Next Article in Journal
An Early Increase in IL-10 and TNF-α Levels Following Atezolizumab Plus Bevacizumab Treatment Predicts Survival in Advanced Hepatocellular Carcinoma Patients: A Prospective Cohort Study
Previous Article in Journal
Psychosocial Burden of Women Who Are to Undergo Additional Diagnostic Procedures Due to Positive Screening for Cervical Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications

by
Angela Dalia Ricci
1,
Alessandro Rizzo
2,*,
Annalisa Schirizzi
3,
Rosalba D’Alessandro
3,
Giorgio Frega
4,
Giovanni Brandi
5,6,
Endrit Shahini
7,
Raffaele Cozzolongo
7,
Claudio Lotesoriere
1 and
Gianluigi Giannelli
8
1
Medical Oncology Unit, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
2
S.S.D. C.O.r.O. Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, Viale Orazio Flacco 65, 70124 Bari, Italy
3
Laboratory of Experimental Oncology, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
4
Osteoncology, Soft Tissue and Bone Sarcomas, Innovative Therapy Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
5
Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
6
Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy
7
Gastroenterology Unit, National Institute of Gastroenterology-IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy
8
Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, 70013 Castellana Grotte, Italy
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(20), 3542; https://doi.org/10.3390/cancers16203542
Submission received: 18 September 2024 / Revised: 6 October 2024 / Accepted: 18 October 2024 / Published: 20 October 2024
(This article belongs to the Section Cancer Therapy)

Simple Summary

Treatment options for intrahepatic cholangiocarcinoma (iCCA), a highly malignant tumor with poor prognosis, are limited. Recent developments in immunotherapy and immune checkpoint inhibitors (ICIs) have offered new hope for treating iCCA. However, several issues remain, including the identification of reliable biomarkers of response to ICIs and immune-based combinations. Tumor immune microenvironment (TIME) of these hepatobiliary tumors has been evaluated and is under assessment in order to boost the efficacy of ICIs and to convert these immunologically “cold” tumors to “hot” tumors. Herein, we examine the role of iCCA TIME, highlighting its mechanisms, current applications and challenges, and future research directions.

Abstract

Treatment options for intrahepatic cholangiocarcinoma (iCCA), a highly malignant tumor with poor prognosis, are limited. Recent developments in immunotherapy and immune checkpoint inhibitors (ICIs) have offered new hope for treating iCCA. However, several issues remain, including the identification of reliable biomarkers of response to ICIs and immune-based combinations. Tumor immune microenvironment (TIME) of these hepatobiliary tumors has been evaluated and is under assessment in this setting in order to boost the efficacy of ICIs and to convert these immunologically “cold” tumors to “hot” tumors. Herein, the review TIME of ICCA and its critical function in immunotherapy. Moreover, this paper also discusses potential avenues for future research, including novel targets for immunotherapy and emerging treatment plans aimed to increase the effectiveness of immunotherapy and survival rates for iCCA patients.

1. Introduction

Over the past few decades, there has been an increase in the incidence of intrahepatic cholangiocarcinoma (iCCA), a malignant tumor arising from the intrahepatic secondary bile ducts and their branches [1,2,3,4]. For most patients, the lack of early symptoms results in diagnosis at an advanced stage, and due to the complexity of biology and high heterogeneity of iCCA, traditional treatments like surgical resection, chemotherapy, and radiotherapy are frequently ineffective [5,6,7,8]. As a result, the 5-year survival rate following radical surgical resection is about 30% [9,10].
The development of cancer immunotherapy in the past twenty years has improved clinical outcome for a variety of hematological and solid tumors [11,12,13,14,15,16]. Since anticancer agents targeting programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), cytotoxic T-lymphocyte antigen 4 (CTLA-4), lymphocyte activation gene 3 (LAG-3), T-cell immunoglobulin domain and mucin domain 3 (TIM-3), and others have reported important results in phase I to III clinical trials [17,18,19]. Immunotherapy fights tumor cells by stimulating and boosting the patient’s immune system, and ICIs represent a fundamental avenue of research in this setting. These agents also offer new hope for biliary tract cancers, including iCCA [20,21,22]. However, unlike other solid tumors, iCCA has a different response to immunotherapy for several possible reasons, including distinct tumor immune microenvironment (TIME) and immune escape mechanisms [23,24,25]. Indeed, several issues remain, including the identification of reliable biomarkers of response to ICIs and immune-based combinations [26]. TIME of these hepatobiliary tumors has been evaluated and is under assessment in order to enhance the efficacy of ICIs and to convert these immunologically “cold” tumors to “hot” tumors [27,28]. However, despite a strong biological rationale and some preclinical reports, TIME is not part of everyday clinical practice in iCCA immunotherapy, and further efforts are needed.
In the current Review, we examine the role of iCCA TIME, highlighting its mechanisms, current applications, challenges, and future directions.

2. The Tumor Immune Microenvironment of Intrahepatic Cholangiocarcinoma

TIME of iCCA comprises a various range of cellular and non-cellular components that are extremely dynamic and intricate [29,30]. Immune cells, cancer cells, fibroblasts, and endothelial cells are among the cellular components of TIME, and these cells interact with each other through a variety of cytokines, chemotactic factors, and growth factors that influence tumor growth and invasion (Figure 1) [31]. Important roles are also played by non-cellular components such as the extracellular matrix (ECM) and soluble signaling mediators like Transforming Growth Factor β (TGF-β), a profibrogenic cytokine converting fibroblasts in myofibroblasts [32,33], and Platelet-Derived Growth Factor (PDGF), a pro-angiogenic and pro-fibrotic molecule [34]. The mutual interaction between tumor cells and their microenvironment results in the activation of Notch signaling pathways involved in the initiation and progression of intrahepatic cholangiocarcinoma. In particular, Notch was shown to be essential for the regulation of angiogenic sprouting and tumor vessel growth in conjunction with Vascular endothelial growth factor A (VEGF-A) [35]. Thus, the proliferating fibrous stroma within the iCCA TIME is highly reactive and has pro-tumorigenic, pro-angiogenic, and immunosuppressive properties that impair the effectiveness of anticancer therapies [36]. Thus, effective immunotherapeutic strategies would require a deep understanding of the cellular and non-cellular components of iCCA TIME and their interactions.

2.1. Cellular Elements of Tumor Immune Microenvironment

2.1.1. Cancer-Associated Fibroblasts

A number of studies have shown that iCCA is characterized by a prominent desmoplastic stroma, which is mainly enriched in cancer-associated fibroblasts (CAFs). CAFs are characterized by strong expression of alpha-smooth muscle actin (alpha-SMA), driven by TGF-β, and widely interact with iCCA cells and immune cells. This enhances the malignant phenotype and promotes tumor fibrosis and progression through several mechanisms, leading to therapy resistance [37,38]. Indeed, CAFs are involved in promoting lymphangiogenesis and angiogenesis and in the formation of a pro-inflammatory tumor microenvironment by secreting growth factors, cytokines, and several chemotactic factors, such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), and C-X-C motif chemokine ligand 12 (CXCL12) [38,39]. Furthermore, CAFs increase tumor invasiveness and growth by altering the ECM and releasing matrix metalloproteinases (MMPs) and other stromal proteins [40]. The remodeled and altered ECM acts as a barrier, limiting the infiltration of immune cells and is associated with reduced cytotoxic T-cell infiltration, increased tumor-associated macrophage (TAM) infiltration, and a poorer prognosis [41]. Hepatic stellate cells, portal fibroblasts, bone marrow mesenchymal stem cells, and fibroblasts derived from epithelial-mesenchymal transition and endothelial-mesenchymal transition are among the cells from which CAFs are activated in iCCA [42]. Recent research has shown that inducing apoptosis in CAFs with pharmacological agents, such as the BH3 mimetic navitoclax, can dramatically reduce tumor growth and prevent metastasis and lymphovascular invasion [43,44]. Moreover, CAFs have complex interactions with both immune and tumor cells, particularly through the CCL2-STAT3 signaling pathway [45]. These cells also exhibit immunosuppressive properties, including the inhibition of T-cell proliferation and function and the suppression of adaptive immune responses, which favor tumor immune escape [46]. Thus, studies and treatment strategies that focus on CAFs and their unique signaling pathways are key to advancing the knowledge of tumor biology and opening the door to novel immunotherapeutic advancements for iCCA.

2.1.2. Tumor-Associated Macrophages

The M2-type of tumor-associated macrophages (TAMs) is especially important for TIME [47]. Due to their primary involvement in angiogenesis and tissue repair, these macrophages play a role in the development of tumors [48]. M2-type TAMs secrete proinflammatory and pro-angiogenic factors, such as TNF-α, IL-6, TGF-β, and VEGF-A, which exacerbate tumor invasiveness and metastasis. In particular, these macrophages actively alter TIME through their interactions with cancer cells by promoting tumor growth and local invasiveness [49]. Additionally, by expressing inhibitory receptors like PD-1 and Siglec-10, which bind to ligands on tumor cells and facilitate tumor immune escape, M2-type TAMs inhibit the immune system’s phagocytic functions [50,51].
Recent reports have suggested a strong correlation between iCCA aggressiveness, increased Treg infiltration, and a grim prognosis when there is a high density of M2-type TAMs [52]. Thus, novel therapeutic approaches are being developed to fight this immunosuppressive state; these strategies include repolarizing M2-type TAMs to M1-type by activating the Notch pathway or employing interferon gamma (IFN-γ), which improves the TAMs’ antitumor properties [53]. Recent research has examined chimeric antigen receptor macrophages (CAR-M), which are genetically modified macrophages that express particular tumor antigen receptors [54]. Several reports have suggested that the effectiveness of immunotherapy could be increased when these CAR-M cells target and phagocytose tumor cells in addition to triggering T-cell responses [55,56,57]. This novel and multidisciplinary strategy opens up new treatment options for iCCA and has great promise to overcome the drawbacks of traditional immunotherapy.

2.1.3. Tumor-Associated Neutrophils

Tumor-associated neutrohils (TANs) play a key role in the TIME, despite their effect on the prognosis of patients, which is still to be defined [58]. While high TAN infiltration has been associated with poor prognosis, recent research suggests that high neutrophil infiltration may be associated with prolonged survival [59]. This disparity may be due to the different TAN phenotypes and behaviors within TIME, and some reports have highlighted that the two polarized TAN phenotypes, N1 and N2, have distinct functions at different stages of tumor progression: N1 shows antitumor characteristics in early stages, while N2 has immunosuppressive properties that promote tumor growth and metastasis in later stages [60]. The primary regulators of TAN function and polarization are TAM-secreted factors such as TGF-β and tumor cells [61]. By up-regulating factors such as arginase 1 (ARG1) and CCL2, TGF-β promotes tumor growth and the potential for metastasis by facilitating the conversion of N1-type TANs to N2-type [62]. Moreover, the function of TANs in boosting the metastatic potential of circulating tumor cells has been revealed by single-cell RNA sequencing [63]. Through the release of growth factors and cytokines, these N2-type TANs promote tumor immune escape by inhibiting effector T-cell function and stimulating angiogenesis [64]. Therapeutically, TAN polarization can be effectively modified and CD8+ T cell antitumor activity increased by exposing tumor-specific antigens with radiotherapy or TGF-β receptor inhibitors combined with granulocyte colony stimulating factor [65]. Based on these premises, neutrophil-based therapeutic approaches are set to become a rapidly evolving area of cancer therapy.

