting the immune landscape of chalangiocarcinoma. Consequently, a potentially clinical application of this developed radiomic model to guide immunotherapy for cholangiocarcinoma was suggested.


Subject(s)
Cholangiocarcinoma , Immunotherapy , Magnetic Resonance Imaging , Humans , Cholangiocarcinoma/diagnostic imaging , Cholangiocarcinoma/immunology , Cholangiocarcinoma/therapy , Cholangiocarcinoma/genetics , Magnetic Resonance Imaging/methods , Immunotherapy/methods , Male , Bile Duct Neoplasms/diagnostic imaging , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/therapy , Female , Middle Aged , Contrast Media , ROC Curve , Aged , Transcriptome
2.
Int Immunopharmacol ; 136: 112273, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38810311

ABSTRACT

Cholangiocarcinoma (CCA) presents a significant clinical challenge which is often identified in advanced stages, therby restricting the effectiveness of surgical interventions for most patients. The high incidence of cancer recurrence and resistance to chemotherapy further contribute to a bleak prognosis and low survival rates. To address this pressing need for effective therapeutic strategies, our study focuses on the development of an innovative cellular immunotherapy, specifically utilizing chimeric antigen receptor (CAR)-engineered natural killer (NK) cells designed to target the cMET receptor tyrosine kinase. In this investigation, we initiated the screening of a phage library displaying human single-chain variable fragment (ScFv) to identify novel ScFv molecules with specificity for cMET. Remarkably, ScFv11, ScFv72, and ScFv114 demonstrated exceptional binding affinity, confirmed by molecular docking analysis. These selected ScFvs, in addition to the well-established anti-cMET ScFvA, were integrated into a CAR cassette harboring CD28 transmembrane region-41BB-CD3ζ domains. The resulting anti-cMET CAR constructs were transduced into NK-92 cells, generating potent anti-cMET CAR-NK-92 cells. To assess the specificity and efficacy of these engineered cells, we employed KKU213A cells with high cMET expression and KKU055 cells with low cMET levels. Notably, co-culture of anti-cMET CAR-NK-92 cells with KKU213A cells resulted in significantly increased cell death, whereas no such effect was observed with KKU055 cells. In summary, our study identified cMET as a promising therapeutic target for CCA. The NK-92 cells, armed with the anti-cMET CAR molecule, have shown strong ability to kill cancer cells specifically, indicating their potential as a promising treatment for CCA in the future.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Killer Cells, Natural , Proto-Oncogene Proteins c-met , Receptors, Chimeric Antigen , Single-Chain Antibodies , Humans , Single-Chain Antibodies/genetics , Single-Chain Antibodies/therapeutic use , Single-Chain Antibodies/immunology , Cholangiocarcinoma/therapy , Cholangiocarcinoma/immunology , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Killer Cells, Natural/immunology , Cell Line, Tumor , Bile Duct Neoplasms/therapy , Bile Duct Neoplasms/immunology , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins c-met/immunology , Immunotherapy, Adoptive/methods , Immunotherapy/methods , Precision Medicine
3.
Front Immunol ; 15: 1376590, 2024.
Article in English | MEDLINE | ID: mdl-38799431

ABSTRACT

Background: Research of immunotherapy for cholangiocarcinoma has yielded some results, but more clinical data are needed to prove its efficacy and safety. Moreover, there is a need to identify accessible indexes for selecting patients who may benefit from such treatments. Methods: The medical records of 66 cholangiocarcinoma patients who underwent immunotherapy were retrospectively collected. The effectiveness of immunotherapy was assessed by tumor response, progression-free survival (PFS), and overall survival (OS), while safety was evaluated by adverse events during treatment. Univariate and multivariate Cox regression analyses were performed to identify prognostic risk factors for PFS and OS, and Kaplan-Meier curves of potential prognostic factors were drawn. Results: Overall, in this study, immunotherapy achieved an objective response rate of 24.2% and a disease control rate of 89.4% for the included patients. The median PFS was 445 days, and the median OS was 772.5 days. Of the 66 patients, 65 experienced adverse events during treatment, but none had severe consequences. Multivariate Cox analysis indicated that tumor number is a prognostic risk factor for disease progression following immunotherapy in cholangiocarcinoma patients, while tumor differentiation and the fibrosis-4 (FIB-4) index are independent risk factors for OS. Conclusion: In general, immunotherapy for cholangiocarcinoma is safe, with adverse events remaining within manageable limits, and it can effectively control disease progression in most patients. The FIB-4 index may reflect the potential benefit of immunotherapy for patients with cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Immunotherapy , Humans , Cholangiocarcinoma/therapy , Cholangiocarcinoma/immunology , Cholangiocarcinoma/mortality , Male , Female , Middle Aged , Bile Duct Neoplasms/therapy , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/immunology , Aged , Prognosis , Immunotherapy/adverse effects , Immunotherapy/methods , Retrospective Studies , Adult , Fibrosis , Aged, 80 and over , Risk Factors
4.
Cancer Lett ; 594: 216992, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38797231