2.1.4. Myeloid-Derived Suppressor Cells

Myeloid-derived suppressor cells (MDSCs) are a subset of immature myeloid cells found in the TIME that mainly stimulate the growth and metastasis of tumors by inhibiting the actions of both innate and adaptive immune systems, with a specific focus on CD8+ T cells [66]. Their accumulation in TIME promotes tumor angiogenesis by secreting pro-angiogenic factors like VEGF, which inhibits T cell activation and function [67]. Moreover, by overexpressing PD-L1, which binds to PD-1 on T cells, MDSCs aid in the depletion of T cells and facilitate immune escape [68].
Due to their variety of functions, MDSCs are important potential targets for immunotherapy [69]. Research shows that MDSCs may prevent T cell activation in tumor patients by catabolizing or depleting cysteine and arginine, which inhibit T cell metabolism and lower the expression of homing receptors on T cell surfaces [70]. MDSC levels in the blood are markedly higher in iCCA patients, and they are highly correlated with the prognosis and severity of the disease [71]. In preclinical animal studies, strategies that target MDSCs, such as lowering their population by using anti-granulocyte differentiation antigen-1-specific antibodies, have shown promise in preventing tumor growth [72,73]. Additionally, decreasing the activation and aggregation of MDSCs at the tumor site may improve immunotherapy’s efficacy and provide iCCA patients with more effective treatment options [74].

2.1.5. Natural Killers

As essential members of the innate immune system, natural killers (NKs) have been shown to have strong antitumor activity, and in the absence of previous antigen exposure, these cells are non-specifically able to identify and attack cancer cells [75,76]. NKs comprise approximately 30–40% of all lymphocytes in the liver, and they mainly use cytotoxic factors such as FasL and TRAIL, as well as cytokines like TNF-α and IFN-γ, to achieve their antitumor effects [77,78]. Furthermore, NKs play a critical role in controlling tumor cell growth and metastasis as well as inducing apoptosis. According to recent research, increasing the number or functionality of NKs may effectively halt the advancement of iCCA, establishing them as possible targets for treatment [79,80].
Some reports have suggested a strong correlation between the prognosis of iCCA patients and the expression of the NK activation receptor NKG2D [81]. Patients with higher expression levels of NKG2D typically have better prognoses, probably because NKG2D helps NKs to identify and destroy cancer cells. Furthermore, by triggering apoptosis in tumor cells and by regulating adaptive immune responses, NKs show promise in precision immunotherapy by lowering off-target effects through cytokine release [82]. The promise of NKs-based immunotherapeutic strategies has been highlighted by experimental studies that suggest a significant inhibition of tumor growth following the infusion of human NKs into a xenograft mouse model by using the human CCA cell line HuCCT-1 [83,84].

2.1.6. Tumor-Infiltrating Lymphocytes (TILs)

Tumor-Infiltrating Lymphocytes (TILs) are immune cells that directly participate in the immune response against tumor cells [82]. They are composed of different subpopulations, such as CD8+ cytotoxic T cells and CD20+ B cells, and they play a complex immunomodulatory role within the TIME [85]. Research has indicated that TILs are able to identify tumor antigens and trigger immune responses against tumors [86,87]. Through encouraging TIL apopoptosis, certain signaling pathways, such as TGF-β, PD-1/PD-L1, and Wnt/β-catenin, boost tumor immune escape. While extensive CD4+ T cell infiltration is correlated with longer overall and recurrence-free survival, high levels of CD8+ TILs are generally linked to improved patient survival [88].
There is a great deal of variation in the TIME in terms of TIL types and functions. For example, CD4+ T cells are more frequently located in the periphery of the tumor, whereas CD8+ TILs usually concentrate in the inner regions of the tumor [89,90]. Because they are unable to produce the proteases necessary to break down the extracellular matrix, CD8+ T cells are unable to permeate dense tumor tissue, although they do tend to concentrate at sparse fibrous junctions and the collagen-rich peripheral matrix [91]. Furthermore, the density and orientation of the matrix structures may have an impact on the distribution of TILs within TIME.
Tumor-associated antigens (TAAs) found in iCCA may be targets for cancer vaccines that seek to boost TILs’ immune responses [92]. The T cell populations in iCCA vary, according to recent transcriptome studies. Notably, CD8+ TILs from highly heterogeneous tumors show reduced cytotoxic potential [93,94]. Furthermore, a distribution that is associated with a worse prognosis was noted: CD8+ TILs are mainly found at the invasive margins of tumors, whereas Tregs and IL-17-positive immune cells are more common inside the tumor [95,96]. In addition, FOXP3, a transcription factor that is primarily overexpressed by Tregs, is linked to elevated CTLA-4 expression and may be a sign of chemoresistance and tumor recurrence. Tregs in TIME alter the phenotypic of the cells by triggering CCL2, which causes the release of immunosuppressive substances such as IL-10 and TGF-β [97]. This encourages tumor immune evasion and inhibits effector T-cell activity. To develop more potent immunotherapeutic strategies, a thorough comprehension of the functional and regulatory mechanisms targeting TILs is essential, especially in overcoming the immunosuppressive state in iCCA.

2.1.7. Dendritic Cells

As specialized antigen-presenting cells, dendritic cells (DCs) are essential elements of the TIME of iCCA [98]. These cells have excellent antigen-uptake, processing, and presentation skills. While mature DCs are good at activating initial T and B cells—which are essential for starting, regulating, and maintaining the immune response—mature DCs are highly capable of migrating [99]. Mature DCs (CD83+) are primarily located at the invasive tumor margins in iCCA, while a considerable number of immature DCs (CD1a+) are found inside the tumor [100,101]. A favorable prognosis is closely associated with the presence of mature DCs, which positively correlate with the quantity of CD4+/CD8+ cells in TIME. This enhances T cell activation and the antitumor immune response [102].
On the other hand, TIME has a significantly lower DC density, which may be due to a decrease in the amounts of chemokines (like CCL4 and CCL5) that attract DCs [103]. These chemokines are usually generated by lymphocytes, including NKs. Increased TIME concentrations of TGF-β, adenosine, IL-10, and hypoxia can induce DCs to exhibit an immunosuppressive phenotype [104,105]. This phenotype promotes the recruitment of MDSCs, M2-type TAMs, N2-type TANs, and the polarization of Tregs in addition to counteracting the tumor-fighting effects of effector T cells through immune checkpoint ligands and causing T cell depletion [106,107]. It does this by secreting IL-6 and triggering the production of TGF-β.
Based on these premises, there are several opportunities for tumor vaccine developments, according to the characteristics of DCs [108]. Clinical trials have tested tumor vaccine technologies for some solid tumors, including prostate cancer and melanoma [109,110,111,112]. These technologies take advantage of the properties of DCs to load tumor antigens and to activate immune effector cells. Enzyme-loaded DCs have enhanced CD3+ lymphocyte infiltration in iCCA animal models, which effectively inhibits tumor growth and metastasis [113,114]. These results suggest that improving DCs’ capacity for immune activation and antigen presentation may increase the effectiveness of immunotherapy against iCCA, especially when used in combination with other immunomodulatory techniques.

2.2. Non-Cellular Elements of Tumor Immune Microenvironment

2.2.1. Extracellular Matrix

In order to facilitate the structural remodeling of tumor tissues through the release of essential components like MMPs, osteoprotegerin, junctional protein-C, and osteonectin, extensive reorganization of the ECM is essential for tumorigenesis and progression [115]. The growth of tumors, increased lymph node metastasis, and overall survival are all strongly correlated with these components’ expression. Through the activation of the RAS-RAF-MEK-ERK and Wnt/β-catenin signaling pathways, elevated osteonectin expression in TIME of iCCA correlates significantly with tumor size, local and distant invasion, and advanced cancer stages [116,117]. Furthermore, osteonectin promotes T cell survival and NK development, highlighting the diverse functions of the ECM in controlling the tumor immune milieu.
Increased ECM stiffness is found to be a characteristic of tumor progression in iCCA studies [118,119,120]. In addition to stimulating cell proliferation, survival, migration, and differentiation, which in turn drives tumor progression, ECM’s increased stiffness also affects tumor behavior through the extracellular vesicles that contain proteins, lipids, and nucleic acids secreted by tumor cells [121]. These vesicles represent a complex and important mechanism in the progression of tumors, as they contribute to immune regulation, tumor angiogenesis, and the reconstitution of TIME [122]. Targeting ECM may have therapeutic benefits, and a particular goal of this research is to improve the prognosis and outcomes of iCCA patients by modifying important ECM elements.

2.2.2. Chemokines

A subclass of small-molecule cytokines called chemokines controls the chemotactic migration of leukocytes, including T lymphocytes, macrophages, and monocytes [123,124]. They are essential for the development of iCCAs because chemokines influence the migration, invasion, and immune escape of cancer cells [125]. Chemokines that facilitate the necessary oxygen and nutrient supply to tumors, such as CCL11, CCL2, CXCL1-CXCL3, CXCL5-CXCL6, CXCL8-CXCL12, and CCL16, either directly or indirectly, promote angiogenesis [126]. To be more precise, angiotensin II and TGF-β both negatively and positively regulate CXCL12 in iCCA, while CXCR4 expression is encouraged by TAM-secreted TNF-α. The activation of multiple signaling pathways, such as the Wnt/β-catenin, extracellular signal-regulated kinase (ERK1/2), and phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) pathways, is facilitated by these chemokines through their specific receptor interactions [127,128,129].
For example, fibroblast activating protein (FAP)-induced CCL2 controls the migration of MDSCs and macrophages, while CXCL5 functions as a chemokine for neutrophils [130]. Chemokine-targeting clinical trials are currently being conducted to investigate the clinical utility of chemokines in oncology treatment, ranging from hematological malignancies and breast cancer to hepatobiliary tumors like iCCA [131,132]. The results of these studies may provide therapeutic targets and important biological insights for the development of novel immunotherapeutic approaches to treat iCCA [133,134].

2.2.3. Extracellular Vesicles

Extracellular Vesicles (EVs) are microscopic membrane structures that serve as messengers between cells and are essential for TIME [135]. These vesicles can be divided into three sizes based on their diameter: exosomes (30–100 nm in diameter), microvesicles (0.1–1 μm), and apoptotic bodies (greater than 1 μm in diameter). The latter are usually removed by phagocytosis shortly after their release [136]. Proteins, lipids, messenger RNAs (mRNAs), microRNAs (miRNAs), long noncoding RNAs (lnRNAs), and circular RNAs (circRNAs) are the constituents of EVs, and the lipid bilayer of the vesicles protects these components from enzymatic degradation [137].
EVs in iCCA stimulate mesenchymal stem cell migration and the release of different cytokines and chemokines that aid in the growth of the tumor [138]. For instance, α-smooth muscle actin and other markers can be expressed by EVs derived from iCCAs [139]. Additionally, EVs are thought to be possible new therapeutic targets because they promote interactions between iCCA cells. Research has suggested that plasminogen activator inhibitor 2 (PAI-2) secretion is enhanced when mRNA-15a is downregulated in CAFs, which in turn promotes tumor cell migration [140,141]. The therapeutic potential of miRNAs has been demonstrated in animal models through the infusion of miRNA-195-enriched EVs, which has been shown to reduce tumor size and improve survival [142,143]. These results highlight the possible role of extracellular vesicles in iCCA immunotherapy approaches and may pave the way for the creation of new treatments that specifically target these molecular messengers.

2.2.4. Platelet-Derived Growth Factor

Angiogenesis, fibrosis, and tumor growth are all significantly aided by PDGF [144,145]. Being a strong mitogen, it increases the fibroblast and smooth muscle cell proliferation in peripheral support tissues, which improves the vascular density and structural integrity of tumors [146]. According to recent research, PDGF stimulates tumor cells directly and through signaling pathways like PI3K/Akt and MAPK to increase tumor cell survival and proliferation [147,148]. Furthermore, there is a strong correlation between the prognosis of iCCA patients and high levels of PDGF in TIME; higher levels generally correspond to worse survival and disease progression [149]. As a result, efforts are being made to develop targeted therapeutic approaches against PDGF and its receptors in order to block the growth factor’s pathway, inhibit tumor growth signals, and investigate novel treatment options for iCCA [150,151].
One of these strategies is the application of particular inhibitors, such as imatinib, a small-molecule drug that effectively inhibits the PDGF receptor and has demonstrated potential efficacy in clinical trials for a number of cancers [152,153].

2.2.5. Notch Signaling Pathway

A number of experimental studies suggest a central role for Notch signaling in cholangiocarcinogenesis, acting on tumor cell proliferation, survival, and migration [154]. The canonical Notch signaling pathway in iCCA is known to be a key player in direct cell-cell interactions between cancer cells and the surrounding stromal cells of the microenvironment [155] and has also been shown to be essential for modulating angiogenic sprouting and blood vessel growth in cooperation with VEGFA. Indeed, it has been shown that Crenigacestat, a selective NOTCH1 inhibitor, reduces tumorigenesis in vivo by downregulating the gene expression and protein levels of VEGFA and matrix metalloproteinase-13 (MMP13) [35]. In this context, further studies were conducted in patient-derived xenografts (PDX), and transcriptomic analysis demonstrated that in Crenigacestat-treated mice there was a downregulation of cluster differentiation 90 (CD90), a critical regulator of cell adhesion, communication, and cell-ECM interaction [37]. An intriguing relationship was found between Notch and CD90, which is closely associated with poor prognosis and shorter survival in iCCA patients. Thus, in a subset of patients with iCCA, CD90 is a potential molecular target for treatment. This is the first demonstrated evidence for molecular stratification of iCCA to predict clinical outcomes. [156]. In addition, it has recently been reported that the Notch signaling pathway also interacts with TGF-β in cholangiocarcinogenesis. Specifically, Crenigacestat treatment of patient-derived CAFs inhibits both Notch and TGF-β signaling. This inhibition led to the inactivation of CAFs, resulting in a decrease in the secretion of ECM proteins [157]. Overall, these works showed that inhibition of Notch signaling is effective at two levels, not only reducing tumorigenesis and angiogenesis but also restoring homeostasis in the microenvironment surrounding iCCA with a reduction of desmoplastic reaction [158]. This suggests a potential new strategy to fight tumor progression. A Notch inhibitor, which reduces the desmoplastic reaction and angiogenesis, could be combined with other drugs, such as immune checkpoint inhibitors, which activate the immune response, to increase their effectiveness.

3. Immune Evasion and Immunophenotyping in Intrahepatic Cholangiocarcinoma

Tumor cells can avoid immune monitoring and elimination by using an intricate network of immune escape mechanisms, which is employed by iCCA (Figure 2) [159]. The tumor cells of ICCA specifically work to thwart immune attacks by modifying surface antigens and increasing the levels of immunosuppressive molecules [160]. PD-L1, for example, is highly expressed in iCCA cells and binds to PD-1 on T cells to prevent T cell activation and suppress immune responses. In addition, TILs have been shown to express higher levels of CTLA-4, a protein that is linked to aggressive tumor behavior and a poor prognosis [161]. In addition to interfering with metabolic and cell signaling pathways, the expression of these checkpoint molecules also promotes immune tolerance and depletes peripheral T cells.
The interaction between tumor cells and immunosuppressive cells such as MDSCs and TAMs strengthens the immune escape mechanisms within TIME. By releasing inhibitory molecules like arginase and inducible nitric oxide synthase (iNOS), these cells block T cell metabolism and signaling. Furthermore, research suggests that increased PD-L1 expression in iCCA is associated with worse clinical outcomes, such as shorter survival [162]. In approximately 45% of iCCA samples, for example, immune checkpoint molecules were found to be upregulated, indicating a worse prognosis, according to a report published by Nakamura and colleagues [163]. Further investigation has revealed that one of the most important mechanisms for immune escape in iCCA is the control of PD-1 and PD-L1. For example, a study by Gani et al. found that overall survival was 60% lower in 72% of iCCA patients with PD-L1 expression than in those without PD-L1 expression [164]. Furthermore, poorer survival and lymph node metastasis were associated with FOXP3 overexpression in iCCA cells, which is frequently associated with elevated CTLA-4 levels. These findings highlight the detrimental prognostic effect of CTLA-4 in iCCA and highlight the critical function of immune checkpoint molecules in the immune escape mechanism of iCCA.
The relationship between the immunophenotype of a tumor and its biological behavior as well as prognosis has been increasingly clear from studies on immune subtypes [165]. Gene expression profiling has recently led to the classification of iCCA into four immune subtypes: immune-indifferent (I1), with very low levels of immune cell infiltration; immune-activated (I2), marked by substantial infiltration of CD4+, CD8+, and CD45RO+ lymphocytes; myeloid-derived (I3), primarily made up of monocytes and macrophages; and mesenchymal (I4), which is characterized by minimal immune infiltration but significant activation of macrophages and monocytes [166]. The immune-activated subtype (I2) is the most promising among them, showing great promise for immunotherapy and a comparatively better prognosis. Because of this classification, iCCA patients may be eligible for customized immunotherapy based on their unique immune profiles [167]. The molecular immunophenotypes of iCCA have also been further improved by multi-omics-based investigations. In a large-sample genome sequencing study, for example, iCCA was classified into two types: proliferative, which indicates a worse prognosis and is characterized by increased activity in tumor proliferation signaling pathways; and inflammatory, which is characterized by enriched immune signals like interleukins and chemokines [168].
These results provide a theoretical framework for more specialized therapies catered to the various molecular subtypes of iCCA [169,170]. Based on protein expression, recent proteomic analyses identified molecular types, which include inflammatory (S1), mesenchymal (S2), metabolic (S3), and differentiated (S4). Determining these subtypes facilitates accurate tumor diagnosis and treatment, and these studies show that combining data from immunology and molecular biology can improve our knowledge of the biology of iCCA and make it easier to create individualized treatment regimens for individual patients.

4. Implications for Treatment and Future Research Avenues

High-throughput sequencing technology has made significant advancements in iCCA molecular typing in recent years. Several classification techniques based on gene mutations (e.g., IDH1/2, FGFR2, TP53), gene expression profiles, and epigenetic modifications are commonly used in molecular typing of iCCA [171]. Research has demonstrated, for instance, that iCCA with IDH1/2 mutations has particular metabolic characteristics and a different TIME, and similarly, FGFR2 fusion gene-positive iCCA shows unique growth patterns and drug response [172]. Based on factors such as immunosuppressive molecule expression, cytokine secretion, and the extent of immune cell infiltration in the tumor tissue, the immune microenvironment can be broadly categorized as “inflammatory” or “non-inflammatory” [173,174].
A non-inflammatory TIME is characterized by enhanced immune escape mechanisms, such as high PD-L1 expression and T-cell depletion, whereas an inflammatory TIME is typically characterized by high levels of T-cell infiltration and immunoreactivity. Certain immune microenvironmental phenotypes are closely linked to distinct molecular subtypes of iCCA [175]. For instance, IDH1/2 mutant iCCA is frequently associated with increased expression of immunosuppressive molecules and decreased T-cell infiltration; iCCA with the FGFR2 fusion gene, on the other hand, exhibits greater levels of immune cell infiltration and might be more amenable to immunotherapy [176,177]. Immunotherapeutic research in iCCA is developing quickly, posing a wide range of research opportunities, and encountering many obstacles. The ongoing investigation of novel immune checkpoints and other immunotherapeutic targets is expected to broaden the range of potential iCCA treatment approaches. Investigating these novel targets holds the potential to enable customized treatment regimens utilizing precision medicine tools like immunomics and genomics. With this method, “cold tumors” could become “hot tumors,” increasing the immune system’s capacity to fight cancer by better stimulating the body’s immune response [177]. Additionally, the focus of iCCA immunotherapy research in the future will be on creating integrative treatment plans. To combat both primary and acquired tumor resistance, these approaches could combine immunotherapy with other treatments like chemotherapy and targeted therapies, since ICIs have been demonstrated to improve treatment efficacy when combined with other antitumor agents [178,179,180,181,182,183]. Immunotherapy provides new hope for treating iCCA and has recently become a central strategy in increasing the prognosis of iCCA patients; however, several issues are to be considered, including the lack of validated predictive biomarkers that could help to identify patient groups that are most likely to benefit from such therapeutic approaches. Future research must focus on the development of novel agents and combinations, but the validation of biomarkers remains an urgent need.

5. Conclusions

While immunotherapy offers a new hope for the treatment of iCCA, the lack of validated predictive biomarkers to determine which patient groups stand to gain the most from these treatments remains an issue. Therefore, to improve the accuracy of therapeutic decisions and boost treatment efficacy, future research must focus on the development and validation of biomarkers. Immunotherapy is set to become a key element in improving the prognosis for patients with iCCA thanks to such extensive research efforts aimed at further comprehending the importance of TIME in this setting.

Author Contributions

Conceptualization, A.D.R. and A.R.; methodology, A.D.R. and A.R.; formal analysis, A.D.R. and A.R.; resources, A.D.R. and A.R.; data curation, A.D.R. and A.R.; writing—original draft preparation, A.D.R. and A.R.; writing—review and editing, all authors; visualization, all authors; supervision, all authors. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest. The authors affiliated to the IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy, are responsible for the views expressed in this article, which do not necessarily represent the Institute.