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is an aggressive cancer with an extremely poor prognosis, and new treatment options are needed. Recently, immunotherapy has emerged as an efficient treatment against malignant tumors, but less effective in iCCA. Activation of stimulator of interferon genes (STING) signaling could reignite immunologically inert tumors, but the expression and role of STING in iCCA remains to be determined. Here, we show STING is expressed in iCCA, and patients with high expression of STING in early-stage iCCA have a longer overall survival than those have low expression. Increased immune cell infiltration in early-stage iCCA corresponds to elevated STING expression. In mice iCCA models, treatment with the STING agonist MSA-2 show stage-specific inhibitory effects on tumors, with beneficial effects in early-stage tumors but not with advanced-stage cancer. This discrepancy was associated with greater programmed cell death ligand 1 (PD-L1) expression in advanced-stage tumors. Combination therapy targeting PD-L1 and MSA-2 strikingly reduced tumor burden in such tumors compared to either monotherapy. Cumulatively, these data demonstrate that STING agonism monotherapy improves the immune landscape of the tumor microenvironment in early-stage iCCA, while combination therapy ameliorates advanced-stage iCCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Membrane Proteins , Cholangiocarcinoma/immunology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cholangiocarcinoma/drug therapy , Animals , Membrane Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/agonists , Humans , Mice , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Neoplasm Staging , Cell Line, Tumor , Tumor Microenvironment/immunology , Male , Female , Signal Transduction
5.
Biomed Pharmacother ; 175: 116718, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38744221

ABSTRACT

Advanced cholangiocarcinoma (CCA) presents a clinical challenge due to limited treatment options, necessitating exploration of innovative therapeutic approaches. Bispecific T cell engager (BTE)-armed T cell therapy shows promise in hematological and solid malignancies, offering potential advantages in safety over continuous BTE infusion. In this context, we developed a novel BTE, targeting CD3 on T cells and integrin αvß6, an antigen elevated in various epithelial malignancies, on cancer cells. The novel BTE was generated by fusing an integrin αvß6-binding peptide (A20) to an anti-CD3 (OKT3) single-chain variable fragment (scFv) through a G4S peptide linker (A20/αCD3 BTE). T cells were then armed with A20/αCD3 BTE (A20/αCD3-armed T cells) and assessed for antitumor activity. Our results highlight the specific binding of A20/αCD3 BTE to CD3 on T cells and integrin αvß6 on target cells, effectively redirecting T cells towards these targets. After co-culture, A20/αCD3-armed T cells exhibited significantly heightened cytotoxicity against integrin αvß6-expressing target cells compared to unarmed T cells in both KKU-213A cells and A375.ß6 cells. Moreover, in a five-day co-culture, A20/αCD3-armed T cells demonstrated superior cytotoxicity against KKU-213A spheroids compared to unarmed T cells. Importantly, A20/αCD3-armed T cells exhibited an increased proportion of the effector memory T cell (Tem) subset, upregulation of T cell activation markers, enhanced T cell proliferation, and increased cytolytic molecule/cytokine production, when compared to unarmed T cells in an integrin αvß6-dependent manner. These findings support the potential of A20/αCD3-armed T cells as a novel therapeutic approach for integrin αvß6-expressing cancers.


Subject(s)
Antigens, Neoplasm , Bile Duct Neoplasms , Cholangiocarcinoma , Integrins , T-Lymphocytes , Humans , Cholangiocarcinoma/immunology , Cholangiocarcinoma/therapy , Cholangiocarcinoma/pathology , Antigens, Neoplasm/immunology , T-Lymphocytes/immunology , Integrins/metabolism , Cell Line, Tumor , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/therapy , CD3 Complex/immunology , Single-Chain Antibodies/pharmacology , Coculture Techniques , Antibodies, Bispecific/pharmacology
6.
Biomed Pharmacother ; 175: 116659, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38692063

ABSTRACT

Cholangiocarcinoma (CCA), a rare yet notably aggressive cancer, has experienced a surge in incidence in recent years. Presently, surgical resection remains the most effective curative strategy for CCA. Nevertheless, a majority of patients with CCA are ineligible for surgical removal at the time of diagnosis. For advanced stages of CCA, the combination of gemcitabine and cisplatin is established as the standard chemotherapy regimen. Despite this, treatment efficacy is often hindered by the development of resistance. In recent times, immune checkpoint inhibitors, particularly those that block programmed death 1 and its ligand (PD1/PD-L1), have emerged as promising strategies against a variety of cancers and are being increasingly integrated into the therapeutic landscape of CCA. A growing body of research supports that the use of PD1/PD-L1 monoclonal antibodies in conjunction with chemotherapy may significantly improve patient outcomes. This article seeks to meticulously review the latest studies on PD1/PD-L1 involvement in CCA, delving into their expression profiles, prognostic significance, contribution to oncogenic processes, and their potential clinical utility.