References

  1. Valle, J.W.; Kelley, R.K.; Nervi, B.; Oh, D.Y.; Zhu, A.X. Biliary tract cancer. Lancet 2021, 397, 428–444. [Google Scholar] [CrossRef] [PubMed]
  2. Lamarca, A.; Edeline, J.; Goyal, L. How I treat biliary tract cancer. ESMO Open 2022, 7, 100378. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  3. Rizzo, A.; Brandi, G. BILCAP trial and adjuvant capecitabine in resectable biliary tract cancer: Reflections on a standard of care. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 483–485. [Google Scholar] [CrossRef] [PubMed]
  4. Lamarca, A.; Barriuso, J.; McNamara, M.G.; Valle, J.W. Molecular targeted therapies: Ready for “prime time” in biliary tract cancer. J. Hepatol. 2020, 73, 170–185. [Google Scholar] [CrossRef] [PubMed]
  5. Scott, A.J.; Sharman, R.; Shroff, R.T. Precision Medicine in Biliary Tract Cancer. J. Clin. Oncol. 2022, 40, 2716–2734. [Google Scholar] [CrossRef] [PubMed]
  6. Bridgewater, J.A.; Goodman, K.A.; Kalyan, A.; Mulcahy, M.F. Biliary Tract Cancer: Epidemiology, Radiotherapy, and Molecular Profiling. Am. Soc. Clin. Oncol. Educ. Book 2016, 35, e194–e203. [Google Scholar] [CrossRef] [PubMed]
  7. Jain, A.; Kwong, L.N.; Javle, M. Genomic Profiling of Biliary Tract Cancers and Implications for Clinical Practice. Curr. Treat. Options Oncol. 2016, 17, 58. [Google Scholar] [CrossRef] [PubMed]
  8. Rizzo, A.; Brandi, G. Pitfalls, challenges, and updates in adjuvant systemic treatment for resected biliary tract cancer. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 547–554. [Google Scholar] [CrossRef] [PubMed]
  9. Lee, S.H.; Song, S.Y. Recent Advancement in Diagnosis of Biliary Tract Cancer through Pathological and Molecular Classifications. Cancers 2024, 16, 1761. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  10. Rizzo, A.; Brandi, G. Neoadjuvant therapy for cholangiocarcinoma: A comprehensive literature review. Cancer Treat. Res. Commun. 2021, 27, 100354. [Google Scholar] [CrossRef] [PubMed]
  11. Riley, R.S.; June, C.H.; Langer, R.; Mitchell, M.J. Delivery technologies for cancer immunotherapy. Nat. Rev. Drug Discov. 2019, 18, 175–196. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  12. Zhang, Y.; Zhang, Z. The history and advances in cancer immunotherapy: Understanding the characteristics of tumor-infiltrating immune cells and their therapeutic implications. Cell. Mol. Immunol. 2020, 17, 807–821. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  13. Guven, D.C.; Erul, E.; Kaygusuz, Y.; Akagunduz, B.; Kilickap, S.; De Luca, R.; Rizzo, A. Immune checkpoint inhibitor-related hearing loss: A systematic review and analysis of individual patient data. Support. Care Cancer 2023, 31, 624. [Google Scholar] [CrossRef] [PubMed]
  14. Abbott, M.; Ustoyev, Y. Cancer and the Immune System: The History and Background of Immunotherapy. Semin. Oncol. Nurs. 2019, 35, 150923. [Google Scholar] [CrossRef] [PubMed]
  15. Rizzo, A. Identifying optimal first-line treatment for advanced non-small cell lung carcinoma with high PD-L1 expression: A matter of debate. Br. J. Cancer 2022, 127, 1381–1382. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  16. Cha, J.H.; Chan, L.C.; Song, M.S.; Hung, M.C. New Approaches on Cancer Immunotherapy. Cold Spring Harb. Perspect. Med. 2020, 10, a036863. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  17. Reck, M.; Rodríguez-Abreu, D.; Robinson, A.G.; Hui, R.; Csőszi, T.; Fülöp, A.; Gottfried, M.; Peled, N.; Tafreshi, A.; Cuffe, S.; et al. Updated Analysis of KEYNOTE-024: Pembrolizumab Versus Platinum-Based Chemotherapy for Advanced Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score of 50% or Greater. J. Clin. Oncol. 2019, 37, 537–546. [Google Scholar] [CrossRef] [PubMed]
  18. Bischoff, P.; Reck, M.; Overbeck, T.; Christopoulos, P.; Rittmeyer, A.; Lüders, H.; Kollmeier, J.; Kulhavy, J.; Kemper, M.; Reinmuth, N.; et al. Outcome of First-Line Treatment With Pembrolizumab According to KRAS/TP53 Mutational Status for Nonsquamous Programmed Death-Ligand 1-High (≥50%) NSCLC in the German National Network Genomic Medicine Lung Cancer. J. Thorac. Oncol. 2024, 19, 803–817. [Google Scholar] [CrossRef] [PubMed]
  19. Rizzo, A.; Mollica, V.; Massari, F. Expression of Programmed Cell Death Ligand 1 as a Predictive Biomarker in Metastatic Urothelial Carcinoma Patients Treated with First-line Immune Checkpoint Inhibitors Versus Chemotherapy: A Systematic Review and Meta-analysis. Eur. Urol. Focus 2022, 8, 152–159. [Google Scholar] [CrossRef] [PubMed]
  20. Chen, X.; Wang, D.; Liu, J.; Qiu, J.; Zhou, J.; Ying, J.; Shi, Y.; Wang, Z.; Lou, H.; Cui, J.; et al. Genomic alterations in biliary tract cancer predict prognosis and immunotherapy outcomes. J. Immunother. Cancer. 2021, 9, e003214. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  21. Ricci, A.D.; Rizzo, A.; Brandi, G. Immunotherapy in Biliary Tract Cancer: Worthy of a Second Look. Cancer Control 2020, 27, 1073274820948047. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  22. Queiroz, M.M.; Lima, N.F., Jr.; Biachi de Castria, T. Immunotherapy and Targeted Therapy for Advanced Biliary Tract Cancer: Adding New Flavors to the Pizza. Cancers 2023, 15, 1970. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  23. Yue, S.; Zhang, Y.; Zhang, W. Recent Advances in Immunotherapy for Advanced Biliary Tract Cancer. Curr. Treat. Options Oncol. 2024, 25, 1089–1111. [Google Scholar] [CrossRef] [PubMed]
  24. Harding, J.J.; Khalil, D.N.; Fabris, L.; Abou-Alfa, G.K. Rational development of combination therapies for biliary tract cancers. J. Hepatol. 2023, 78, 217–228. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  25. Neureiter, D.; Mayr, C.; Kiesslich, T. The challenges of combinatory immunotherapy for biliary tract cancer. Expert. Opin. Investig. Drugs 2021, 30, 591–594. [Google Scholar] [CrossRef] [PubMed]
  26. Carloni, R.; Rizzo, A.; Ricci, A.D.; Federico, A.D.; De Luca, R.; Guven, D.C.; Yalcin, S.; Brandi, G. Targeting tumor microenvironment for cholangiocarcinoma: Opportunities for precision medicine. Transl. Oncol. 2022, 25, 101514. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  27. Kam, A.E.; Masood, A.; Shroff, R.T. Current and emerging therapies for advanced biliary tract cancers. Lancet Gastroenterol. Hepatol. 2021, 6, 956–969. [Google Scholar] [CrossRef] [PubMed]
  28. Rizzo, A.; Ricci, A.D.; Brandi, G. PD-L1, TMB, MSI, and Other Predictors of Response to Immune Checkpoint Inhibitors in Biliary Tract Cancer. Cancers 2021, 13, 558. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  29. Job, S.; Rapoud, D.; Dos Santos, A.; Gonzalez, P.; Desterke, C.; Pascal, G.; Elarouci, N.; Ayadi, M.; Adam, R.; Azoulay, D.; et al. Identification of Four Immune Subtypes Characterized by Distinct Composition and Functions of Tumor Microenvironment in Intrahepatic Cholangiocarcinoma. Hepatology 2020, 72, 965–981. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  30. Gao, B.; Wang, Y.; Zhang, X.; Jiang, H.; Han, F.; Li, C.; Lu, S. Identification and validation of inflammatory subtypes in intrahepatic cholangiocellular carcinoma. J. Transl. Med. 2024, 22, 730. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  31. Cheng, K.; Cai, N.; Zhu, J.; Yang, X.; Liang, H.; Zhang, W. Tumor-associated macrophages in liver cancer: From mechanisms to therapy. Cancer Commun. 2022, 42, 1112–1140. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  32. Clapéron, A.; Mergey, M.; Aoudjehane, L.; Ho-Bouldoires, T.H.N.; Wendum, D.; Prignon, A.; Merabtene, F.; Firrincieli, D.; Desbois-Mouthon, C.; Scatton, O.; et al. Hepatic myofibroblasts promote the progression of human cholangiocarcinoma through activation of epidermal growth factor receptor. Hepatology 2013, 58, 2001–2011. [Google Scholar] [CrossRef] [PubMed]
  33. Sung, P.S. Crosstalk between tumor-associated macrophages and neighboring cells in hepatocellular carcinoma. Clin. Mol. Hepatol. 2022, 28, 333–350. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  34. Wang, C.; Chen, C.; Hu, W.; Tao, L.; Chen, J. Revealing the role of necroptosis microenvironment: FCGBP + tumor-associated macrophages drive primary liver cancer differentiation towards cHCC-CCA or iCCA. Apoptosis 2024, 29, 460–481. [Google Scholar] [CrossRef] [PubMed]
  35. Mancarella, S.; Serino, G.; Dituri, F.; Cigliano, A.; Ribback, S.; Wang, J.; Chen, X.; Calvisi, D.F.; Giannelli, G. Crenigacestat, a selective NOTCH1 inhibitor, reduces intrahepatic cholangiocarcinoma progression by blocking VEGFA/DLL4/MMP13 axis. Cell Death Differ. 2020, 27, 2330–2343. [Google Scholar] [CrossRef]
  36. Czekay, R.P.; Cheon, D.J.; Samarakoon, R.; Kutz, S.M.; Higgins, P.J. Cancer-Associated Fibroblasts: Mechanisms of Tumor Progression and Novel Therapeutic Targets. Cancers 2022, 14, 1231. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  37. Mancarella, S.; Serino, G.; Coletta, S.; Armentano, R.; Dituri, F.; Ardito, F.; Ruzzenente, A.; Fabregat, I.; Giannelli, G. The Tumor Microenvironment Drives Intrahepatic Cholangiocarcinoma Progression. Int. J. Mol. Sci. 2022, 23, 4187. [Google Scholar] [CrossRef]
  38. Khan, G.J.; Sun, L.; Khan, S.; Yuan, S.; Nongyue, H. Versatility of Cancer Associated Fibroblasts: Commendable Targets for Anti-tumor Therapy. Curr. Drug Targets 2018, 19, 1573–1588. [Google Scholar] [CrossRef] [PubMed]
  39. Belhabib, I.; Zaghdoudi, S.; Lac, C.; Bousquet, C.; Jean, C. Extracellular Matrices and Cancer-Associated Fibroblasts: Targets for Cancer Diagnosis and Therapy? Cancers 2021, 13, 3466. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  40. Lin, Y.; Cai, Q.; Chen, Y.; Shi, T.; Liu, W.; Mao, L.; Deng, B.; Ying, Z.; Gao, Y.; Luo, H.; et al. CAFs shape myeloid-derived suppressor cells to promote stemness of intrahepatic cholangiocarcinoma through 5-lipoxygenase. Hepatology 2022, 75, 28–42. [Google Scholar] [CrossRef] [PubMed]
  41. Fabris, L.; Perugorria, M.J.; Mertens, J.; Björkström, N.K.; Cramer, T.; Lleo, A.; Solinas, A.; Sänger, H.; Lukacs-Kornek, V.; Moncsek, A.; et al. The tumour microenvironment and immune milieu of cholangiocarcinoma. Liver Int. 2019, 39 (Suppl. S1), 63–78. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  42. Yang, X.; Lin, Y.; Shi, Y.; Li, B.; Liu, W.; Yin, W.; Dang, Y.; Chu, Y.; Fan, J.; He, R. FAP Promotes Immunosuppression by Cancer-Associated Fibroblasts in the Tumor Microenvironment via STAT3-CCL2 Signaling. Cancer Res. 2016, 76, 4124–4135. [Google Scholar] [CrossRef] [PubMed]
  43. Biffi, G.; Tuveson, D.A. Diversity and Biology of Cancer-Associated Fibroblasts. Physiol. Rev. 2021, 101, 147–176. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  44. Lavie, D.; Ben-Shmuel, A.; Erez, N.; Scherz-Shouval, R. Cancer-associated fibroblasts in the single-cell era. Nat. Cancer. 2022, 3, 793–807. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  45. Chen, Y.; McAndrews, K.M.; Kalluri, R. Clinical and therapeutic relevance of cancer-associated fibroblasts. Nat. Rev. Clin. Oncol. 2021, 18, 792–804. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  46. Kennel, K.B.; Bozlar, M.; De Valk, A.F.; Greten, F.R. Cancer-Associated Fibroblasts in Inflammation and Antitumor Immunity. Clin. Cancer Res. 2023, 29, 1009–1016. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  47. Cencini, E.; Sicuranza, A.; Ciofini, S.; Fabbri, A.; Bocchia, M.; Gozzetti, A. Tumor-Associated Macrophages in Multiple Myeloma: Key Role in Disease Biology and Potential Therapeutic Implications. Curr. Oncol. 2023, 30, 6111–6133. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  48. Rihawi, K.; Ricci, A.D.; Rizzo, A.; Brocchi, S.; Marasco, G.; Pastore, L.V.; Llimpe, F.L.R.; Golfieri, R.; Renzulli, M. Tumor-Associated Macrophages and Inflammatory Microenvironment in Gastric Cancer: Novel Translational Implications. Int. J. Mol. Sci. 2021, 22, 3805. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  49. Zhang, X.; Sun, Y.; Ma, Y.; Gao, C.; Zhang, Y.; Yang, X.; Zhao, X.; Wang, W.; Wang, L. Tumor-associated M2 macrophages in the immune microenvironment influence the progression of renal clear cell carcinoma by regulating M2 macrophage-associated genes. Front. Oncol. 2023, 13, 1157861. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  50. Nam, S.J.; Kim, S.; Kwon, D.; Kim, H.; Kim, S.; Lee, E.; Kim, T.M.; Heo, D.S.; Park, S.H.; Lim, M.S.; et al. Prognostic implications of tumor-infiltrating macrophages, M2 macrophages, regulatory T-cells, and indoleamine 2,3-dioxygenase-positive cells in primary diffuse large B-cell lymphoma of the central nervous system. Oncoimmunology 2018, 7, e1442164. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  51. Vadevoo, S.M.P.; Gunassekaran, G.R.; Yoo, J.D.; Kwon, T.H.; Hur, K.; Chae, S.; Lee, B. Epigenetic therapy reprograms M2-type tumor-associated macrophages into an M1-like phenotype by upregulating miR-7083-5p. Front. Immunol. 2022, 13, 976196. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  52. Luo, C.; Xin, H.; Zhou, Z.; Hu, Z.; Sun, R.; Yao, N.; Sun, Q.; Borjigin, U.; Wu, X.; Fan, J.; et al. Tumor-derived exosomes induce immunosuppressive macrophages to foster intrahepatic cholangiocarcinoma progression. Hepatology 2022, 76, 982–999. [Google Scholar] [CrossRef] [PubMed]
  53. Zhou, Z.; Wang, P.; Sun, R.; Li, J.; Hu, Z.; Xin, H.; Luo, C.; Zhou, J.; Fan, J.; Zhou, S. Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J. Immunother. Cancer 2021, 9, e001946. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  54. Maalej, K.M.; Merhi, M.; Inchakalody, V.P.; Mestiri, S.; Alam, M.; Maccalli, C.; Cherif, H.; Uddin, S.; Steinhoff, M.; Marincola, F.M.; et al. CAR-cell therapy in the era of solid tumor treatment: Current challenges and emerging therapeutic advances. Mol. Cancer 2023, 22, 20. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  55. Klichinsky, M.; Ruella, M.; Shestova, O.; Lu, X.M.; Best, A.; Zeeman, M.; Schmierer, M.; Gabrusiewicz, K.; Anderson, N.R.; Petty, N.E.; et al. Human chimeric antigen receptor macrophages for cancer immunotherapy. Nat. Biotechnol. 2020, 38, 947–953. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  56. Shin, M.H.; Oh, E.; Kim, Y.; Nam, D.H.; Jeon, S.Y.; Yu, J.H.; Minn, D. Recent Advances in CAR-Based Solid Tumor Immunotherapy. Cells 2023, 12, 1606. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  57. Chen, Y.; Yu, Z.; Tan, X.; Jiang, H.; Xu, Z.; Fang, Y.; Han, D.; Hong, W.; Wei, W.; Tu, J. CAR-macrophage: A new immunotherapy candidate against solid tumors. Biomed. Pharmacother. 2021, 139, 111605. [Google Scholar] [CrossRef] [PubMed]
  58. Wang, X.; Li, X.; Wu, Y.; Hong, J.; Zhang, M. The prognostic significance of tumor-associated neutrophils and circulating neutrophils in glioblastoma (WHO CNS5 classification). BMC Cancer 2023, 23, 20. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  59. Wang, J.; Bo, X.; Suo, T.; Liu, H.; Ni, X.; Shen, S.; Li, M.; Xu, J.; Liu, H.; Wang, Y. Tumor-infiltrating neutrophils predict prognosis and adjuvant chemotherapeutic benefit in patients with biliary cancer. Cancer Sci. 2018, 109, 2266–2274. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  60. Ohms, M.; Möller, S.; Laskay, T. An Attempt to Polarize Human Neutrophils Toward N1 and N2 Phenotypes in vitro. Front. Immunol. 2020, 11, 532. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  61. Mishalian, I.; Bayuh, R.; Levy, L.; Zolotarov, L.; Michaeli, J.; Fridlender, Z.G. Tumor-associated neutrophils (TAN) develop pro-tumorigenic properties during tumor progression. Cancer Immunol. Immunother. 2013, 62, 1745–1756. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  62. Zhang, F.; Wang, H.; Wang, X.; Jiang, G.; Liu, H.; Zhang, G.; Wang, H.; Fang, R.; Bu, X.; Cai, S.; et al. TGF-β induces M2-like macrophage polarization via SNAIL-mediated suppression of a pro-inflammatory phenotype. Oncotarget 2016, 7, 52294–52306. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  63. Shen, Y.; Wei, Y.; Wang, Z.; Jing, Y.; He, H.; Yuan, J.; Li, R.; Zhao, Q.; Wei, L.; Yang, T.; et al. TGF-β regulates hepatocellular carcinoma progression by inducing Treg cell polarization. Cell. Physiol. Biochem. 2015, 35, 1623–1632. [Google Scholar] [CrossRef] [PubMed]
  64. de Boer, S.A.; Reijrink, M.; Abdulahad, W.H.; Hoekstra, E.S.; Slart, R.H.J.A.; Heerspink, H.J.L.; Westra, J.; Mulder, D.J. Angiogenic T cells are decreased in people with type 2 diabetes mellitus and recruited by the dipeptidyl peptidase-4 inhibitor Linagliptin: A subanalysis from a randomized, placebo-controlled trial (RELEASE study). Diabetes Obes. Metab. 2020, 22, 1220–1225. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  65. Barcellos-Hoff, M.H.; Gulley, J.L. Molecular Pathways and Mechanisms of TGFβ in Cancer Therapy. Clin. Cancer Res. 2023, 29, 2025–2033. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  66. Zhao, Y.; Du, J.; Shen, X. Targeting myeloid-derived suppressor cells in tumor immunotherapy: Current, future and beyond. Front. Immunol. 2023, 14, 1157537. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  67. Ostrand-Rosenberg, S.; Lamb, T.J.; Pawelec, G. Here, There, and Everywhere: Myeloid-Derived Suppressor Cells in Immunology. J. Immunol. 2023, 210, 1183–1197. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  68. Ballbach, M.; Dannert, A.; Singh, A.; Siegmund, D.M.; Handgretinger, R.; Piali, L.; Rieber, N.; Hartl, D. Expression of checkpoint molecules on myeloid-derived suppressor cells. Immunol. Lett. 2017, 192, 1–6. [Google Scholar] [CrossRef] [PubMed]
  69. Sui, H.; Dongye, S.; Liu, X.; Xu, X.; Wang, L.; Jin, C.Q.; Yao, M.; Gong, Z.; Jiang, D.; Zhang, K.; et al. Immunotherapy of targeting MDSCs in tumor microenvironment. Front. Immunol. 2022, 13, 990463. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  70. Fletcher, M.; Ramirez, M.E.; Sierra, R.A.; Raber, P.; Thevenot, P.; Al-Khami, A.A.; Sanchez-Pino, D.; Hernandez, C.; Wyczechowska, D.D.; Ochoa, A.C.; et al. l-Arginine depletion blunts antitumor T-cell responses by inducing myeloid-derived suppressor cells. Cancer Res. 2015, 75, 275–283. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  71. Srivastava, M.K.; Sinha, P.; Clements, V.K.; Rodriguez, P.; Ostrand-Rosenberg, S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010, 70, 68–77. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  72. Bayik, D.; Lauko, A.J.; Roversi, G.A.; Serbinowski, E.; Acevedo-Moreno, L.A.; Lanigan, C.; Orujov, M.; Lo, A.; Alban, T.J.; Kim, A.; et al. Hepatobiliary malignancies have distinct peripheral myeloid-derived suppressor cell signatures and tumor myeloid cell profiles. Sci. Rep. 2020, 10, 18848. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  73. Rose, P.; van den Engel, N.K.; Kovács, J.R.; Hatz, R.A.; Boon, L.; Winter, H. Anti-Gr-1 Antibody Provides Short-Term Depletion of MDSC in Lymphodepleted Mice with Active-Specific Melanoma Therapy. Vaccines 2022, 10, 560. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  74. Petrova, V.; Groth, C.; Bitsch, R.; Arkhypov, I.; Simon, S.C.S.; Hetjens, S.; Müller, V.; Utikal, J.; Umansky, V. Immunosuppressive capacity of circulating MDSC predicts response to immune checkpoint inhibitors in melanoma patients. Front. Immunol. 2023, 14, 1065767. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  75. Mishra, H.K.; Pore, N.; Michelotti, E.F.; Walcheck, B. Anti-ADAM17 monoclonal antibody MEDI3622 increases IFNγ production by human NK cells in the presence of antibody-bound tumor cells. Cancer Immunol. Immunother. 2018, 67, 1407–1416. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  76. Fahrner, R.; Trochsler, M.; Corazza, N.; Graubardt, N.; Keogh, A.; Candinas, D.; Brunner, T.; Stroka, D.; Beldi, G. Tumor necrosis factor-related apoptosis-inducing ligand on NK cells protects from hepatic ischemia-reperfusion injury. Transplantation 2014, 97, 1102–1109. [Google Scholar] [CrossRef] [PubMed]