Subject(s)
B7-H1 Antigen , Bile Duct Neoplasms , Cholangiocarcinoma , Immune Checkpoint Inhibitors , Immunotherapy , Programmed Cell Death 1 Receptor , Cholangiocarcinoma/therapy , Cholangiocarcinoma/immunology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Humans , B7-H1 Antigen/antagonists & inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Bile Duct Neoplasms/therapy , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/pathology , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Animals
7.
J Hepatol ; 80(6): 892-903, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38458319

ABSTRACT

BACKGROUND & AIMS: Cholangiocarcinoma (CCA) is a poorly immunogenic malignancy associated with limited survival. Syngeneic immunocompetent mouse models of CCA are an essential tool to elucidate the tumor immune microenvironment (TIME), understand mechanisms of tumor immune evasion, and test novel immunotherapeutic strategies. The scope of this study was to develop and characterize immunocompetent CCA models with distinct genetic drivers, and correlate tumor genomics, immunobiology, and therapeutic response. METHODS: A multifaceted approach including scRNA-seq, CITE-seq, whole exome and bulk RNA sequencing was employed. FDA-approved PD-1/PD-L1 antibodies were tested in humanized PD-1/PD-L1 mice (HuPD-H1). RESULTS: A genetic mouse model of intrahepatic CCA (iCCA) driven by intrabiliary transduction of Fbxw7ΔF/Akt that mimics human iCCA was generated. From the Fbxw7ΔF/Akt tumors, a murine cell line (FAC) and syngeneic model with genetic and phenotypic characteristics of human iCCA were developed. Established SB1 (YAPS127A/Akt) and KPPC (KrasG12Dp53L/L) models were compared to the FAC model. Although the models had transcriptomic similarities, they had substantial differences as well. Mutation patterns of FAC, SB1, and KPPC cells matched different mutational signatures in Western and Japanese CCA patient cohorts. KPPC tumors had a high tumor mutation burden. FAC tumors had a T cell-infiltrated TIME, while SB1 tumors had a preponderance of suppressive myeloid cells. FAC, SB1, and KPPC tumors matched different immune signatures in human iCCA cohorts. Moreover, FAC, SB1, and KPPC tumor-bearing HuPD-H1 mice displayed differential responses to nivolumab or durvalumab. CONCLUSIONS: Syngeneic iCCA models display a correlation between tumor genotype and TIME phenotype, with differential responses to FDA-approved immunotherapies. This study underscores the importance of leveraging multiple preclinical models to understand responses to immunotherapy in different genetic subsets of human CCA. IMPACT AND IMPLICATIONS: Understanding the relationship between tumor genotype and the phenotype of the immune microenvironment is an unmet need in cholangiocarcinoma (CCA). Herein, we use syngeneic murine models of intrahepatic CCA with different genetic drivers to demonstrate a correlation between tumor genotype and immune microenvironment phenotype in murine models, which is associated with differential responses to FDA-approved immunotherapies. This information will help guide other preclinical studies. Additionally, it emphasizes that immune checkpoint inhibition in patients with CCA is not a "one-size-fits-all" approach. Our observations suggest that, as for targeted therapies, patients should be stratified and selected for treatment according to their tumor genetics.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Disease Models, Animal , Tumor Microenvironment , Animals , Cholangiocarcinoma/immunology , Cholangiocarcinoma/genetics , Mice , Tumor Microenvironment/immunology , Humans , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/genetics , F-Box-WD Repeat-Containing Protein 7/genetics , Cell Line, Tumor
8.
Curr Cancer Drug Targets ; 24(7): 681-700, 2024.
Article in English | MEDLINE | ID: mdl-38213139

ABSTRACT

Cholangiocarcinoma (CCA) is an epithelial cancer distinguished by bile duct cell differentiation and is also a fibroproliferative tumor. It is characterized by a dense mesenchyme and a complex tumor immune microenvironment (TME). The TME comprises both cellular and non-cellular components. The celluar component includes CCA cells, immune cells and mesenchymal cells represented by the cancer-associated fibroblasts (CAFs), while the non-cellular component is represented by mesenchymal elements such as the extracellular matrix (ECM). Recent studies have demonstrated the important role of the TME in the development, progression, and treatment resistance of CCA. These cell-associated prognostic markers as well as intercellular connections, may serve as potential therapeutic targets and could inspire new treatment approaches for CCA in the future. This paper aims to summarize the current understanding of CCA's immune microenvironment, focusing on immune cells, mesenchymal cells, ECM, intercellular interactions, and metabolism within the microenvironment.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Disease Progression , Tumor Microenvironment , Cholangiocarcinoma/immunology , Cholangiocarcinoma/pathology , Humans , Tumor Microenvironment/immunology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/pathology , Cancer-Associated Fibroblasts/immunology , Cancer-Associated Fibroblasts/pathology , Extracellular Matrix/immunology , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Animals
9.
Neoplasia ; 43: 100920, 2023 09.
Article in English | MEDLINE | ID: mdl-37515847

ABSTRACT

BACKGROUND: Most patients with resected bile tract cancers (BTCs) survive for less than 5 years; however, some achieve better prognosis. The tumor microbiome can improve survival by regulating the tumor immune microenvironment. However, whether the tumor microbiome promotes immune cell infiltration in BTCs is unknown. This study aimed to determine the association between CD8+ T lymphocyte infiltration and the tumor microbiome in patients with resected BTCs. METHODS: Archived formalin-fixed paraffin-embedded tumor specimens were collected from patients with resected BTCs and analyzed using 16S rRNA gene sequencing to identify that prognosis-related and significantly differentially enriched taxa. Gene ontology (GO) analysis of the differentially enriched taxa was used to assess how CD8+ T lymphocyte infiltration is affected by the tumor microbiome of BTCs. RESULTS: We enrolled 32 patients with resected BTCs. The high CD8+ lymphocyte-infiltration (CD8hi) group had four significantly enriched taxa, and in the low CD8+ lymphocyte-infiltration (CD8low) group comprised one significantly enriched taxon. Patients with higher Clostridia abundance (enriched in the CD8hi group) experienced longer overall survival than those with lower abundance. The enrichment of Clostridia in the CD8hi group corresponded with lower CCL2 expression and downregulation of phosphatidylinositol 3-kinase activity, which might decrease myeloid-derived suppressor cell recruitment to the tumor milieu, thus increasing CD8+ lymphocyte infiltration in BTCs. CONCLUSIONS: The tumor microbiome is related to CD8+ T lymphocyte infiltration in patients with resected BTCs. The relationship between tumor Clostridia and high infiltration of CD8+ T lymphocytes might reflect decreased recruitment of myeloid-derived suppressor cells via the PI3K-CCL2-CCR2 axis.


Subject(s)
Bile Duct Neoplasms , CD8-Positive T-Lymphocytes , Cholangiocarcinoma , Clostridium , Lymphocytes, Tumor-Infiltrating , Microbiota , Humans , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL2/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid-Derived Suppressor Cells/immunology , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Receptors, CCR2/metabolism , RNA, Ribosomal, 16S , Tumor Microenvironment/genetics , Cholangiocarcinoma/immunology , Cholangiocarcinoma/microbiology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/microbiology , Clostridium/immunology
10.
Br J Cancer ; 126(2): 219-227, 2022 02.
Article in English | MEDLINE | ID: mdl-34616011

ABSTRACT

BACKGROUND: Caveolin-1 (CAV1) in cancer-associated fibroblasts (CAFs) has pro- or anti-tumourigenic effect depending on the cancer type. However, its effect in intrahepatic carcinoma (ICC) remains unknown. Therefore, this study aimed to investigate the relationship between CAV1 in CAFs and tumour-infiltrating lymphocyte (TIL) numbers or PD-L1 levels in ICC patients. METHODS: Consecutive ICC patients (n = 158) were enrolled in this study. The levels of CAV1 in CAFs, CD8 + TILs, Foxp3+ TILs and PD-L1 in cancer cells were analysed using immunohistochemistry. Their association with the clinicopathological factors and prognosis were evaluated. The correlation between these factors was evaluated. RESULTS: CAV1 upregulation in CAFs was associated with a poor overall survival (OS) (P < 0.001) and recurrence-free survival (P = 0.008). Clinicopathological factors were associated with high CA19-9 levels (P < 0.001), advanced tumour stage (P = 0.046) and lymph node metastasis (P = 0.004). CAV1 level was positively correlated with Foxp3+ TIL numbers (P = 0.01). There were no significant correlations between CAV1 levels and CD8 + TIL numbers (P = 0.80) and PD-L1 levels (P = 0.97). An increased CD8 + TIL number and decreased Foxp3+ TIL number were associated with an increased OS. In multivariate analysis, positive CAV1 expression in CAFs (P = 0.013) and decreased CD8 + TIL numbers (P = 0.021) were independent poor prognostic factors. CONCLUSION: Cellular senescence, represented by CAV1 levels, may be a marker of CAFs and a prognostic indicator of ICC through Foxp3+ TIL regulation. CAV1 expression in CAFs can be a therapeutic target for ICC.