  77. Dianat-Moghadam, H.; Mahari, A.; Heidarifard, M.; Parnianfard, N.; Pourmousavi-Kh, L.; Rahbarghazi, R.; Amoozgar, Z. NK cells-directed therapies target circulating tumor cells and metastasis. Cancer Lett. 2021, 497, 41–53. [Google Scholar] [CrossRef] [PubMed]
  78. Vacca, P.; Pietra, G.; Tumino, N.; Munari, E.; Mingari, M.C.; Moretta, L. Exploiting Human NK Cells in Tumor Therapy. Front. Immunol. 2020, 10, 3013. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  79. Kim, N.; Kim, H.S. Targeting Checkpoint Receptors and Molecules for Therapeutic Modulation of Natural Killer Cells. Front. Immunol. 2018, 9, 2041. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  80. Qin, X.; Ning, W.; Liu, H.; Liu, X.; Luo, W.; Xia, N. Stepping forward: T-cell redirecting bispecific antibodies in cancer therapy. Acta Pharm. Sin. B 2024, 14, 2361–2377. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  81. Tsukagoshi, M.; Wada, S.; Yokobori, T.; Altan, B.; Ishii, N.; Watanabe, A.; Kubo, N.; Saito, F.; Araki, K.; Suzuki, H.; et al. Overexpression of natural killer group 2 member D ligands predicts favorable prognosis in cholangiocarcinoma. Cancer Sci. 2016, 107, 116–122. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  82. Vogler, M.; Shanmugalingam, S.; Särchen, V.; Reindl, L.M.; Grèze, V.; Buchinger, L.; Kühn, M.; Ullrich, E. Unleashing the power of NK cells in anticancer immunotherapy. J. Mol. Med. 2022, 100, 337–349. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  83. Panwong, S.; Wathikthinnakon, M.; Kaewkod, T.; Sawasdee, N.; Tragoolpua, Y.; Yenchitsomanus, P.T.; Panya, A. Cordycepin Sensitizes Cholangiocarcinoma Cells to Be Killed by Natural Killer-92 (NK-92) Cells. Molecules 2021, 26, 5973. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  84. Chiawpanit, C.; Wathikthinnakorn, M.; Sawasdee, N.; Phanthaphol, N.; Sujjitjoon, J.; Junking, M.; Yamabhai, M.; Panaampon, J.; Yenchitsomanus, P.T.; Panya, A. Precision immunotherapy for cholangiocarcinoma: Pioneering the use of human-derived anti-cMET single chain variable fragment in anti-cMET chimeric antigen receptor (CAR) NK cells. Int. Immunopharmacol. 2024, 136, 112273. [Google Scholar] [CrossRef] [PubMed]
  85. Li, B. Why do tumor-infiltrating lymphocytes have variable efficacy in the treatment of solid tumors? Front. Immunol. 2022, 13, 973881. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  86. Levi, S.T.; Copeland, A.R.; Nah, S.; Crystal, J.S.; Ivey, G.D.; Lalani, A.; Jafferji, M.; White, B.S.; Parikh, N.B.; Leko, V.; et al. Neoantigen Identification and Response to Adoptive Cell Transfer in Anti-PD-1 Naïve and Experienced Patients with Metastatic Melanoma. Clin. Cancer Res. 2022, 28, 3042–3052. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  87. Khalili-Tanha, G.; Fiuji, H.; Gharib, M.; Moghbeli, M.; Khalili-Tanha, N.; Rahmani, F.; Shakour, N.; Maftooh, M.; Hassanian, S.M.; Asgharzadeh, F.; et al. Dual targeting of TGF-β and PD-L1 inhibits tumor growth in TGF-β/PD-L1-driven colorectal carcinoma. Life Sci. 2023, 328, 121865. [Google Scholar] [CrossRef] [PubMed]
  88. Kumar, A.; Watkins, R.; Vilgelm, A.E. Cell Therapy With TILs: Training and Taming T Cells to Fight Cancer. Front. Immunol. 2021, 12, 690499. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  89. Stanton, S.E.; Disis, M.L. Clinical significance of tumor-infiltrating lymphocytes in breast cancer. J. Immunother. Cancer. 2016, 4, 59. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  90. Zhou, Y.; Tian, Q.; Wang, B.Y.; Yang, J.; Zhao, S.D.; Yang, J. The prognostic significance of TILs as a biomarker in triple-negative breast cancer: What is the role of TILs in TME of TNBC? Eur. Rev. Med. Pharmacol. Sci. 2021, 25, 2885–2897. [Google Scholar] [CrossRef] [PubMed]
  91. Moreno, V.; Salazar, R.; Gruber, S.B. The prognostic value of TILs in stage III colon cancer must consider sidedness. Ann. Oncol. 2022, 33, 1094–1096. [Google Scholar] [CrossRef] [PubMed]
  92. Liu, D.; Heij, L.R.; Czigany, Z.; Dahl, E.; Lang, S.A.; Ulmer, T.F.; Luedde, T.; Neumann, U.P.; Bednarsch, J. The role of tumor-infiltrating lymphocytes in cholangiocarcinoma. J. Exp. Clin. Cancer Res. 2022, 41, 127. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  93. Chiawpanit, C.; Panwong, S.; Sawasdee, N.; Yenchitsomanus, P.T.; Panya, A. Genistein Sensitizes Human Cholangiocarcinoma Cell Lines to Be Susceptible to Natural Killer Cells. Biology 2022, 11, 1098. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  94. Paillet, J.; Kroemer, G.; Pol, J.G. Immune contexture of cholangiocarcinoma. Curr. Opin. Gastroenterol. 2020, 36, 70–76. [Google Scholar] [CrossRef] [PubMed]
  95. Zhang, M.; Yang, H.; Wan, L.; Wang, Z.; Wang, H.; Ge, C.; Liu, Y.; Hao, Y.; Zhang, D.; Shi, G.; et al. Single-cell transcriptomic architecture and intercellular crosstalk of human intrahepatic cholangiocarcinoma. J. Hepatol. 2020, 73, 1118–1130. [Google Scholar] [CrossRef] [PubMed]
  96. Yu, W.D.; Sun, G.; Li, J.; Xu, J.; Wang, X. Mechanisms and therapeutic potentials of cancer immunotherapy in combination with radiotherapy and/or chemotherapy. Cancer Lett. 2019, 452, 66–70. [Google Scholar] [CrossRef] [PubMed]
  97. Fridman, W.H.; Zitvogel, L.; Sautès-Fridman, C.; Kroemer, G. The immune contexture in cancer prognosis and treatment. Nat. Rev. Clin. Oncol. 2017, 14, 717–734. [Google Scholar] [CrossRef] [PubMed]
  98. Sung, E.; Ko, M.; Won, J.Y.; Jo, Y.; Park, E.; Kim, H.; Choi, E.; Jung, U.J.; Jeon, J.; Kim, Y.; et al. LAG-3xPD-L1 bispecific antibody potentiates antitumor responses of T cells through dendritic cell activation. Mol. Ther. 2022, 30, 2800–2816. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  99. Diggs, L.P.; Ruf, B.; Ma, C.; Heinrich, B.; Cui, L.; Zhang, Q.; McVey, J.C.; Wabitsch, S.; Heinrich, S.; Rosato, U.; et al. CD40-mediated immune cell activation enhances response to anti-PD-1 in murine intrahepatic cholangiocarcinoma. J. Hepatol. 2021, 74, 1145–1154. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  100. Hu, Z.Q.; Zhou, Z.J.; Luo, C.B.; Xin, H.Y.; Li, J.; Yu, S.Y.; Zhou, S.L. Peritumoral plasmacytoid dendritic cells predict a poor prognosis for intrahepatic cholangiocarcinoma after curative resection. Cancer Cell Int. 2020, 20, 582. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  101. Lin, J.; Dai, Y.; Sang, C.; Song, G.; Xiang, B.; Zhang, M.; Dong, L.; Xia, X.; Ma, J.; Shen, X.; et al. Multimodule characterization of immune subgroups in intrahepatic cholangiocarcinoma reveals distinct therapeutic vulnerabilities. J. Immunother. Cancer. 2022, 10, e004892. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  102. Sun, B.Y.; Wang, Z.T.; Chen, K.Z.; Song, Y.; Wu, J.F.; Zhang, D.; Sun, G.Q.; Zhou, J.; Fan, J.; Hu, B.; et al. Mobilization and activation of tumor-infiltrating dendritic cells inhibits lymph node metastasis in intrahepatic cholangiocarcinoma. Cell Death Discov. 2024, 10, 304. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  103. Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef] [PubMed]
  104. Gardner, A.; Ruffell, B. Dendritic Cells and Cancer Immunity. Trends Immunol. 2016, 37, 855–865. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  105. Patente, T.A.; Pinho, M.P.; Oliveira, A.A.; Evangelista, G.C.M.; Bergami-Santos, P.C.; Barbuto, J.A.M. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front. Immunol. 2019, 9, 3176. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  106. Bryant, C.E.; Sutherland, S.; Kong, B.; Papadimitrious, M.S.; Fromm, P.D.; Hart, D.N.J. Dendritic cells as cancer therapeutics. Semin. Cell Dev. Biol. 2019, 86, 77–88. [Google Scholar] [CrossRef] [PubMed]
  107. Lee, Y.S.; Radford, K.J. The role of dendritic cells in cancer. Int. Rev. Cell Mol. Biol. 2019, 348, 123–178. [Google Scholar] [CrossRef] [PubMed]
  108. Santos, P.M.; Butterfield, L.H. Dendritic Cell-Based Cancer Vaccines. J. Immunol. 2018, 200, 443–449. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  109. Wang, C.; Barnoud, C.; Cenerenti, M.; Sun, M.; Caffa, I.; Kizil, B.; Bill, R.; Liu, Y.; Pick, R.; Garnier, L.; et al. Dendritic cells direct circadian anti-tumour immune responses. Nature 2023, 614, 136–143. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  110. Roberts, E.W.; Broz, M.L.; Binnewies, M.; Headley, M.B.; Nelson, A.E.; Wolf, D.M.; Kaisho, T.; Bogunovic, D.; Bhardwaj, N.; Krummel, M.F. Critical Role for CD103(+)/CD141(+) Dendritic Cells Bearing CCR7 for Tumor Antigen Trafficking and Priming of T Cell Immunity in Melanoma. Cancer Cell 2016, 30, 324–336. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  111. Rizzo, A.; Santoni, M.; Mollica, V.; Fiorentino, M.; Brandi, G.; Massari, F. Microbiota and prostate cancer. Semin. Cancer Biol. 2022, 86 Pt 3, 1058–1065. [Google Scholar] [CrossRef] [PubMed]
  112. Liu, P.; Zhao, L.; Kroemer, G.; Kepp, O. Conventional type 1 dendritic cells (cDC1) in cancer immunity. Biol. Direct. 2023, 18, 71. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  113. Junking, M.; Grainok, J.; Thepmalee, C.; Wongkham, S.; Yenchitsomanus, P.T. Enhanced cytotoxic activity of effector T-cells against cholangiocarcinoma by dendritic cells pulsed with pooled mRNA. Tumour Biol. 2017, 39, 1010428317733367. [Google Scholar] [CrossRef] [PubMed]