Subject(s)
B7-H1 Antigen/metabolism , Cancer-Associated Fibroblasts/pathology , Caveolin 1/metabolism , Cellular Senescence , Cholangiocarcinoma/pathology , Forkhead Transcription Factors/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Aged , B7-H1 Antigen/immunology , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cancer-Associated Fibroblasts/metabolism , Cholangiocarcinoma/immunology , Cholangiocarcinoma/metabolism , Female , Forkhead Transcription Factors/immunology , Humans , Male , Prognosis , Survival Rate
11.
Br J Cancer ; 126(1): 10-23, 2022 01.
Article in English | MEDLINE | ID: mdl-34400801

ABSTRACT

Liver cancer is one of the most prevalent cancers, and the third most common cause of cancer-related mortality worldwide. The therapeutic options for the main types of primary liver cancer-hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA)-are very limited. HCC and CCA are immunogenic cancers, but effective immune-mediated tumour control is prevented by their immunosuppressive tumour microenvironment. Despite the critical involvement of key co-inhibitory immune checkpoint interactions in immunosuppression in liver cancer, only a minority of patients with HCC respond to monotherapy using approved checkpoint inhibitor antibodies. To develop effective (combinatorial) therapeutic immune checkpoint strategies for liver cancer, in-depth knowledge of the different mechanisms that contribute to intratumoral immunosuppression is needed. Here, we review the co-inhibitory pathways that are known to suppress intratumoral T cells in HCC and CCA. We provide a detailed description of insights from preclinical studies in cellular crosstalk within the tumour microenvironment that results in interactions between co-inhibitory receptors on different T-cell subsets and their ligands on other cell types, including tumour cells. We suggest alternative immune checkpoints as promising targets, and draw attention to the possibility of combined targeting of co-inhibitory and co-stimulatory pathways to abrogate immunosuppression.


Subject(s)
Cholangiocarcinoma/immunology , Immune Checkpoint Proteins/immunology , Immunosuppression Therapy/methods , Immunotherapy/methods , Liver Neoplasms/pathology , Tumor Microenvironment , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/pathology , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/pathology , Clinical Trials as Topic , Humans , Immune Checkpoint Proteins/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology
12.
Hepatology ; 75(2): 297-308, 2022 02.
Article in English | MEDLINE | ID: mdl-34510503

ABSTRACT

BACKGROUND AND AIMS: Cholangiocarcinoma (CCA) is a deadly and highly therapy-refractory cancer of the bile ducts, with early results from immune checkpoint blockade trials showing limited responses. Whereas recent molecular assessments have made bulk characterizations of immune profiles and their genomic correlates, spatial assessments may reveal actionable insights. APPROACH AND RESULTS: Here, we have integrated immune checkpoint-directed immunohistochemistry with next-generation sequencing of resected intrahepatic CCA samples from 96 patients. We found that both T-cell and immune checkpoint markers are enriched at the tumor margins compared to the tumor center. Using two approaches, we identify high programmed cell death protein 1 or lymphocyte-activation gene 3 and low CD3/CD4/inducible T-cell costimulator specifically in the tumor center as associated with poor survival. Moreover, loss-of-function BRCA1-associated protein-1 mutations are associated with and cause elevated expression of the immunosuppressive checkpoint marker, B7 homolog 4. CONCLUSIONS: This study provides a foundation on which to rationally improve and tailor immunotherapy approaches for this difficult-to-treat disease.


Subject(s)
Antigens, CD/metabolism , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Programmed Cell Death 1 Receptor/metabolism , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , B7 Antigens/genetics , Bile Duct Neoplasms/immunology , Bile Ducts, Intrahepatic , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , CD4-Positive T-Lymphocytes , Cell Line, Tumor , Cholangiocarcinoma/immunology , Female , Gene Expression , Genes, Tumor Suppressor , Genomics , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Inducible T-Cell Co-Stimulator Protein/genetics , Inducible T-Cell Co-Stimulator Protein/metabolism , Loss of Function Mutation , Male , Middle Aged , Oncogenes/genetics , Programmed Cell Death 1 Receptor/genetics , Survival Rate , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , V-Set Domain-Containing T-Cell Activation Inhibitor 1/genetics , Young Adult , Lymphocyte Activation Gene 3 Protein
13.
Clin Cancer Res ; 28(3): 540-551, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34785581

ABSTRACT

PURPOSE: Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a rare malignancy associated with an overall poor prognosis. We aimed to investigate the immune profile of cHCC-CCA and determine its impact on disease outcome. EXPERIMENTAL DESIGN: We performed a multicenter study of 96 patients with cHCC-CCA. Gene expression profile was analyzed using nCounter PanCancer IO 360 Panel. Densities of main immune cells subsets were quantified from digital slides of IHC stainings. Genetic alterations were investigated using targeted next-generation sequencing. RESULTS: Two main immune subtypes of cHCC-CCA were identified by clustering analysis: an "immune-high" (IH) subtype (57% of the cases) and an "immune-low" (IL) subtype (43% of the cases). Tumors classified as IH showed overexpression of genes related to immune cells recruitment, adaptive and innate immunity, antigen presentation, cytotoxicity, immune suppression, and inflammation (P < 0.0001). IH cHCC-CCAs also displayed activation of gene signatures recently shown to be associated with response to immunotherapy in patients with HCC. Quantification of immunostainings confirmed that IH tumors were also characterized by higher densities of immune cells. Immune subtypes were not associated with any genetic alterations. Finally, multivariate analysis showed that the IH subtype was an independent predictor of improved overall survival. CONCLUSIONS: We have identified a subgroup of cHCC-CCA that displays features of an ongoing intratumor immune response, along with an activation of gene signatures predictive of response to immunotherapy in HCC. This tumor subclass is associated with an improved clinical outcome. These findings suggest that a subset of patients with cHCC-CCA may benefit from immunomodulating therapeutic approaches.


Subject(s)
Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Cholangiocarcinoma/immunology , Cholangiocarcinoma/therapy , Immunotherapy , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Neoplasms, Multiple Primary/immunology , Neoplasms, Multiple Primary/therapy , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/diagnosis , Bile Duct Neoplasms/genetics , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/genetics , Cholangiocarcinoma/diagnosis , Cholangiocarcinoma/genetics , Female , Forecasting , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/genetics , Male , Middle Aged , Neoplasms, Multiple Primary/diagnosis , Neoplasms, Multiple Primary/genetics , Treatment Outcome
14.
J Exp Med ; 218(10)2021 10 04.
Article in English | MEDLINE | ID: mdl-34495298

ABSTRACT

Cholangiocarcinoma (CCA) results from the malignant transformation of cholangiocytes. Primary sclerosing cholangitis (PSC) and primary biliary cholangitis (PBC) are chronic diseases in which cholangiocytes are primarily damaged. Although PSC is an inflammatory condition predisposing to CCA, CCA is almost never found in the autoimmune context of PBC. Here, we hypothesized that PBC might favor CCA immunosurveillance. In preclinical murine models of cholangitis challenged with syngeneic CCA, PBC (but not PSC) reduced the frequency of CCA development and delayed tumor growth kinetics. This PBC-related effect appeared specific to CCA as it was not observed against other cancers, including hepatocellular carcinoma. The protective effect of PBC was relying on type 1 and type 2 T cell responses and, to a lesser extent, on B cells. Single-cell TCR/RNA sequencing revealed the existence of TCR clonotypes shared between the liver and CCA tumor of a PBC host. Altogether, these results evidence a mechanistic overlapping between autoimmunity and cancer immunosurveillance in the biliary tract.


Subject(s)
Autoimmunity , Bile Duct Neoplasms/immunology , Cholangiocarcinoma/immunology , Cholangitis/immunology , Animals , Bile Duct Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cholangiocarcinoma/pathology , Cholangitis/pathology , Cytokines/metabolism , Female , Forkhead Transcription Factors/metabolism , Liver/immunology , Liver/pathology , Mice, Inbred C57BL , Monitoring, Immunologic , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology
15.
Front Immunol ; 12: 705378, 2021.
Article in English | MEDLINE | ID: mdl-34526987

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is highly invasive and carries high mortality due to limited therapeutic strategies. In other solid tumors, immune checkpoint inhibitors (ICIs) target cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) and programmed death 1 (PD1), and the PD1 ligand PD-L1 has revolutionized treatment and improved outcomes. However, the relationship and clinical significance of CTLA-4 and PD-L1 expression in ICC remains to be addressed. Deciphering CTLA-4 and PD-L1 interactions in ICC enable targeted therapy for this disease. In this study, immunohistochemistry (IHC) was used to detect and quantify CTLA-4, forkhead box protein P3 (FOXP3), and PD-L1 in samples from 290 patients with ICC. The prognostic capabilities of CTLA-4, FOXP3, and PD-L1 expression in ICC were investigated with the Kaplan-Meier method. Independent risk factors related to ICC survival and recurrence were assessed by the Cox proportional hazards models. Here, we identified that CTLA-4+ lymphocyte density was elevated in ICC tumors compared with peritumoral hepatic tissues (P <.001), and patients with a high density of CTLA-4+ tumor-infiltrating lymphocytes (TILsCTLA-4 High) showed a reduced overall survival (OS) rate and increased cumulative recurrence rate compared with patients with TILsCTLA-4 Low (P <.001 and P = .024, respectively). Similarly, patients with high FOXP3+ TILs (TILsFOXP3 High) had poorer prognoses than patients with low FOXP3+ TILs (P = .021, P = .034, respectively), and the density of CTLA-4+ TILs was positively correlated with FOXP3+ TILs (Pearson r = .31, P <.001). Furthermore, patients with high PD-L1 expression in tumors (TumorPD-L1 High) and/or TILsCTLA-4 High presented worse OS and a higher recurrence rate than patients with TILsCTLA-4 LowTumorPD-L1 Low. Moreover, multiple tumors, lymph node metastasis, and high TumorPD-L1/TILsCTLA-4 were independent risk factors of cumulative recurrence and OS for patients after ICC tumor resection. Furthermore, among ICC patients, those with hepatolithiasis had a higher expression of CTLA-4 and worse OS compared with patients with HBV infection or undefined risk factors (P = .018). In conclusion, CTLA-4 is increased in TILs in ICC and has an expression profile distinct from PD1/PD-L1. TumorPD-L1/TILsCTLA-4 is a predictive factor of OS and ICC recurrence, suggesting that combined therapy targeting PD1/PD-L1 and CTLA-4 may be useful in treating patients with ICC.


Subject(s)
B7-H1 Antigen/physiology , Bile Duct Neoplasms/immunology , CTLA-4 Antigen/physiology , Cholangiocarcinoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Proteins/physiology , Programmed Cell Death 1 Receptor/physiology , Aged , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , CTLA-4 Antigen/biosynthesis , CTLA-4 Antigen/genetics , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Female , Forkhead Transcription Factors/analysis , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Lithiasis/etiology , Liver Diseases/etiology , Lymphocytes, Tumor-Infiltrating/chemistry , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Programmed Cell Death 1 Receptor/biosynthesis , Programmed Cell Death 1 Receptor/genetics , Proportional Hazards Models , Tumor Microenvironment , Up-Regulation
16.
J Surg Oncol ; 124(8): 1561-1568, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34351633

ABSTRACT

BACKGROUND AND OBJECTIVES: We evaluated the changes in natural killer cell activity (NKA) during the entire treatment period of patients with resectable biliopancreatic cancers and investigated the predictors of the failure of recovery of NKA after surgery. METHODS: A total of 202 patients who underwent curative resection for biliopancreatic cancer were enrolled in the study. NKA levels were measured six times during the treatment period. We investigated whether there was any difference in postoperative NKA recovery according to the period-by-time NKA value. RESULTS: NKA decreased after surgery (mean, 40 pg/ml) compared to the NKA value at admission (200.2 pg/ml), then began to increase from 3 weeks after surgery (139.7 pg/ml) and rose to normal NKA levels at 5 weeks (217.1 pg/ml). The pattern of NKA changes was distinct according to the NKA values at admission. In multivariate analysis, NKA values of less than 250 pg/ml at admission (odds ratio = 5.898, p = 0.044) were a predictor of NKA recovery failure 5 weeks after surgery. CONCLUSIONS: NKA rapidly decreased after curative surgery for biliopancreatic cancer and recovered to normal levels about 5 weeks later. Clinicians should be aware and cautious that patients with low NKA at admission may fail to recover NKA postoperatively.


Subject(s)
Bile Duct Neoplasms/pathology , Biliary Tract Neoplasms/pathology , Biliary Tract Surgical Procedures/methods , Killer Cells, Natural/immunology , Pancreatectomy/methods , Pancreatic Neoplasms/pathology , Recovery of Function , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/surgery , Biliary Tract Neoplasms/immunology , Biliary Tract Neoplasms/surgery , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/surgery , Prognosis , Retrospective Studies
17.
Int Immunopharmacol ; 99: 108039, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34426102

ABSTRACT

Cholangiocarcinoma (CHOL), the second most common malignant liver tumor, is clinically heterogeneous. In this study, we used gene expression profiles of CHOL obtained from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases to identify novel mutation signatures in CHOL. Hepcidin antimicrobial peptide (HAMP) was identified as a novel diagnostic biomarker for CHOL using the intersection of mutation analysis and receiver operating characteristic (ROC) analysis. We then explored the expression signatures of HAMP in CHOL. HAMP-related differentially expressed genes (DEGs) were selected for the identification of hub genes related to HAMP and for prognostic prediction model analysis. Gene set enrichment analysis (GSEA) showed that the HAMP-related DEGs were mainly enriched for signaling pathways related to cholangiocarcinoma development. Through immunohistochemistry validation, clinical cohorts analysis, and TCGA analysis, we investigated the association between HAMP and clinical parameters and found that decreased HAMP expression was correlated with advanced pathological grade and poor prognosis. Besides, we estimated the immune infiltration level in CHOL and its relationship with HAMP expression. The proportion of tumor-infiltrating cells revealed that gamma delta T cells and monocytes were positively correlated with HAMP expression. Besides, HAMP was also correlated with chemokine, CCL16. This evidence suggested that HAMP might contribute to immune activation in the CHOL microenvironment. Therefore, HAMP may play a synergistic role with these immune cells and chemokines to inhibit CHOL development. HAMP serves as a valuable biomarker in CHOL and is closely correlated with its progression.


Subject(s)
Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Hepcidins/genetics , Mutation , Bile Duct Neoplasms/immunology , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Biomarkers, Tumor/genetics , Chemokines, CC/metabolism , Cholangiocarcinoma/immunology , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Computational Biology , Correlation of Data , Data Mining , Databases, Genetic , Gene Expression Regulation, Neoplastic , Hepcidins/immunology , Hepcidins/metabolism , Humans , Kaplan-Meier Estimate , Neutrophils/immunology , Prognosis , Protein Interaction Maps , Transcriptome , Tumor Microenvironment/immunology
18.
J Immunother ; 44(7): 254-263, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34191790

ABSTRACT

Cholangiocarcinoma (CC) is an uncommon malignancy with increasing incidence and dismal prognosis. We conducted a comprehensive analysis of the CC tumor immune microenvironment (TIME) based on tumor location to identify therapeutic targets. We hypothesized that the TIME of CC would vary by primary tumor location and that high tumor infiltration by CD8+ T cells and low infiltration by M2 macrophages would be associated with improved survival. A retrospective analysis was conducted of 99 CC tumor samples surgically resected between 2000 and 2014. Tissue microarrays were constructed from each tumor and stained by immunohistochemistry for 24 markers of immune cells, immune activation or inhibition, programmed cell death-ligand 1, and mesothelin. Most tumors were amply infiltrated with by CD4+, CD8+, and FoxP3+ T cells, as well as by myeloid cells. Mesothelin expression ≥1+ by immunohistochemistry was found in 68% of tumors. We identified higher densities of M1 macrophages in primary distal extrahepatic CC, as well as metastatic lesions. Mesothelin expression was also significantly higher in distal extrahepatic CC. There was no association with survival of infiltration by CD4+, CD8+, or FoxP3+ T cells, mesothelin expression, or programmed cell death-ligand 1 percentage expression, however, high CD14+ myeloid cells and high CD163+ M2 macrophages were associated with worse survival. In conclusion, the CC TIME is a heterogenous milieu highly infiltrated by innate and adaptive immune cells, which differs based on primary tumor location and between primary tumors and metastatic lesions. The correlation of intratumoral M2 macrophages and myeloid cells with a worse prognosis may suggest promising immunotherapeutic targets in CC.


Subject(s)
Bile Duct Neoplasms/immunology , Cholangiocarcinoma/immunology , Myeloid Cells/immunology , Aged , B7-H1 Antigen/immunology , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/mortality , Cholangiocarcinoma/pathology , Female , Humans , Male , Mesothelin/immunology , Middle Aged , Prognosis , Proportional Hazards Models , Retrospective Studies , T-Lymphocytes/immunology
20.
Immunotherapy ; 13(13): 1113-1134, 2021 09.
Article in English | MEDLINE | ID: mdl-34190581

ABSTRACT

Cholangiocarcinoma (CCA) is a rare malignancy with generally dismal prognosis. Immunotherapy has revolutionized the management of cancer patients during the last decade, offering durable responses with an acceptable safety profile, but there are still no significant advances regarding CCA. Novel immunotherapeutic methods, such as cancer vaccines, oncolytic viruses, adoptive cell therapy and combinations of immune checkpoint inhibitors with other agents are currently under investigation and may improve prognosis. Efforts to find robust biomarkers for response are also ongoing. In this review, we discuss the rationale for the use of immunotherapy in CCA and available clinical data. Ongoing trials will also be presented, as well as key findings from each study.


Subject(s)
Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/immunology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/immunology , Immunotherapy/methods , Humans
SELECTION OF CITATIONS
SEARCH DETAIL