  114. Panya, A.; Thepmalee, C.; Sawasdee, N.; Sujjitjoon, J.; Phanthaphol, N.; Junking, M.; Wongkham, S.; Yenchitsomanus, P.T. Cytotoxic activity of effector T cells against cholangiocarcinoma is enhanced by self-differentiated monocyte-derived dendritic cells. Cancer Immunol. Immunother. 2018, 67, 1579–1588. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  115. Minini, M.; Fouassier, L. Cancer-Associated Fibroblasts and Extracellular Matrix: Therapeutical Strategies for Modulating the Cholangiocarcinoma Microenvironment. Curr. Oncol. 2023, 30, 4185–4196. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  116. Cantallops Vilà, P.; Ravichandra, A.; Agirre Lizaso, A.; Perugorria, M.J.; Affò, S. Heterogeneity, crosstalk, and targeting of cancer-associated fibroblasts in cholangiocarcinoma. Hepatology 2024, 79, 941–958. [Google Scholar] [CrossRef] [PubMed]
  117. Giguelay, A.; Turtoi, E.; Khelaf, L.; Tosato, G.; Dadi, I.; Chastel, T.; Poul, M.A.; Pratlong, M.; Nicolescu, S.; Severac, D.; et al. The landscape of cancer-associated fibroblasts in colorectal cancer liver metastases. Theranostics 2022, 12, 7624–7639. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  118. Carpino, G.; Overi, D.; Melandro, F.; Grimaldi, A.; Cardinale, V.; Di Matteo, S.; Mennini, G.; Rossi, M.; Alvaro, D.; Barnaba, V.; et al. Matrisome analysis of intrahepatic cholangiocarcinoma unveils a peculiar cancer-associated extracellular matrix structure. Clin. Proteom. 2019, 16, 37. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  119. van Tienderen, G.S.; Rosmark, O.; Lieshout, R.; Willemse, J.; de Weijer, F.; Elowsson Rendin, L.; Westergren-Thorsson, G.; Doukas, M.; Groot Koerkamp, B.; van Royen, M.E.; et al. Extracellular matrix drives tumor organoids toward desmoplastic matrix deposition and mesenchymal transition. Acta Biomater. 2023, 158, 115–131. [Google Scholar] [CrossRef] [PubMed]
  120. Ying, F.; Chan, M.S.M.; Lee, T.K.W. Cancer-Associated Fibroblasts in Hepatocellular Carcinoma and Cholangiocarcinoma. Cell. Mol. Gastroenterol. Hepatol. 2023, 15, 985–999. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  121. Yi, X.; Zhu, J.; Liu, W.; Peng, L.; Lu, C.; Sun, P.; Huang, L.; Nie, X.; Huang, S.; Guo, T.; et al. Proteome Landscapes of Human Hepatocellular Carcinoma and Intrahepatic Cholangiocarcinoma. Mol. Cell. Proteom. 2023, 22, 100604. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  122. Zhu, C.; Ma, J.; Zhu, K.; Yu, L.; Zheng, B.; Rao, D.; Zhang, S.; Dong, L.; Gao, Q.; Zhang, X.; et al. Spatial immunophenotypes predict clinical outcome in intrahepatic cholangiocarcinoma. JHEP Rep. 2023, 5, 100762. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  123. Märkl, F.; Huynh, D.; Endres, S.; Kobold, S. Utilizing chemokines in cancer immunotherapy. Trends Cancer 2022, 8, 670–682. [Google Scholar] [CrossRef] [PubMed]
  124. Propper, D.J.; Balkwill, F.R. Harnessing cytokines and chemokines for cancer therapy. Nat. Rev. Clin. Oncol. 2022, 19, 237–253. [Google Scholar] [CrossRef] [PubMed]
  125. Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  126. Korbecki, J.; Kojder, K.; Simińska, D.; Bohatyrewicz, R.; Gutowska, I.; Chlubek, D.; Baranowska-Bosiacka, I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of the Ligands of Receptors CCR1, CCR2, CCR3, and CCR4. Int. J. Mol. Sci. 2020, 21, 8412. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  127. Drouillard, D.; Craig, B.T.; Dwinell, M.B. Physiology of chemokines in the cancer microenvironment. Am. J. Physiol. Cell Physiol. 2023, 324, C167–C182. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  128. Mollica Poeta, V.; Massara, M.; Capucetti, A.; Bonecchi, R. Chemokines and Chemokine Receptors: New Targets for Cancer Immunotherapy. Front. Immunol. 2019, 10, 379. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  129. Korbecki, J.; Grochans, S.; Gutowska, I.; Barczak, K.; Baranowska-Bosiacka, I. CC Chemokines in a Tumor: A Review of Pro-Cancer and Anti-Cancer Properties of Receptors CCR5, CCR6, CCR7, CCR8, CCR9, and CCR10 Ligands. Int. J. Mol. Sci. 2020, 21, 7619. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  130. Masih, M.; Agarwal, S.; Kaur, R.; Gautam, P.K. Role of chemokines in breast cancer. Cytokine 2022, 155, 155909. [Google Scholar] [CrossRef] [PubMed]
  131. Cheng, D.; Wang, J.; Wang, Y.; Xue, Y.; Yang, Q.; Yang, Q.; Zhao, H.; Huang, J.; Peng, X. Chemokines: Function and therapeutic potential in bone metastasis of lung cancer. Cytokine 2023, 172, 156403. [Google Scholar] [CrossRef] [PubMed]
  132. Mehraj, U.; Mushtaq, U.; Mir, M.A.; Saleem, A.; Macha, M.A.; Lone, M.N.; Hamid, A.; Zargar, M.A.; Ahmad, S.M.; Wani, N.A. Chemokines in triple-negative breast cancer heterogeneity: New challenges for clinical implications. Semin. Cancer Biol. 2022, 86 Pt 2, 769–783. [Google Scholar] [CrossRef] [PubMed]
  133. Miyata, T.; Yamashita, Y.I.; Yoshizumi, T.; Shiraishi, M.; Ohta, M.; Eguchi, S.; Aishima, S.; Fujioka, H.; Baba, H. CXCL12 expression in intrahepatic cholangiocarcinoma is associated with metastasis and poor prognosis. Cancer Sci. 2019, 110, 3197–3203. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  134. Caligiuri, A.; Pastore, M.; Lori, G.; Raggi, C.; Di Maira, G.; Marra, F.; Gentilini, A. Role of Chemokines in the Biology of Cholangiocarcinoma. Cancers 2020, 12, 2215. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  135. Urabe, F.; Kosaka, N.; Ito, K.; Kimura, T.; Egawa, S.; Ochiya, T. Extracellular vesicles as biomarkers and therapeutic targets for cancer. Am. J. Physiol. Cell Physiol. 2020, 318, C29–C39. [Google Scholar] [CrossRef] [PubMed]
  136. Zhang, X.; Zhang, H.; Gu, J.; Zhang, J.; Shi, H.; Qian, H.; Wang, D.; Xu, W.; Pan, J.; Santos, H.A. Engineered Extracellular Vesicles for Cancer Therapy. Adv. Mater. 2021, 33, e2005709. [Google Scholar] [CrossRef] [PubMed]
  137. Weng, Z.; Zhang, B.; Wu, C.; Yu, F.; Han, B.; Li, B.; Li, L. Therapeutic roles of mesenchymal stem cell-derived extracellular vesicles in cancer. J. Hematol. Oncol. 2021, 14, 136. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  138. Xie, F.; Feng, S.; Yang, H.; Mao, Y. Extracellular vesicles in hepatocellular cancer and cholangiocarcinoma. Ann. Transl. Med. 2019, 7, 86. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  139. Trifylli, E.M.; Kriebardis, A.G.; Koustas, E.; Papadopoulos, N.; Vasileiadi, S.; Fortis, S.P.; Tzounakas, V.L.; Anastasiadi, A.T.; Sarantis, P.; Papageorgiou, E.G.; et al. The Arising Role of Extracellular Vesicles in Cholangiocarcinoma: A Rundown of the Current Knowledge Regarding Diagnostic and Therapeutic Approaches. Int. J. Mol. Sci. 2023, 24, 15563. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  140. Zhang, N.; Shu, L.; Liu, Z.; Shi, A.; Zhao, L.; Huang, S.; Sheng, G.; Yan, Z.; Song, Y.; Huang, F.; et al. The role of extracellular vesicles in cholangiocarcinoma tumor microenvironment. Front. Pharmacol. 2024, 14, 1336685. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  141. Wang, J.; Shi, R.; Yin, Y.; Luo, H.; Cao, Y.; Lyu, Y.; Luo, H.; Zeng, X.; Wang, D. Clinical significance of small extracellular vesicles in cholangiocarcinoma. Front. Oncol. 2024, 14, 1334592. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  142. Cao, L.; Hong, J.; Wu, J. Potential of extracellular vesicles and exosomes as diagnostic markers for cholangiocarcinoma. Hepatobiliary Surg. Nutr. 2022, 11, 436–438. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  143. Lapitz, A.; Azkargorta, M.; Milkiewicz, P.; Olaizola, P.; Zhuravleva, E.; Grimsrud, M.M.; Schramm, C.; Arbelaiz, A.; O’Rourke, C.J.; La Casta, A.; et al. Liquid biopsy-based protein biomarkers for risk prediction, early diagnosis, and prognostication of cholangiocarcinoma. J. Hepatol. 2023, 79, 93–108. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  144. Gradilone, S.A. Extracellular vesicles as therapeutic carriers of microRNAs for cholangiocarcinoma. Hepatology 2017, 65, 404–406. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  145. Zou, X.; Tang, X.Y.; Qu, Z.Y.; Sun, Z.W.; Ji, C.F.; Li, Y.J.; Guo, S.D. Targeting the PDGF/PDGFR signaling pathway for cancer therapy: A review. Int. J. Biol. Macromol. 2022, 202, 539–557. [Google Scholar] [CrossRef] [PubMed]
  146. Turrell, F.K.; Orha, R.; Guppy, N.J.; Gillespie, A.; Guelbert, M.; Starling, C.; Haider, S.; Isacke, C.M. Age-associated microenvironmental changes highlight the role of PDGF-C in ER+ breast cancer metastatic relapse. Nat. Cancer 2023, 4, 468–484. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  147. Kaps, L.; Schuppan, D. Targeting Cancer Associated Fibroblasts in Liver Fibrosis and Liver Cancer Using Nanocarriers. Cells 2020, 9, 2027. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  148. Lv, D.; Gimple, R.C.; Zhong, C.; Wu, Q.; Yang, K.; Prager, B.C.; Godugu, B.; Qiu, Z.; Zhao, L.; Zhang, G.; et al. PDGF signaling inhibits mitophagy in glioblastoma stem cells through N6-methyladenosine. Dev. Cell 2022, 57, 1466–1481.e6. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  149. Cadamuro, M.; Brivio, S.; Mertens, J.; Vismara, M.; Moncsek, A.; Milani, C.; Fingas, C.; Cristina Malerba, M.; Nardo, G.; Dall’Olmo, L.; et al. Platelet-derived growth factor-D enables liver myofibroblasts to promote tumor lymphangiogenesis in cholangiocarcinoma. J. Hepatol. 2019, 70, 700–709. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  150. Yan, J.; Xiao, G.; Yang, C.; Liu, Q.; Lv, C.; Yu, X.; Zhou, Z.; Lin, S.; Bai, Z.; Lin, H.; et al. Cancer-Associated Fibroblasts Promote Lymphatic Metastasis in Cholangiocarcinoma via the PDGF-BB/PDGFR-β Mediated Paracrine Signaling Network. Aging Dis. 2024, 15, 369–389. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  151. Shi, A.; Liu, Z.; Fan, Z.; Li, K.; Liu, X.; Tang, Y.; Hu, J.; Li, X.; Shu, L.; Zhao, L.; et al. Function of mast cell and bile-cholangiocarcinoma interplay in cholangiocarcinoma microenvironment. Gut 2024, 73, 1350–1363. [Google Scholar] [CrossRef] [PubMed]
  152. Waller, C.F. Imatinib Mesylate. Recent Results Cancer Res. 2018, 212, 1–27. [Google Scholar] [CrossRef] [PubMed]
  153. Yao, F.; Luo, Y.; Liu, Y.C.; Chen, Y.H.; Li, Y.T.; Hu, X.Y.; You, X.Y.; Yu, S.S.; Li, Z.Y.; Chen, L.; et al. Imatinib inhibits pericyte-fibroblast transition and inflammation and promotes axon regeneration by blocking the PDGF-BB/PDGFRβ pathway in spinal cord injury. Inflamm. Regen. 2022, 42, 44. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  154. Fan, B.; Malato, Y.; Calvisi, D.F.; Naqvi, S.; Razumilava, N.; Ribback, S.; Gores, G.J.; Dombrowski, F.; Evert, M.; Chen, X.; et al. Cholangiocarcinomas can originate from hepatocytes in mice. J. Clin. Invest. 2012, 122, 2911–2915. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  155. Meurette, O.; Mehlen, P. Notch Signaling in the Tumor Microenvironment. Cancer Cell 2018, 34, 536–548. [Google Scholar] [CrossRef] [PubMed]
  156. Mancarella, S.; Serino, G.; Gigante, I.; Cigliano, A.; Ribback, S.; Sanese, P.; Grossi, V.; Simone, C.; Armentano, R.; Evert, M.; et al. CD90 is regulated by notch1 and hallmarks a more aggressive intrahepatic cholangiocarcinoma phenotype. J. Exp. Clin. Cancer Res. 2022, 41, 65, Erratum in J. Exp. Clin. Cancer Res. 2023, 42, 88. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  157. Mancarella, S.; Gigante, I.; Serino, G.; Pizzuto, E.; Dituri, F.; Valentini, M.F.; Wang, J.; Chen, X.; Armentano, R.; Calvisi, D.F.; et al. Crenigacestat blocking notch pathway reduces liver fibrosis in the surrounding ecosystem of intrahepatic CCA viaTGF-β inhibition. J. Exp. Clin. Cancer Res. 2022, 41, 331. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  158. Yang, R.; Wang, D.; Han, S.; Gu, Y.; Li, Z.; Deng, L.; Yin, A.; Gao, Y.; Li, X.; Yu, Y.; et al. MiR-206 suppresses the deterioration of intrahepatic cholangiocarcinoma and promotes sensitivity to chemotherapy by inhibiting interactions with stromal CAFs. Int. J. Biol. Sci. 2022, 18, 43–64, Erratum in Int. J. Biol. Sci. 2022, 18, 4466–4467. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  159. Lin, Y.; Peng, L.; Dong, L.; Liu, D.; Ma, J.; Lin, J.; Chen, X.; Lin, P.; Song, G.; Zhang, M.; et al. Geospatial Immune Heterogeneity Reflects the Diverse Tumor-Immune Interactions in Intrahepatic Cholangiocarcinoma. Cancer Discov. 2022, 12, 2350–2371. [Google Scholar] [CrossRef] [PubMed]
  160. Tomlinson, J.L.; Valle, J.W.; Ilyas, S.I. Immunobiology of cholangiocarcinoma. J. Hepatol. 2023, 79, 867–875. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  161. Zhong, X.; Ji, C.; Ren, D.; Ke, A.; Yang, Z. Circular RNA circEIF3C promotes intrahepatic cholangiocarcinoma progression and immune evasion via the miR-34a-5p/B7-H4 axis. Genes Dis. 2022, 10, 370–372. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  162. Mauro, E.; Forner, A. Immunotherapy in biliary tract cancer: The race has begun! Liver Int. 2023, 43, 1620–1622. [Google Scholar] [CrossRef] [PubMed]
  163. Nakamura, H.; Arai, Y.; Totoki, Y.; Shirota, T.; Elzawahry, A.; Kato, M.; Hama, N.; Hosoda, F.; Urushidate, T.; Ohashi, S.; et al. Genomic spectra of biliary tract cancer. Nat. Genet. 2015, 47, 1003–1010. [Google Scholar] [CrossRef] [PubMed]
  164. Gani, F.; Nagarajan, N.; Kim, Y.; Zhu, Q.; Luan, L.; Bhaijjee, F.; Anders, R.A.; Pawlik, T.M. Program Death 1 Immune Checkpoint and Tumor Microenvironment: Implications for Patients With Intrahepatic Cholangiocarcinoma. Ann. Surg. Oncol. 2016, 23, 2610–2617. [Google Scholar] [CrossRef] [PubMed]
  165. Qiu, X.; Yang, S.; Wang, S.; Wu, J.; Zheng, B.; Wang, K.; Shen, S.; Jeong, S.; Li, Z.; Zhu, Y.; et al. M6A Demethylase ALKBH5 Regulates PD-L1 Expression and Tumor Immunoenvironment in Intrahepatic Cholangiocarcinoma. Cancer Res. 2021, 81, 4778–4793. [Google Scholar] [CrossRef] [PubMed]
  166. Cho, S.Y.; Hwang, H.; Kim, Y.H.; Yoo, B.C.; Han, N.; Kong, S.Y.; Baek, M.J.; Kim, K.H.; Lee, M.R.; Park, J.G.; et al. Refining Classification of Cholangiocarcinoma Subtypes via Proteogenomic Integration Reveals New Therapeutic Prospects. Gastroenterology 2023, 164, 1293–1309, Erratum in Gastroenterology 2023, 165, 1313. [Google Scholar] [CrossRef] [PubMed]
  167. Wardell, C.P.; Fujita, M.; Yamada, T.; Simbolo, M.; Fassan, M.; Karlic, R.; Polak, P.; Kim, J.; Hatanaka, Y.; Maejima, K.; et al. Genomic characterization of biliary tract cancers identifies driver genes and predisposing mutations. J. Hepatol. 2018, 68, 959–969. [Google Scholar] [CrossRef] [PubMed]
  168. Qiang, S.; Fu, F.; Wang, J.; Dong, C. Definition of immune molecular subtypes with distinct immune microenvironment, recurrence, and PANoptosis features to aid clinical therapeutic decision-making. Front. Genet. 2022, 13, 1007108. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  169. Dong, L.; Lu, D.; Chen, R.; Lin, Y.; Zhu, H.; Zhang, Z.; Cai, S.; Cui, P.; Song, G.; Rao, D.; et al. Proteogenomic characterization identifies clinically relevant subgroups of intrahepatic cholangiocarcinoma. Cancer Cell 2022, 40, 70–87.e15. [Google Scholar] [CrossRef] [PubMed]
  170. Liao, H.; Chen, X.; Wang, H.; Lin, Y.; Chen, L.; Yuan, K.; Liao, M.; Jiang, H.; Peng, J.; Wu, Z.; et al. Whole-Genome DNA Methylation Profiling of Intrahepatic Cholangiocarcinoma Reveals Prognostic Subtypes with Distinct Biological Drivers. Cancer Res. 2024, 84, 1747–1763. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  171. Zhang, Y.; Ma, Z.; Li, C.; Wang, C.; Jiang, W.; Chang, J.; Han, S.; Lu, Z.; Shao, Z.; Wang, Y.; et al. The genomic landscape of cholangiocarcinoma reveals the disruption of post-transcriptional modifiers. Nat. Commun. 2022, 13, 3061. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  172. Mahipal, A.; Tella, S.H.; Kommalapati, A.; Anaya, D.; Kim, R. FGFR2 genomic aberrations: Achilles heel in the management of advanced cholangiocarcinoma. Cancer Treat. Rev. 2019, 78, 1–7. [Google Scholar] [CrossRef] [PubMed]
  173. Deng, M.; Ran, P.; Chen, L.; Wang, Y.; Yu, Z.; Cai, K.; Feng, J.; Qin, Z.; Yin, Y.; Tan, S.; et al. Proteogenomic characterization of cholangiocarcinoma. Hepatology 2023, 77, 411–429. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  174. European Association for the Study of the Liver. EASL-ILCA Clinical Practice Guidelines on the management of intrahepatic cholangiocarcinoma. J. Hepatol. 2023, 79, 181–208, Erratum in J. Hepatol. 2023, 79, 1342. [CrossRef] [PubMed]
  175. Kitagawa, A.; Osawa, T.; Noda, M.; Kobayashi, Y.; Aki, S.; Nakano, Y.; Saito, T.; Shimizu, D.; Komatsu, H.; Sugaya, M.; et al. Convergent genomic diversity and novel BCAA metabolism in intrahepatic cholangiocarcinoma. Br. J. Cancer. 2023, 128, 2206–2217. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  176. Zhang, J.; Huang, D.; Saw, P.E.; Song, E. Turning cold tumors hot: From molecular mechanisms to clinical applications. Trends Immunol. 2022, 43, 523–545. [Google Scholar] [CrossRef] [PubMed]
  177. Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef] [PubMed]
  178. Liu, Y.T.; Sun, Z.J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365–5386. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  179. Greten, T.F.; Schwabe, R.; Bardeesy, N.; Ma, L.; Goyal, L.; Kelley, R.K.; Wang, X.W. Immunology and immunotherapy of cholangiocarcinoma. Nat. Rev. Gastroenterol. Hepatol. 2023, 20, 349–365. [Google Scholar] [CrossRef] [PubMed]
  180. Ricci, A.D.; Rizzo, A.; Brandi, G. The DNA damage repair (DDR) pathway in biliary tract cancer (BTC): A new Pandora’s box? ESMO Open 2020, 5, e001042. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  181. Al-Rajabi, R.; Sun, W. Immunotherapy in cholangiocarcinoma. Curr. Opin. Gastroenterol. 2021, 37, 105–111. [Google Scholar] [CrossRef] [PubMed]
  182. Shang, T.; Jiang, T.; Lu, T.; Wang, H.; Cui, X.; Pan, Y.; Xu, M.; Pei, M.; Ding, Z.; Feng, X.; et al. Tertiary lymphoid structures predict the prognosis and immunotherapy response of cholangiocarcinoma. Front. Immunol. 2023, 14, 1166497. [Google Scholar] [CrossRef] [PubMed] [PubMed Central]
  183. Rizzo, A.; Ricci, A.D.; Brandi, G. Recent advances of immunotherapy for biliary tract cancer. Expert Rev. Gastroenterol. Hepatol. 2021, 15, 527–536. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Overview of cancer-associated changes in tumor immune microenvironment.
Figure 1. Overview of cancer-associated changes in tumor immune microenvironment.
Cancers 16 03542 g001
Figure 2. Overview of some mechanisms of immune evasion in cancer.
Figure 2. Overview of some mechanisms of immune evasion in cancer.
Cancers 16 03542 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Ricci, A.D.; Rizzo, A.; Schirizzi, A.; D’Alessandro, R.; Frega, G.; Brandi, G.; Shahini, E.; Cozzolongo, R.; Lotesoriere, C.; Giannelli, G. Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications. Cancers 2024, 16, 3542. https://doi.org/10.3390/cancers16203542

AMA Style

Ricci AD, Rizzo A, Schirizzi A, D’Alessandro R, Frega G, Brandi G, Shahini E, Cozzolongo R, Lotesoriere C, Giannelli G. Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications. Cancers. 2024; 16(20):3542. https://doi.org/10.3390/cancers16203542

Chicago/Turabian Style

Ricci, Angela Dalia, Alessandro Rizzo, Annalisa Schirizzi, Rosalba D’Alessandro, Giorgio Frega, Giovanni Brandi, Endrit Shahini, Raffaele Cozzolongo, Claudio Lotesoriere, and Gianluigi Giannelli. 2024. "Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications" Cancers 16, no. 20: 3542. https://doi.org/10.3390/cancers16203542

APA Style

Ricci, A. D., Rizzo, A., Schirizzi, A., D’Alessandro, R., Frega, G., Brandi, G., Shahini, E., Cozzolongo, R., Lotesoriere, C., & Giannelli, G. (2024). Tumor Immune Microenvironment in Intrahepatic Cholangiocarcinoma: Regulatory Mechanisms, Functions, and Therapeutic Implications. Cancers, 16(20), 3542. https://doi.org/10.3390/cancers16203542

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop