Download PDF
Review  |  Open Access  |  27 Jul 2023

Tumor microenvironment highlighting tumor-associated macrophages and immune cells

Views: 362 |  Downloads: 105 |  Cited:   1
Hepatoma Res 2023;9:32.
10.20517/2394-5079.2023.32 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Cholangiocarcinoma (CCA) grows within a highly desmoplastic microenvironment, exhibiting a continuous interconnection with the immune infiltrate, which is characterized by an abundance of immune cells, including natural killer cells, T lymphocytes, and macrophages. The presence of inflammatory cells within the tumor microenvironment plays a crucial role in determining the aggressiveness and growth of CCA. The immune cell population engages in diverse and dynamic interactions with cancer cells. The balance of different subpopulations within CCA can generate varying responses, either inhibiting or promoting tumoral progression. The purpose of this review is to offer a comprehensive overview of the role of various immune infiltrate subpopulations within the tumor microenvironment, with a particular focus on the actions of tumor-associated macrophages (TAMs) and their critical regulation in the development and progression of CCA. TAMs play vital roles in maintaining homeostasis, facilitating tissue repair, and contributing to immune responses due to their significant functional diversity. Macrophages are present in numerous types of cancer, and their emerging role has also been observed in CCA. Recognizing and attaining a deeper comprehension of the intricate interplay between infiltrating immune cells and CCA cells is essential to identify new opportunities to advance treatment strategies.

Keywords

Cholangiocarcinoma (CCA), immune infiltrate, tumor-associated macrophages (TAMs), macrophage, macrophage polarization, tumor microenvironment

INTRODUCTION

CCA represents the second most prevalent form of primary liver cancer, comprising approximately 10%-15% of all cases of primary liver malignancies. It arises from different sections of the biliary tree and expresses epithelial markers of cholangiocyte differentiation[1,2].

Worldwide, CCA affects more men than women, and there is a substantial increase in the mortality rate with age[3,4].

The incidence and mortality rates of CCA exhibit significant geographical variation worldwide. The incidence ranges from 85 per 100,000 in Eastern countries (such as northeastern Thailand) to 0.4 per 100,000 in Northern countries (like Canada). Similarly, Eastern countries exhibit the highest mortality rates for cholangiocarcinoma, with a rate of 2.5 per 100,000, whereas the lowest mortality rates, ranging from 0.2 to 0.5 per 100,000, are observed in South America[3,5,6].

Geographical variations in incidence and mortality likely reflect a multifactorial etiology based on regional risk factors and genetic predispositions[6].

Classification and histology of cholangiocarcinoma

The classification of CCA is based on the anatomical site of the biliary ducts and distinguishes among intrahepatic (iCCA), perihilar (pCCA), and distal (dCCA) subtypes[7,8].

Intrahepatic CCA

Intrahepatic CCA (iCCA) represents approximately 10% of all Cholangiocarcinoma cases and is associated with a rising global incidence and poor outcomes. It develops within the liver parenchyma, originating from the secondary bile ducts. Following hepatocellular carcinoma, cholangiocarcinoma is the second most common primary intrahepatic liver cancer[9,10].

According to the most recent tumor classification by the World Health Organization (WHO)[11], iCCA can be classified into two distinct types: perihilar large duct type (iCCAphl) and peripheral small duct type (iCCApps).This new classification was introduced following the recognition of distinct tumor cell types and anatomical locations associated with iCCA.

ICCApps arises from intrahepatic bile ductules or Canals of Hering. It has a mass-forming growth pattern within the liver tissue and displays histological features characteristic of a tubular or acinar adenocarcinoma.

Additionally, iCCApps is associated with poor or absent mucin production[12,13]. On the contrary, iCCAphl originates from large intrahepatic bile ducts or their peribiliary glands and is characterized by extended mucin production. Histologically, iCCAphl demonstrates a growth pattern with large tubular or papillary architecture[12,14]

ICCApps typically exhibits larger tumor size, less frequent lymph node metastasis, lower expression of Ki67, CA19-9, and CEA, and a better prognosis compared to iCCAphl[13,15]. ICCA can be further macroscopically divided into different subtypes, including mass-forming (60%-80%), periductal-infiltrating (15%-35%), intraductal (8%-29%), and undefined and mixed subtypes[14]. This classification is based on distinct growth patterns observed in CCA. These classes differ in terms of prognosis and their association with specific CCA types.

The most common growth pattern observed in iCCA is the mass-forming type. On the other hand, intraductal iCCA is a papillary tumor with a slow growth rate, which is associated with a more favorable prognosis. Conversely, periductal infiltrating CCA exhibits a growth pattern along the bile duct without forming a distinct mass. Instead, it manifests as diffuse bile duct thickening or small lesions. This rare condition is often found concurrently with mass-forming CCA[16,17].

Perihilar CCA and Distal CCA

Perihilar CCA (pCCA) arises in the common bile duct, specifically below the secondary biliary branches and above the cystic duct. It represents the most prevalent subtype of cholangiocarcinoma, accounting for an incidence of 50%-60%.

Distal CCA (dCCA) is localized to the region between the origin of the cystic duct and Vater's ampulla. It represents approximately 20%-30% of all cholangiocarcinoma cases[18].

Both pCCA and dCCA often present with similar characteristics, appearing as flat or poorly defined nodular sclerosing tumors or, less commonly, as intraductal papillary masses. Histologically, the majority of both pCCA and dCCA cases are conventional mucin-producing adenocarcinomas or papillary tumors[19].

PCCA and dCCA are often regarded as single entity due to their similarities and are collectively referred to as extrahepatic cholangiocarcinoma (eCCA).

PCCA, dCCA, and large bile duct iCCA share a common origin from columnar mucous cholangiocytes, as well as some typical mutations, including frequent mutations of KRAS and/or P53[12,14,20,21].

Moreover, they share a typical presentation characterized by periductal infiltration or an intraductal growth pattern[18,19].

On the other hand, iCCA pps is characterized by a mass-forming pattern and the presence of fibroblast growth receptor 2 (FGFR2) fusions and isocitrate dehydrogenase (IDH 1-2)[22,23].

Risk factors

Currently, cholangiocarcinoma is predominantly observed to have a sporadic origin, however, Multiple risk factors have been associated with the development of CCA[18].

Some risk factors for CCA, such as autoimmune disease (type I diabetes), inflammatory bowel disease (IBD), and peptic ulcers can be considered common to all the subtypes, while others specifically influence certain CCA subtypes or are region-specific[6,24].

Likewise, alcohol consumption, non-alcoholic fatty liver diseases (NAFLD), lupus, obesity, alcohol-related disorders, HBV and HCV infections, and hemochromatosis were more strongly associated with iCCA. On the other hand, cholangitis, chronic pancreatitis, choledochal cysts, cholelithiasis, choledocholithiasis, and tobacco smoking tended to be more strongly related to eCCA[24-26].

Caroli disease (CD) demonstrates the most significant associations with CCA compared to any other medical condition, resulting in a 38-fold higher risk of iCCA and a 97-fold higher risk of eCCA[24].

Caroli disease is an extremely rare liver disease, with an incidence of 1 in 1,000,000. It is characterized by non-obstructive cystic dilatations of the embryonic bile ducts, leading to an anomalous expansion[27].

Furthermore, Primary sclerosing cholangitis (PSC) is a rare condition strongly related to CCA, particularly eCCA in northern Europe and the United States. Approximately 10% of all the patients with PSC develop CCA, resulting in an overall average risk of cholangiocarcinoma that is approximately 400 times higher than the non-affected population. Cholangiocarcinoma affects approximately 10% of all patients with PSC[28].

The distinct relationship between CCA subtypes and specific risk factors has provided insight into regional incidence trends.

Liver flukes, such as Opisthorchis viverrini and Clonorchis sinensis, are prevalent in eastern countries, influencing the rates of CCA in those regions. However, in Western countries, liver fluke infection is an unlikely factor. Infections caused by liver flukes induce biliary epithelial hyperplasia, potentially facilitating changes in cholangiocarcinogenesis and exerting a promoting effect[29].

Underlining the importance of regional risk factors, different studies have demonstrated that the rates of iCCA in different countries correspond to the trends in alcohol consumption. For example, in countries such as Italy and France, where a decrease in alcohol-associated liver disease has been observed, there has also been a reduction in the incidence of iCCA. Conversely, countries that have experienced an increase in HCV infections, obesity, or NAFLD, have a higher incidence of iCCA[24,30,31] [Figure 1].

Tumor microenvironment highlighting tumor-associated macrophages and immune cells

Figure 1. The image represents the anatomical classification of CCA into iCCA and eCCA, further divided into pCCA and dCCA subtypes[7]. Additionally, the image illustrates the different gross patterns observed in intrahepatic CCA, including mass-forming, intraductal, and periductal patterns[14]. The surrounding circle highlights the main risk factors associated with cholangiocarcinoma, which are common to all CCA types but may have a more typical cause for each subtype. These risk factors are categorized into three groups: light blue indicates the primary risk factors for extrahepatic CCA, light green signifies the main risk factors for intrahepatic CCA, and yellow represents the shared risk factors between the two types of CCA[6,29]. IBD: inflammatory bowel disease; PSC: primary sclerosing cholangitis; NAFLD: non-alcoholic fatty liver diseases; eCCA: extrahepatic cholangiocarcinoma; iCCA: intrahepatic cholangiocarcinoma; pCCA: perihilar cholangiocarcinoma; dCCA: distal cholangiocarcinoma.

Genetics alterations in cholangiocarcinoma

Cholangiocarcinoma (CCA) exhibits considerable genetic heterogeneity attributed to chromosomal aberrations, as well as genetic and epigenetic mutations.

The mutations are likely to arise as a result of chronic inflammation, which directly damages the DNA molecules and impairs DNA repair mechanisms. These DNA lesions give rise to molecular abnormalities, leading to the accumulation of additional mutations and the subsequent development of the tumor[32].

Despite the diverse phenotypic presentations and activated molecular pathways in CCA, several common chromosomal alterations have been identified. These include losses at 1p, 4q, 8p, 9p, 17p, and 18p, as well as gains at 1q, 5p, 7p, 8q, 17q, 19p, and 20q[33].

Some of these common chromosomal alterations involved important genes, such as ERBB2 and MAP2K2/MEK, underlying the significance of constitutive activation of the growth factors pathway in cancer development[34,35].

The major oncogenic networks that are deregulated in CCA, involve key molecules including transforming growth factor (TGF-β), mitogen-activated protein kinase-1/2 (MAPK 1/2), and in genes enriched in key networks controlled by VEGF/ERRB, CTNNB1/MYC, and tumor necrosis factor (TNF)[36].

Genetic studies have identified a direct relationship between gene expression and patient prognosis. Mutations in KRAS, TP53, and ARID2 were found to be significantly associated with poorer patient outcomes[37].

Furthermore, distinct distributions of mutations can be observed among the different subtypes of CCA. This highlights the importance of categorizing CCA based on subtype to understand the specific mutation profiles and their implications.

Genetic alterations such as FGFR2 (fibroblast growth factor receptor 2) fusion genes, hotspot IDH mutations in IDH1 and IDH2, BAP1, and RNF43 genes are much more present in iCCA[38].

Additionally, the JAK-STAT signaling pathway, which is important for the activation of immunosuppressive effects, is specifically expressed in iCCA due to a mutation of STAT3[39].

On the other hand, mutations in ARID1B and ELF3 are more frequently observed in eCCA compared to iCCA. Additionally, specific gene fusions such as ATP1B-PRKACA and ATP1B-PRKACB have been detected exclusively in eCCA cases.

Furthermore, when considering dCCA and pCCA subtypes, a significant differentiation is observed primarily in the expression of TP53, which is more frequently mutated in distal cases[37].

It is important to emphasize how these mutations can result in alterations not only within the tumor cells but also in the surrounding tumor microenvironment. Mutations in genes such as FGFR2, and the JAK-STAT pathway can disrupt the immune infiltrate and influence the tumor microenvironment (TME). The differential expression of mutations in various subtypes of cholangiocarcinoma has the potential to play a pivotal role in elucidating the distinct characteristics of the tumor microenvironment associated with each subtype.

IMMUNE INFILTRATE

The progression of the tumor is maintained by the interplay between inflammatory cells and tumor cells, where inflammatory cells play a crucial role at different stages of tumor growth, including initiation, promotion, malignant transformation, invasion, and metastasis[40]. Within the tissue, there exists a complex and ever-changing network of cytokines, chemokines, growth factors, inflammatory mediators, and enzymes involved in modifying the extracellular matrix. This network facilitates intercellular communication amidst notable alterations in the physical and chemical properties of the tissue. This leads to the reprogramming of the surrounding cells, enabling them to assume a vital role in the survival and advancement of the tumor[41].

In TME, various types of cellular components are present: cancer-associated fibroblasts (CAFs), vascular endothelial cells, lymphatic endothelial cells, and immune cells. The immune cell composition includes various types such as tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAM), tumor-associated neutrophils (TAN), dendritic cells (DC), natural killer cells (NK), and myeloid-derived suppressor cells (MDSC), has been observed[2].

CCA grows in a pro-inflammatory microenvironment in which CAFs, ECM, and the immune cells, play a pivotal role in keeping these conditions that promote tumor progression and diffusion[42].

CAFs play a central role in shaping the TME in CCA by promoting increased production of extracellular matrix proteins and collagen. This leads to the development of a desmoplastic TME and a hypovascularized stroma, characterized by the formation of adhesions and fibrous connective tissue within the tumor[43,44].

The desmoplastic and hypo vascularized microenvironment poses a barrier to the infiltration of immune cells, leading to a higher concentration of immune cells in the peritumor area surrounding the tumor rather than within the tumor itself.

However, as evidenced in multiple studies outlined below, immune cells capable of infiltrating the tumor site play a pivotal role in the tumor's development and progression. Modulating the immune response in CCA is a primary target for future therapeutic interventions, aiming to activate the immune response and reduce genetic instability and tumor development.

Tumor-infiltrating lymphocytes

Of the various inflammatory cells that infiltrate the tumor, tumor-infiltrating lymphocytes (TILs) are recognized as the primary contributors to the host immune response against tumor cells[45].

TILS is composed of two main populations: T cells and B cells[46].

T cells can be classified based on their T cell receptor (TCR) subunits, which are responsible for recognizing membrane-bound peptides presented on the Major Histocompatibility Complex (MHC). The two major populations of T cells are distinguished by the expression of cell surface molecules CD4 and CD8, respectively.

CD8+ T cells are capable of recognizing peptides presented by MHC class I molecules, whereas CD4+ T cells rely on MHC class II molecules for peptide recognition[47].

Both CD4+ and CD8+ T cells are present in the tumor microenvironment of cholangiocarcinoma, but their distribution differs. Studies revealed that CD4+ cells are predominantly located in the interface area surrounding the tumor, while CD8+ cells infiltrate the tumor tissue[48].

CD4+ is an antigenic peptide expressed in approximately 60% of mature T cells. CD4+ cells differentiate into T helper lymphocytes, which can activate and stimulate B cells, CD8+ cells, and TAMs. T-helper lymphocytes involvement in the immune response against cancer has been associated with better overall survival in patients[49].

Moreover, the immunosuppressive effects are mediated by a subgroup of T cells expressing CD4+CD25+Foxp3+, known as regulatory T cells (Tregs). Tregs represent approximately 5%-10% of the total number of CD4+.

Tregs suppress T cells’ activity by inhibiting the costimulatory molecule CD28. This is achieved through the overexpression of Foxp3+, a transcription factor associated with the up-regulation of CTLA-4 on the cell surface[50].

Single-cell RNA-sequencing analysis of T cells comparing the tumoral and peritumoral areas of iCCA has shown an altered network of transcription factors. This altered network leads to increased infiltration of hyperactivated CD4+ Tregs and a reduction in the effector functions ofCD8+ T cells. This dysregulated immune response is likely the cause of the negative impact of Tregs in CCA[51].

Tregs have been implicated in cancer progression and a TME that is rich in Treg cells and has low levels of CD8+ T cells is associated with poor overall survival. This suggests that the presence of Tregs in the TME contributes to tumor progression and is indicative of a worse prognosis in CCA[52].

On the other hand, cytotoxic CD8+ T lymphocytes (CTL) can selectively detect and induce tumor cell death through the release of cytotoxic granules.

Among the antigens expressed by tumor cells, tumor-specific neoantigens play a significant role. Upon encountering antigens in an acute inflammatory environment, naïve antigen-specific CD8+ T cells undergo clonal expansion and differentiate into cytolytic effector T cells. Despite the presence of cytolytic effector T cells in a neoantigen-enriched tumor microenvironment, cancer can develop evasive strategies that enable its progression[53,54].

Based on immunohistochemical staining, CD8+ T cells are found to be the most abundant tumor-infiltrating lymphocytes (TILs) within the tumor regions.

In contrast to regulatory T cells (Tregs), CD8+ cytotoxic T lymphocytes exhibit anti-cancer activities, and a higher density of CTLs is associated with longer overall survival or disease-free survival[49].

Lymphocyte B cells play a significant role in inhibiting tumor development through various mechanisms. They are involved in the production of tumor-reactive antibodies, priming CD4+ and CD8+ T cells, and releasing cytokines that influence the activities of other immune cells such as NK cells, DCs, and TAMs[55,56]. Although B cells are less represented among TILs, a high infiltration of B cells in CCA has been associated with better overall survival[57].

Natural killer cells

NK cells, a type of innate lymphoid cells, possess the capacity to identify and eradicate tumor cells without requiring prior antigenic exposure. In humans, NK cells are characterized as CD3-CD56+ lymphocytes.

NK cell recognition is mediated by multiple receptor families, such as CD94-NKG2 and KIRs, which specifically bind to MHC class I molecules. These receptors exhibit either inhibitory or activating actions. Inhibitory receptors function to inhibit the killing of healthy cells, whereas activating receptors interact with ligands expressed on rapidly proliferating cells, infected cells, transformed cells, and tumor cells.

NK cell binding between activating receptors and their ligands leads to the formation of an immunological synapse, triggering the release of granules containing perforin and granzymes. Additionally, NK cell activation results in the expression of death-inducing ligands like FAS ligand and TNF-related apoptosis-inducing ligand (TRAIL), which initiate the apoptosis cascade[58].

The liver, due to its exposure to a large number of antigens, is rich in NK cells, accounting for nearly 50% of all liver lymphocytes[59,60].

Currently, only a limited number of studies have been conducted to fully characterize the role of NK cells in CCA. However, the current study has demonstrated the cytolytic activity of NK cells against CCA cells through in vitro and in vivo experiments[61,62].

Despite the demonstrated cytotoxic activity of NK cells, cancer cells have been reported to employ evasive strategies. Carnevale et al. have shown that iCCA cells can induce apoptosis in NK cells and T cells (CD4+ and CD8+) through the Fas/FasL pathway[63].

Tumor-associated neutrophils

Neutrophils are the most abundant leukocyte in human blood, constituting approximately 50%-70% of the total leukocyte population. Neutrophils are the initial responders to cellular damage, and their infiltration contributes to persistent inflammation.

Under various cytokine stimulations, neutrophils can differentiate into two distinct phenotypes: antitumoral (N1) or protumor (N2). N1 neutrophils exert their antitumoral activity through direct cytotoxicity, antibody-dependent recognition, and immune cell activation via cytokine release. Conversely, N2 neutrophils activation leads to a persistent release of enzymes, resulting in inflammatory angiogenesis and the production of reactive oxygen species. These conditions collectively facilitate cancer growth[64-66].

Another crucial aspect of neutrophil action is their interaction with and production of chemokines. Zhou et al. have demonstrated that CXCL5, a member of the CXC-type chemokine family, is overexpressed in cholangiocarcinoma cells. This chemokine plays a role in promoting the growth and metastasis of iCCA by recruiting infiltrating neutrophils within the tumor microenvironment[67].

Numerous studies have extensively highlighted the significance of neutrophil infiltration, with the Neutrophil-to-Lymphocyte Ratio (NLR) emerging as a valuable prognostic marker for long-term outcomes. Independent associations have been observed between NLR and worse Overall Survival and Disease-Free Survival following curative resection of both pCCA and iCCA. A high NLR is thought to reflect an aberrant inflammatory response with a pro-tumor activity, contributing to an unfavorable prognosis[68,69].

Dendritic cells

Within healthy liver tissue, Dendritic cells DCs are typically found in limited quantities, yet they retain their specialized role as antigen-presenting cells (APCs) capable of initiating adaptive immune responses. Like other APCs, when activated, DCs capture antigens and migrate to the lymph nodes. There, they present the antigens on their cell membrane, leading to the activation of specific B and T cells. Additionally, DCs play a crucial role in facilitating the selection of T cell receptors (TCRs) and B cell receptors (BCRs)[70].

DCs can be categorized into two primary subgroups: conventional DCs (cDCs) and plasmacytoid DCs (pDCs). Among cDCs, there are two distinct subclusters: cDC1s, which play a role in sustaining T cell restimulation within the TME, and cDC2s, which are responsible for presenting antigens to naive CD4+ T cells and inducing their differentiation[70,71].

On the other hand, the activity of pDCs remains unclear, but they are believed to have primarily tolerogenic functions. This is partially supported by evidence of poor prognosis in various types of cancer[72].

Currently, limited research has been conducted to thoroughly characterize DCs in CCA. However, recent studies have revealed intriguing findings regarding their functionality. Observations have indicated that blocking IL-10 and TGF-β receptors on DCs can result in elevated IFN-γ levels and heightened cytolytic activity of effector T cells in CCA. This suggests a potential role for DCs in promoting an immune response against CCA.

Furthermore, DCs have been found to stimulate the proliferation of NK cells through a combination of cytokine signaling and direct cell-to-cell contact. This highlights the significance of DCs in facilitating NK cell activation and their potential impact on the immune response in CCA[73,74].

Myeloid-derived suppressor cells

Myeloid-derived suppressor cells (MDSCs) are a heterogenous population of cells that undergo expansion in inflammatory conditions, including the TME. MDSCs can be classified into two main groups based on their origins: granulocytic or polymorphonuclear MDSCs (PMN-MDSCs) and monocytic MDSCs (M-MDSCs). The primary characteristic of MDSCs is their capacity to suppress immune responses, thereby directly impacting crucial immune cells[75].

The two populations, PMN-MDSCs, and M-MDSCs employ different pathways of action. PMN-MDSCs release reactive oxygen species (ROS), peroxynitrite, arginase 1, and prostaglandin E2 (PGE2) into the environment to mediate immune suppression. On the other hand, M-MDSCs secrete nitric oxide (NO), and immunosuppressive cytokines such as IL-10 and TGFβ, and upregulate the expression of programmed death-ligand 1 (PDL1)[76].

An increase in circulating MDSCs has been reported in patients with cholangiocarcinoma[77].

Furthermore, the potential clinical importance of MDSCs is underlined by Ma et al. in their study, which demonstrated, using an in vivo model of cholangiocarcinoma, that concurrent depletion of PMN-MDSCs and macrophages sensitized the tumor to anti-PD-L1 therapy[75] [Figure 2].

Tumor microenvironment highlighting tumor-associated macrophages and immune cells

Figure 2. Representation of the immune infiltrate interaction with CCA and its microenvironment. TANs: tumor-associated neutrophils; NK: natural killer; DCs: dendritic cells; MDSCs: myeloid-derived suppressor cells; CCA: cholangiocarcinoma.

Lymphocyte T CD8+, T CD4+, and CCA have a mutual inhibiting action, high-density CD4+ and CD8+ determine a longer OS and CCA cells can induce apoptosis in NK cells and T-cells (CD4+, CD8+), via the Fas/FasL pathway[49,63]. Lymphocyte Treg overexpression of CTLA 4 promotes CCA proliferation through the inhibition of lymphocyte T; An environment rich in Treg has poor overall survival[50,52]. High Lymphocyte B infiltration in CCA was related to better overall survival[57]. NLR and neutrophil infiltration combined with intratumor IL-17 cells are related to worse overall survival, demonstrating the importance of TANs infiltration as a promoter of CCA cell proliferation[68,69,78]. On the other hand, CCA cells overexpress CXCL5, which promotes iCCA growth and metastasis by recruiting infiltrative intertumoral neutrophils[67]. Has been demonstrated an in vitro and in vivo cytolytic activity of NK cells against CCA[61,62]. Dendritic cells interact with lymphocyte T and Natural Killer cells to promote their activities against CCA cells[73,74]. MDSCs act on the activity of the fibroblasts and of the macrophages causing an inhibition of CCA cells[75].

MACROPHAGES

Macrophages, known for their remarkable plasticity, represent the most versatile cells within the hematopoietic system. They are present in all tissues[79] and demonstrate significant functional diversity, playing vital roles in processes such as development, tissue homeostasis, repair, and immune responses[80]. They regulate immune responses by pathogen phagocytosis and antigen presentation and additionally, they contribute to tissue formation and reorganization, wound healing, coagulation, and inflammation[81,82].

Macrophages originate from hematopoietic stem cells (HSCs) located in the bone marrow and undergo a series of sequential differentiation stages. These stages include the common myeloid progenitor (CMP), the granulocyte-macrophage progenitor (GMP)[83], the common macrophage and DC precursor (MDP)[84], and finally, the committed monocyte progenitor (cMoP)[85]. The homeostatic regulation of macrophage development is influenced by macrophage colony-stimulating factor (M-CSF), also referred to as colony-stimulating factor-1 (CSF-1)[86]. During the inflammatory state, granulocyte-macrophage colony-stimulating factor (GM-CSF) involves in the development of macrophages[87]. Macrophages are referred to by various names depending on which tissue they are found in, such as osteoclasts in bone, alveolar macrophages in the lung, microglial cells in the central nervous system, histiocytes in connective tissue, and Kupffer cells in the liver. These tissue-resident macrophage populations have such diverse transcriptional characteristics that they could potentially be classified as distinct classes of macrophages[88].

As previously stated, macrophages are plastic[79] and “ exist in a continuum of functional states ”[89]. The mononuclear phagocyte system is widely recognized for its functional versatility, and the M1 and M2 polarization paradigm represents the two extremes of the entire spectrum of macrophage functional activity[90,91]. As a result, macrophages present in distinct portions of TME display properties of both M1 and M2, depending on the tumor type[92]. The diverse signals within the TME can influence the diversity and function of TAMs, leading to their dual role in tumor progression, which can either promote or suppress tumor growth[93]. TAMs can be classified into classically activated (M1) and alternatively activated (M2) phenotypes[94,95] analogous to T helper type-1 (Th1) and T helper type-2 (Th2) of T cells[91].

M1 macrophages, also called classically activated M1 macrophages, are activated under the condition of Th1 cytokines, such as interferon-gamma (INF-γ)[96], tumor necrosis factor (TNF-α), GM-CSF[97,98] and toll-like receptor (TLR) ligands[99] alone or together with lipopolysaccharide (LPS)[100] [Figure 3]. Their activation is characterized by high antigen presenting[101] and high phagocytic capacity[102], expressing high levels of pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, IL-12, IL-23, C-X-C motif chemokine (CXCL)9, and CXCL10, and major histocompatibility complex (MHC) molecules[103]. The expression of surface proteins CD68, CD80, CD86[104], and intracellular protein suppressor cytokine signaling 3 (SOCS3)[105] are upregulated. M1 macrophages are involved in the Th1 response to infection[106].

Tumor microenvironment highlighting tumor-associated macrophages and immune cells

Figure 3. Classically activated M1 macrophages are activated by interferon-gamma (INF-γ)[96], tumor necrosis factor (TNF-α), GM-CSF[97,98] and lipopolysaccharide (LPS)[100], whereas alternatively activated macrophages are activated by M-CSF[107], prostaglandin F (PGF), prostaglandin E2 (PGE)[79], and vitamin D3. M1 phenotype is characterized by IL-1β, IL-6, IL-12, IL-23, CXCL9, CXCL10, and major MHC molecules[103] expression and M2 phenotype is characterized by IL-10, TGF-β, VEGF-A, MMP-2 and arginase-1[40] expression. M1 macrophages also express surface proteins CD68, CD80 and CD86[104], whereas M2 macrophages express CD163, CD204[99], CD206, and CD301.

M2s are activated by M-CSF[107], prostaglandin F (PGF), prostaglandin E2 (PGE2)[79], and vitamin D3[108], and they produce transforming growth factor beta (TGF-β), vascular endothelial growth factor A (VEGF-A), and matrix metalloproteinase-2 (MMP-2)[109]. M2 macrophages exhibit elevated IL-10, scavenger receptor A (CD163, CD204)[99], arginase-1[40], mannose receptor-1 (CD206), and c-type lectin (CD301) expression[110]. M2 macrophages downregulate the expression of MHC class II and IL-12 expression[40]. M2 macrophage induction can be enhanced by IL-25 and IL-33 indirectly through Th2 cells[111,112].

TAMs appear in the majority of human and experimental murine cancer models[113]. The release of macrophage components into the tumor stroma, such as epidermal growth factor (EGF), fibroblast growth factor (FGF) family proteins, TGF-β, VEGF, distinct chemokines and cytokines, enhances tumor progression and promotes cell migration and metastasis[91]. The phenotype of TAMs is defined by their gene expression profile rather than deterministic differentiation pathways and lineage choices, as seen in Th1 and Th2 cells[40]. TAMs undergo M1-like or M2-like activation in response to the effects of tumor-derived growth factors, especially M-CSF[91], after being derived from peripheral blood monocytes and recruited to TME[103]. TAMs are associated with elevated levels of anti-inflammatory cytokine expression, which can alter the immunosuppressive capacity of TME and promote tumor development via TAM-derived angiogenesis factors and proteases[106]. TAM-related subpopulations, such as, TEK tyrosine kinase receptor TIE-2 expressing monocyte subgroup (TEM)[114], myeloid-derived suppressor cells (MDSCs), and myeloid dendritic cells (mDCs) have been associated with a pro-tumorigenic inflammatory microenvironment[115].

Recent evidence shows that TAMs are obligated for tumor cell motility, invasion, and metastasis[116]. Data show that extracellular matrix (ECM) fragments produced by tumors or exosomes establish pre-metastatic niches, which is an appropriate environment for tumor cell survival and expansion induced at distant sites by the tumor[117], to be responsive to the circulating tumor cells by recruiting CD11b and VEGFR1 positive myeloid cells[118,119]. Metastatic cells must resist detachment-induced cell death, also known as anoikis, as they infiltrate into circulation. TAMs can be physically linked to cancer cells via the secretion of cytokines and assist them to travel through circulation[116].

Transcriptional profiling on oligonucleotide arrays enriched TAMs shows overexpression of angiogenic molecules[120]. Angiogenic TAMs, which are characterized by the expression of angiopoietin receptor TIE2[121,122], have an essential role in angiogenesis, regulating the angiogenic switch[123].

TAMs accumulate in tumor necrotic zones, which are characterized by low oxygen levels[124]. It is proposed that TAMs are attracted to hypoxic regions of tumors due to the release of hypoxia-induced chemo-attractants such as VEGF, endothelins, endothelial-monocyte-activating polypeptide II (EMAP2, also known as AIMP1)[125], and hypoxia-inducible factor (HIF-1) dependent factors modulating TAM migration in avascular areas[126].

Tumor-associated macrophages in cholangiocarcinoma

TAMs are immensely staged in various cancers, and CCA is one of them[18,127-129]. CCA cells and various constituents of the TME can attract TAMs by releasing factors such as monocyte chemo-attractant protein-1 (MCP-1, also known as chemokine ligand-2 or CCL2), macrophage colony-stimulating factor (M-CSF), and vascular endothelial growth factor-A (VEGF-A)[53,130-132]. Recent evidence shows that CCA upregulates TNF-like weak inducer of apoptosis (TWEAK)/fibroblast growth factor-inducible 14 (Fn14), resulting in the induce of MCP-1, CX3CL1, IL-6, IL-8, M-CSF, and GM-CSF secretion through NF-kB[132,133]. MCP-1 regulates TAM recruitment and is associated with an increase in TAM marker CD206+ and tumor aggressiveness after invading the tumor sites, straying to a pro-tumorigenic phenotype despite the initial role of macrophages[132].

Research reveals that macrophages are likely to display polarization changes during the specific stages of infection. C. sinensis (Clonorchis sinensis infection which is closely associated with CCA formation)[134] infection models in mice showed M1 phenotype polarization at the early stage of infection, followed by an increase in CD47 secretion of CCA cells which indicates blockage of phagocytosis[135], and M2 phenotype polarization at the late stages of cholangiocarcinogenesis, contributing to fibrosis and remodeling of the bile duct[136].

Pre-clinical and clinical data suggest that rich TAM content in TME is linked with poor prognosis[137]. Many studies have found a linkage between the presence of TAMs and their clinical outcomes in CCA[52,138-142]. In a study of 39 patients with intrahepatic cholangiocarcinoma (iCCA), TAM infiltration has shown an association with angiogenesis, regulatory T cell (Treg) infiltration, and poor prognosis, with CCA cells inducing M2-like phenotype via signal transducer and activator of transcription 3 (STAT3) activation[138,139]. In an immunohistochemistry (IHC) analysis of 114 patients with CCA, it has shown that there is a positive interaction between TAMs, Tregs, and tumor-infiltrating neutrophils[52]. An investigation of the mechanism of TAM-derived progression of CCA demonstrated that chronic liver injury results in the inducement of mitochondrial dysfunction, oxidative stress, and recruitment of Kupffer cells[140]. Macrophage deficiency in pre-clinical models showed inhibition of Wnt signaling and limitation in tumor progression with the promotion of apoptosis[141].

As TAM infiltration has been linked to poor patient prognosis, it has been proposed that CCA cells may alter the surrounding stroma to create a tumor-promoting immune niche. Cellular spheroids derived from CCA cells modified macrophages into TAM phenotype which express great invasive capability[142]. TAMs obtained from resected human CCA tissues recited the characteristics of CCA-educated macrophages in vitro[142].

An insight into immunotherapeutic strategies in Cholangiocarcinoma

Immune checkpoint inhibitors in Cholangiocarcinoma

Immune checkpoints are receptors expressed by immune cells that are crucial for maintaining immune balance and regulating the activity of T cells. In the TME, immune checkpoints can become overexpressed leading to a state of T cell exhaustion, characterized by impaired effector function reducing cytotoxicity and cytokine production[143].

Immune checkpoint inhibitors (ICIs) are designed to disrupt the signaling pathways mediated by immune checkpoints, thereby facilitating enhanced activation of the immune system within the tumor microenvironment[144]. Currently, the role of immunotherapy in the management of CCA is delineated based on two separate situations[145]. The first scenario evolves around the identification of specific molecular alterations in the tumor, allowing for the selection of patients who would benefit from targeted treatment. In this context, pembrolizumab, an immune ICI, has demonstrated efficacy in patients with dMMR, MSI-H, and a high TMB, leading to its approval for these specific subsets of CCA patients[146-148].

The second involves the application of immunotherapy in unselected populations of patients with advanced-stage CCA. Initially, clinical trials focused on utilizing monotherapy with ICIs. However, this approach yielded limited effectiveness in patients[149].

Consequently, alternative strategies have been pursued, primarily involving the combination of ICIs with cytotoxic chemotherapy. The positive outcomes observed from combining ICIs with cytotoxic chemotherapy have led to the approval of a specific regimen by the U.S. Food and Drug Administration (FDA) in 2022[150].

Moreover, the latest version of the National Comprehensive Cancer Network (NCCN) guidelines for hepatobiliary cancer endorses this combination as the preferred first-line treatment option for advanced-stage CCA[151]. These regulatory approvals and guidelines reflect the growing evidence supporting the efficacy of combining ICIs with cytotoxic chemotherapy in advanced-stage CCA. The gemcitabine, cisplatin, and durvalumab regimen represents a significant advancement in the treatment landscape, providing a potential treatment option that has demonstrated favorable outcomes in this patient population.

Chimeric antigen receptor in cholangiocarcinoma

Genetically engineered T cells represent a promising and potent therapeutic approach with the potential for curative responses in cancer patients. Chimeric antigen receptor (CAR)-T cell therapies have shown significant success in the treatment of hematological malignancies. However, their efficacy against solid tumors, such as solid cancers, has been comparatively limited[152]. Various strategies are being explored to target frequently expressed antigens in CCA. One approach involves the use of EGFR-specific CAR-engineered autologous CAR-T cells, which have been tested in a study involving 19 CCA patients[153]. Additionally, CAR-T cells targeting antigens such as MUC1 and CD133 have been developed and have shown promising efficacy in early in vitro studies for potential therapeutic use in CCA patients[154,155]. Further research and development are required to improve the efficacy of these immune cells against solid tumors and broaden their application to a broader spectrum of malignancies.

Novel approach to immunotherapy

Recent advances in the field of CCA treatment have emphasized the significant role of TAMs and granulocytic MDSCs[156].

TAMs are considered the primary source of programmed death-ligand 1 (PD-L1) expression in CCA. However, blocking PD-L1+ TAMs alone did not result in a reduction in tumor progression. This lack of response was attributed to the compensatory emergence of granulocytic MDSCs. These cells play a role in suppressing the immune response and promoting tumor growth. A dual approach targeting both TAMs and granulocytic MDSCs has shown promise in enhancing the response to immune checkpoint inhibitors in mice[157]. Another study has introduced a novel approach using different mouse models of CCA, which demonstrated that activating macrophages and dendritic cells through CD40 signaling significantly improves the response to anti-PD1 therapy in iCCA.

To further enhance the therapeutic response, a combination treatment involving anti-PD1, anti-CD40, gemcitabine, and cisplatin was employed. Remarkably, this combination led to a significant increase in mouse survival[158].

The recent studies highlighted the crucial significance of gaining a deeper understanding of TAMs within the TME to enhance the effectiveness of immunotherapies.

CONCLUSION

The ongoing investigation into distinct immune subpopulations and their potential role in future therapies is progressively advancing, although many aspects still lack clarity. This can be attributed to the intricate interplay among different subpopulations within the TME. In order to advance the development of innovative therapies, it is essential to gain a comprehensive understanding of the specific characteristics and functions of immune cells in CCA. We will direct our attention towards future research and investigations that explore the interaction between macrophages and dendritic cells, as these cells play a crucial role in suppressing the immune response and promoting tumor growth. This interaction suggests that targeting these cells could hold promise as a viable approach for treatment[158].

Considering the substantial impact of immune cells in shaping the characteristics of the TME, the regulation of immune cell effects in CCA presents a promising strategy for suppressing tumor progression[159].

DECLARATIONS

Authors’ contributions

Manuscript writing: Lodetti Zangrandi G, Tirpanlar D

Critical revision of the manuscript: Lleo A, Soldani C, Raggi C, Pastore M

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

All authors declared that there are no conflicts of interest.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. Razumilava N, Gores GJ. Cholangiocarcinoma. Lancet 2014;383:2168-79.

2. Cadamuro M, Fabris L, Zhang X, Strazzabosco M. Tumor microenvironment and immunology of cholangiocarcinoma. Hepatoma Res 2022;8:11.

3. Bertuccio P, Malvezzi M, Carioli G, et al. Global trends in mortality from intrahepatic and extrahepatic cholangiocarcinoma. J Hepatol 2019;71:104-14.

4. Yao KJ, Jabbour S, Parekh N, Lin Y, Moss RA. Increasing mortality in the United States from cholangiocarcinoma: an analysis of the National Center for Health Statistics Database. BMC Gastroenterol 2016;16:117.

5. Strijker M, Belkouz A, van der Geest LG, et al. Dutch Pancreatic Cancer Group. Treatment and survival of resected and unresected distal cholangiocarcinoma: a nationwide study. Acta Oncol 2019;58:1048-55.

6. Khan SA, Tavolari S, Brandi G. Cholangiocarcinoma: epidemiology and risk factors. Liver Int 2019;39 Suppl 1:19-31.

7. Lendvai G, Szekerczés T, Illyés I, et al. Cholangiocarcinoma: classification, histopathology and molecular carcinogenesis. Pathol Oncol Res 2020;26:3-15.

8. Kendall T, Verheij J, Gaudio E, et al. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int 2019;39 Suppl 1:7-18.

9. Gerber TS, Müller L, Bartsch F, et al. Integrative analysis of intrahepatic cholangiocarcinoma subtypes for improved patient stratification: clinical, pathological, and radiological considerations. Cancers 2022;14:3156.

10. Nakeeb A, Pitt HA, Sohn TA, et al. Cholangiocarcinoma. A spectrum of intrahepatic, perihilar, and distal tumors. Ann Surg 1996;224:463-73; discussion 473.

11. Nagtegaal ID, Odze RD, Klimstra D, et al. WHO Classification of Tumours Editorial Board. The 2019 WHO classification of tumours of the digestive system. Histopathology 2020;76:182-8.

12. Komuta M, Govaere O, Vandecaveye V, et al. Histological diversity in cholangiocellular carcinoma reflects the different cholangiocyte phenotypes. Hepatology 2012;55:1876-88.

13. Aishima S, Oda Y. Pathogenesis and classification of intrahepatic cholangiocarcinoma: different characters of perihilar large duct type versus peripheral small duct type. J Hepatobiliary Pancreat Sci 2015;22:94-100.

14. Nakanuma Y, Sato Y, Harada K, Sasaki M, Xu J, Ikeda H. Pathological classification of intrahepatic cholangiocarcinoma based on a new concept. World J Hepatol 2010;2:419-27.

15. Song G, Shi Y, Meng L, et al. Single-cell transcriptomic analysis suggests two molecularly subtypes of intrahepatic cholangiocarcinoma. Nat Commun 2022;13:1642.

16. Buettner S, van Vugt JL, IJzermans JN, Groot Koerkamp B. Intrahepatic cholangiocarcinoma: current perspectives. Onco Targets Ther 2017;10:1131-42.

17. DeOliveira ML, Cunningham SC, Cameron JL, et al. Cholangiocarcinoma: thirty-one-year experience with 564 patients at a single institution. Ann Surg 2007;245:755-62.

18. Banales JM, Marin JJG, Lamarca A, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol 2020;17:557-88.

19. Nakanuma Y, Kakuda Y. Pathologic classification of cholangiocarcinoma: new concepts. Best Pract Res Clin Gastroenterol 2015;29:277-93.

20. Liau JY, Tsai JH, Yuan RH, Chang CN, Lee HJ, Jeng YM. Morphological subclassification of intrahepatic cholangiocarcinoma: etiological, clinicopathological, and molecular features. Mod Pathol 2014;27:1163-73.

21. Carpino G, Cardinale V, Folseraas T, et al. Neoplastic transformation of the peribiliary stem cell niche in cholangiocarcinoma arisen in primary sclerosing cholangitis. Hepatology ;2019, 69:622-38.

22. Arai Y, Totoki Y, Hosoda F, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology 2014;59:1427-34.

23. Borger DR, Tanabe KK, Fan KC, et al. Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. Oncologist 2012;17:72-9.

24. Petrick JL, Yang B, Altekruse SF, et al. Risk factors for intrahepatic and extrahepatic cholangiocarcinoma in the United States: a population-based study in SEER-Medicare. PLoS One 2017;12:e0186643.

25. Cardinale V, Semeraro R, Torrice A, et al. Intra-hepatic and extra-hepatic cholangiocarcinoma: new insight into epidemiology and risk factors. World J Gastrointest Oncol 2010;2:407-16.

26. El-Serag HB, Engels EA, Landgren O, et al. Risk of hepatobiliary and pancreatic cancers after hepatitis C virus infection: a population-based study of U.S. veterans. Hepatology 2009;49:116-23.

27. Abdalla EK, Forsmark CE, Lauwers GY, Vauthey JN. Monolobar Caroli's disease and cholangiocarcinoma. HPB Surg 1999;11:271-6; discussion 276.

28. Mehta TI, Weissman S, Fung BM, Tabibian JH. Geoepidemiologic variation in outcomes of primary sclerosing cholangitis. World J Hepatol 2020;12:116-24.

29. Choi BI, Han JK, Hong ST, Lee KH. Clonorchiasis and cholangiocarcinoma: etiologic relationship and imaging diagnosis. Clin Microbiol Rev 2004;17:540-52, table of contents.

30. La Vecchia C, Bosetti C, Bertuccio P, et al. Trends in alcohol consumption in Europe and their impact on major alcohol-related cancers. Eur J Cancer Prev ;, 2014, 23:319-22.

31. Michelotti GA, Machado MV, Diehl AM. NAFLD, NASH and liver cancer. Nat Rev Gastroenterol Hepatol 2013;10:656-65.

32. Wise C, Pilanthananond M, Perry BF, Alpini G, McNeal M, Glaser SS. Mechanisms of biliary carcinogenesis and growth. World J Gastroenterol 2008;14:2986-9.

33. Andersen JB, Thorgeirsson SS. Genetic profiling of intrahepatic cholangiocarcinoma. Curr Opin Gastroenterol 2012;28:266-72.

34. Andersen JB. Molecular pathogenesis of intrahepatic cholangiocarcinoma. J Hepatobiliary Pancreat Sci 2015;22:101-13.

35. Höpfner M, Schuppan D, Scherübl H. Growth factor receptors and related signalling pathways as targets for novel treatment strategies of hepatocellular cancer. World J Gastroenterol 2008;14:1-14.

36. Andersen JB, Spee B, Blechacz BR, et al. Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology 2012;142:1021-1031.e15.

37. Nakamura H, Arai Y, Totoki Y, et al. Genomic spectra of biliary tract cancer. Nat Genet 2015;47:1003-10.

38. Testa U, Pelosi E, Castelli G. Cholangiocarcinoma: molecular abnormalities and cells of origin. Technol Cancer Res Treat 2023;22:15330338221128689.

39. Yang R, Song Y, Shakoor K, Yi W, Peng C, Liu S. Insights into the role of STAT3 in intrahepatic cholangiocarcinoma (Review). Mol Med Rep 2022;25:171.

40. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, and cancer. Cell 2010;140:883-99.

41. Balkwill FR, Capasso M, Hagemann T. The tumor microenvironment at a glance. J Cell Sci 2012;125:5591-6.

42. Zhou Z, Wang P, Sun R, et al. Tumor-associated neutrophils and macrophages interaction contributes to intrahepatic cholangiocarcinoma progression by activating STAT3. J Immunother Cancer 2021;9:e001946.

43. Høgdall D, Lewinska M, Andersen JB. Desmoplastic tumor microenvironment and immunotherapy in cholangiocarcinoma. Trends Cancer 2018;4:239-55.

44. Sirica AE, Gores GJ. Desmoplastic stroma and cholangiocarcinoma: clinical implications and therapeutic targeting. Hepatology 2014;59:2397-402.

45. Badalamenti G, Fanale D, Incorvaia L, et al. Role of tumor-infiltrating lymphocytes in patients with solid tumors: can a drop dig a stone? Cell Immunol 2019;343:103753.

46. Fabris L, Sato K, Alpini G, Strazzabosco M. The tumor microenvironment in cholangiocarcinoma progression. Hepatology 2021;73 Suppl 1:75-85.

47. Paijens ST, Vledder A, de Bruyn M, Nijman HW. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell Mol Immunol 2021;18:842-59.

48. Cao H, Huang T, Dai M, et al. Tumor microenvironment and its implications for antitumor immunity in cholangiocarcinoma: future perspectives for novel therapies. Int J Biol Sci 2022;18:5369-90.

49. Liu D, Heij LR, Czigany Z, et al. The role of tumor-infiltrating lymphocytes in cholangiocarcinoma. J Exp Clin Cancer Res 2022;41:127.

50. Walker LS. Treg and CTLA-4: two intertwining pathways to immune tolerance. J Autoimmun 2013;45:49-57.

51. Alvisi G, Termanini A, Soldani C, et al. Multimodal single-cell profiling of intrahepatic cholangiocarcinoma defines hyperactivated Tregs as a potential therapeutic target. J Hepatol 2022;77:1359-72.

52. Kitano Y, Okabe H, Yamashita YI, et al. Tumour-infiltrating inflammatory and immune cells in patients with extrahepatic cholangiocarcinoma. Br J Cancer 2018;118:171-80.

53. Malenica I, Donadon M, Lleo A. Molecular and immunological characterization of biliary tract cancers: a paradigm shift towards a personalized medicine. Cancers 2020; 12:2190.

54. Philip M, Schietinger A. CD8+ T cell differentiation and dysfunction in cancer. Nat Rev Immunol 2022;22:209-23.

55. Loeuillard E, Conboy CB, Gores GJ, Rizvi S. Immunobiology of cholangiocarcinoma. JHEP Rep 2019;1:297-311.

56. Shen M, Sun Q, Wang J, Pan W, Ren X. Positive and negative functions of B lymphocytes in tumors. Oncotarget 2016;7:55828-39.

57. Chen Z, Yu M, Yan J, et al. PNOC expressed by B cells in cholangiocarcinoma was survival related and LAIR2 could be a t cell exhaustion biomarker in tumor microenvironment: characterization of immune microenvironment combining single-cell and bulk sequencing technology. Front Immunol 2021;12:647209.

58. Morvan MG, Lanier LL. NK cells and cancer: you can teach innate cells new tricks. Nat Rev Cancer 2016;16:7-19.

59. Martín-Sierra C, Martins R, Laranjeira P, et al. Functional and phenotypic characterization of tumor-infiltrating leukocyte subsets and their contribution to the pathogenesis of hepatocellular carcinoma and cholangiocarcinoma. Transl Oncol 2019;12:1468-79.

60. Mikulak J, Bruni E, Oriolo F, Di Vito C, Mavilio D. Hepatic natural killer cells: organ-specific sentinels of liver immune homeostasis and physiopathology. Front Immunol 2019;10:946.

61. Polidoro MA, Mikulak J, Cazzetta V, et al. Tumor microenvironment in primary liver tumors: a challenging role of natural killer cells. World J Gastroenterol 2020;26:4900-18.

62. Jung IH, Kim DH, Yoo DK, et al. In vivo study of natural killer (NK) cell cytotoxicity against cholangiocarcinoma in a nude mouse model. In Vivo 2018;32:771-81.

63. Carnevale G, Carpino G, Cardinale V, et al. Activation of Fas/FasL pathway and the role of c-FLIP in primary culture of human cholangiocarcinoma cells. Sci Rep 2017;7:14419.

64. Giese MA, Hind LE, Huttenlocher A. Neutrophil plasticity in the tumor microenvironment. Blood 2019;133:2159-67.

65. Masucci MT, Minopoli M, Del Vecchio S, Carriero MV. The emerging role of neutrophil extracellular traps (NETs) in tumor progression and metastasis. Front Immunol 2020;11:1749.

66. Masucci MT, Minopoli M, Carriero MV. Tumor associated neutrophils. Their role in tumorigenesis, metastasis, prognosis and therapy. Front Oncol 2019;9:1146.

67. Zhou SL, Dai Z, Zhou ZJ, et al. CXCL5 contributes to tumor metastasis and recurrence of intrahepatic cholangiocarcinoma by recruiting infiltrative intratumoral neutrophils. Carcinogenesis 2014;35:597-605.

68. Ge MY, Liu ZP, Pan Y, et al. Assessment of the prognostic value of the neutrophil-to-lymphocyte ratio and platelet-to-lymphocyte ratio in perihilar cholangiocarcinoma patients following curative resection: a multicenter study of 333 patients. Front Oncol 2022;12:1104810.

69. Lin G, Liu Y, Li S, et al. Elevated neutrophil-to-lymphocyte ratio is an independent poor prognostic factor in patients with intrahepatic cholangiocarcinoma. Oncotarget 2016;7:50963-71.

70. Anderson DA 3rd, Murphy KM, Briseño CG. Development, diversity, and function of dendritic cells in mouse and human. Cold Spring Harb Perspect Biol 2018;10:a028613.

71. Spranger S, Dai D, Horton B, Gajewski TF. Tumor-Residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 2017;31:711-723.e4.

72. Conrad C, Gregorio J, Wang YH, et al. Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3+ T-regulatory cells. Cancer Res 2012;72:5240-9.

73. Thepmalee C, Panya A, Junking M, Chieochansin T, Yenchitsomanus PT. Inhibition of IL-10 and TGF-β receptors on dendritic cells enhances activation of effector T-cells to kill cholangiocarcinoma cells. Hum Vaccin Immunother 2018;14:1423-31.

74. Cazzetta V, Franzese S, Carenza C, Della Bella S, Mikulak J, Mavilio D. Natural killer-dendritic cell interactions in liver cancer: implications for immunotherapy. Cancers 2021;13:2184.

75. Ma C, Zhang Q, Greten TF. MDSCs in liver cancer: A critical tumor-promoting player and a potential therapeutic target. Cell Immunol 2021;361:104295.

76. Veglia F, Sanseviero E, Gabrilovich DI. Myeloid-derived suppressor cells in the era of increasing myeloid cell diversity. Nat Rev Immunol 2021;21:485-98.

77. Xu XD, Hu J, Wang M, et al. Circulating myeloid-derived suppressor cells in patients with pancreatic cancer. Hepatobiliary Pancreat Dis Int 2016;15:99-105.

78. Gu FM, Gao Q, Shi GM, et al. Intratumoral IL-17+ cells and neutrophils show strong prognostic significance in intrahepatic cholangiocarcinoma. Ann Surg Oncol 2012;19:2506-14.

79. Mosser DM, Edwards JP. Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008;8:958-69.

80. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature 2013;496:445-55.

81. Anderson NM, Simon MC. The tumor microenvironment. Curr Biol 2020;30:R921-5.

82. Peiseler M, Kubes P. Macrophages play an essential role in trauma-induced sterile inflammation and tissue repair. Eur J Trauma Emerg Surg 2018;44:335-49.

83. Akashi K, Traver D, Miyamoto T, Weissman IL. A clonogenic common myeloid progenitor that gives rise to all myeloid lineages. Nature 2000;404:193-7.

84. Fogg DK, Sibon C, Miled C, et al. A clonogenic bone marrow progenitor specific for macrophages and dendritic cells. Science 2006;311:83-7.

85. Hettinger J, Richards DM, Hansson J, et al. Origin of monocytes and macrophages in a committed progenitor. Nat Immunol 2013;14:821-30.

86. Hamilton JA. Colony-stimulating factors in inflammation and autoimmunity. Nat Rev Immunol 2008;8:533-44.

87. Kumar A, Taghi Khani A, Sanchez Ortiz A, Swaminathan S. GM-CSF: a double-edged sword in cancer immunotherapy. Front Immunol 2022;13:901277.

88. Gautier EL, Shay T, Miller J, et al. Immunological Genome Consortium. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat Immunol 2012;13:1118-28.

89. Kim J, Bae JS. Tumor-Associated macrophages and neutrophils in tumor microenvironment. Mediators Inflamm 2016;2016:6058147.

90. Mantovani A, Allavena P, Sica A. Tumour-associated macrophages as a prototypic type II polarised phagocyte population: role in tumour progression. Eur J Cancer 2004;40:1660-7.

91. Sica A, Allavena P, Mantovani A. Cancer related inflammation: the macrophage connection. Cancer Lett 2008;267:204-15.

92. Hinshaw DC, Shevde LA. The tumor microenvironment innately modulates cancer progression. Cancer Res 2019;79:4557-66.

93. Salmaninejad A, Valilou SF, Soltani A, et al. Tumor-associated macrophages: role in cancer development and therapeutic implications. Cell Oncol 2019;42:591-608.

94. Mills CD, Kincaid K, Alt JM, Heilman MJ, Hill AM. M-1/M-2 macrophages and the Th1/Th2 paradigm J Immunol 2000. pp. 6166-73.

95. Biswas SK, Mantovani A. Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 2010;11:889-96.

96. Billiau A, Matthys P. Interferon-gamma: a historical perspective. Cytokine Growth Factor Rev 2009;20:97-113.

97. Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. Granulocyte-macrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation. J Immunol 2007;178:5245-52.

98. Zhu J, Zhi Q, Zhou BP, Tao M, Liu J, Li W. The role of tumor associated macrophages in the tumor microenvironment: mechanism and functions. Anticancer Agents Med Chem 2016;16:1133-41.

99. Jayasingam SD, Citartan M, Thang TH, Mat Zin AA, Ang KC, Ch'ng ES. Evaluating the polarization of tumor-associated macrophages into M1 and M2 phenotypes in human cancer tissue: technicalities and challenges in routine clinical practice. Front Oncol 2019;9:1512.

100. Sica A, Larghi P, Mancino A, et al. Macrophage polarization in tumour progression. Semin Cancer Biol 2008;18:349-55.

101. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity 2010;32:593-604.

102. Tamura R, Tanaka T, Yamamoto Y, Akasaki Y, Sasaki H. Dual role of macrophage in tumor immunity. Immunotherapy 2018;10:899-909.

103. Wu K, Lin K, Li X, et al. Redefining tumor-associated macrophage subpopulations and functions in the tumor microenvironment. Front Immunol 2020;11:1731.

104. de Sousa JR, de Sousa RP, Aarão TL, et al. In situ expression of M2 macrophage subpopulation in leprosy skin lesions. Acta Trop 2016;157:108-14.

105. Huang X, Li Y, Fu M, Xin H. Polarizing macrophages in vitro. In: Rousselet G, editor. Macrophages. New York: Springer; 2018. pp. 119-26.

106. Yang L, Zhang Y. Tumor-associated macrophages: from basic research to clinical application. J Hematol Oncol 2017;10:58.

107. Satoh T, Takeuchi O, Vandenbon A, et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol 2010;11:936-44.

108. Kaler P, Augenlicht L, Klampfer L. Macrophage-derived IL-1beta stimulates Wnt signaling and growth of colon cancer cells: a crosstalk interrupted by vitamin D3. Oncogene 2009;28:3892-902.

109. Chen Y, Zhang X. Pivotal regulators of tissue homeostasis and cancer: macrophages. Exp Hematol Oncol 2017;6:23.

110. Martinez F O, Gordon S, Locati M, et al. Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol 2006;177:7303-7311.

111. Caccamo N, Todaro M, Sireci G, Meraviglia S, Stassi G, Dieli F. Mechanisms underlying lineage commitment and plasticity of human γδ T cells. Cell Mol Immunol 2013;10:30-4.

112. O'Shea JJ, Paul WE. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 2010;327:1098-102.

113. Qian B Z, Pollard J W. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141:39-51.

114. De Palma M, Venneri MA, Galli R, et al. Tie2 identifies a hematopoietic lineage of proangiogenic monocytes required for tumor vessel formation and a mesenchymal population of pericyte progenitors. Cancer Cell 2005;8:211-26.

115. Sica A, Bronte V. Altered macrophage differentiation and immune dysfunction in tumor development. J Clin Invest 2007;117:1155-66.

116. Condeelis J, Pollard JW. Macrophages: obligate partners for tumor cell migration, invasion, and metastasis. Cell 2006;124:263-6.

117. Peinado H, Zhang H, Matei IR, et al. Pre-metastatic niches: organ-specific homes for metastases. Nat Rev Cancer 2017;17:302-17.

118. Psaila B, Lyden D. The metastatic niche: adapting the foreign soil. Nat Rev Cancer 2009;9:285-93.

119. Peinado H, Alečković M, Lavotshkin S, et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 2012;18:883-91.

120. Ojalvo LS, Whittaker CA, Condeelis JS, Pollard JW. Gene expression analysis of macrophages that facilitate tumor invasion supports a role for Wnt-signaling in mediating their activity in primary mammary tumors. J Immunol 2010;184:702-12.

121. Mazzieri R, Pucci F, Moi D, et al. Targeting the ANG2/TIE2 axis inhibits tumor growth and metastasis by impairing angiogenesis and disabling rebounds of proangiogenic myeloid cells. Cancer Cell 2011;19:512-26.

122. Murdoch C, Muthana M, Coffelt SB, Lewis CE. The role of myeloid cells in the promotion of tumour angiogenesis. Nat Rev Cancer 2008;8:618-31.

123. Lin EY, Li JF, Gnatovskiy L, et al. Macrophages regulate the angiogenic switch in a mouse model of breast cancer. Cancer Res 2006;66:11238-46.

124. Lewis C, Murdoch C. Macrophage responses to hypoxia: implications for tumor progression and anti-cancer therapies. Am J Pathol 2005;167:627-35.

125. Murdoch C, Giannoudis A, Lewis CE. Mechanisms regulating the recruitment of macrophages into hypoxic areas of tumors and other ischemic tissues. Blood 2004;104:2224-34.

126. Talks KL, Turley H, Gatter KC, et al. The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol 2000;157:411-21.

127. Vigano L, Soldani C, Franceschini B, et al. Tumor-Infiltrating lymphocytes and macrophages in intrahepatic cholangiocellular carcinoma. Impact on prognosis after complete surgery. J Gastrointest Surg 2019;23:2216-24.

128. Donadon M, Torzilli G, Cortese N, et al. Macrophage morphology correlates with single-cell diversity and prognosis in colorectal liver metastasis. J Exp Med 2020:217.

129. Gazzillo A, Polidoro MA, Soldani C, Franceschini B, Lleo A, Donadon M. Relationship between epithelial-to-mesenchymal transition and tumor-associated macrophages in colorectal liver metastases. Int J Mol Sci 2022;23:16197.

130. Fabris L, Perugorria MJ, Mertens J, et al. The tumour microenvironment and immune milieu of cholangiocarcinoma. Liver Int 2019;39 Suppl 1:63-78.

131. Ziani L, Chouaib S, Thiery J. Alteration of the antitumor immune response by cancer-associated fibroblasts. Front Immunol 2018;9:414.

132. Dwyer BJ, Jarman EJ, Gogoi-Tiwari J, et al. TWEAK/Fn14 signalling promotes cholangiocarcinoma niche formation and progression. J Hepatol 2021;74:860-72.

133. Ruffolo LI, Jackson KM, Kuhlers PC, et al. GM-CSF drives myelopoiesis, recruitment and polarisation of tumour-associated macrophages in cholangiocarcinoma and systemic blockade facilitates antitumour immunity. Gut 2022;71:1386-98.

134. Pak JH, Lee JY, Jeon BY, Dai F, Yoo WG, Hong SJ. Cytokine production in cholangiocarcinoma cells in response to clonorchis sinensis excretory-secretory products and their putative protein components. Korean J Parasitol 2019;57:379-87.

135. Vaeteewoottacharn K, Kariya R, Pothipan P, et al. Attenuation of CD47-SIRPα signal in cholangiocarcinoma potentiates tumor-associated macrophage-mediated phagocytosis and suppresses intrahepatic metastasis. Transl Oncol 2019;12:217-25.

136. Kim EM, Kwak YS, Yi MH, Kim JY, Sohn WM, Yong TS. Clonorchis sinensis antigens alter hepatic macrophage polarization in vitro and in vivo. PLoS Negl Trop Dis 2017;11:e0005614.

137. Bingle L, Brown NJ, Lewis CE. The role of tumour-associated macrophages in tumour progression: implications for new anticancer therapies. J Pathol 2002;196:254-65.

138. Hasita H, Komohara Y, Okabe H, et al. Significance of alternatively activated macrophages in patients with intrahepatic cholangiocarcinoma. Cancer Sci 2010;101:1913-9.

139. Sun D, Luo T, Dong P, et al. M2-polarized tumor-associated macrophages promote epithelial-mesenchymal transition via activation of the AKT3/PRAS40 signaling pathway in intrahepatic cholangiocarcinoma. J Cell Biochem 2020;121:2828-38.

140. Yuan D, Huang S, Berger E, et al. Kupffer cell-derived Tnf triggers cholangiocellular tumorigenesis through jnk due to chronic mitochondrial dysfunction and ROS. Cancer Cell 2017;31:771-789.e6.

141. Boulter L, Guest RV, Kendall TJ, et al. WNT signaling drives cholangiocarcinoma growth and can be pharmacologically inhibited. J Clin Invest 2015;125:1269-85.

142. Raggi C, Correnti M, Sica A, et al. Cholangiocarcinoma stem-like subset shapes tumor-initiating niche by educating associated macrophages. J Hepatol 2017;66:102-15.

143. Johnson DB, Nebhan CA, Moslehi JJ, Balko JM. Immune-checkpoint inhibitors: long-term implications of toxicity. Nat Rev Clin Oncol 2022;19:254-67.

144. Ilyas SI, Affo S, Goyal L, et al. Cholangiocarcinoma-novel biological insights and therapeutic strategies. Nat Rev Clin Oncol 2023;20:470-86.

145. Lamarca A, Edeline J, Goyal L. How I treat biliary tract cancer. ESMO Open 2022;7:100378.

146. Maio M, Ascierto PA, Manzyuk L, et al. Pembrolizumab in microsatellite instability high or mismatch repair deficient cancers: updated analysis from the phase II KEYNOTE-158 study. Ann Oncol 2022;33:929-38.

147. . Food and Drug Administration US Food and Drug Administration. FDA grants accelerated approval to pembrolizumab for first tissue/site agnostic indication; 2017.

148. . Food and Drug Administration US Food and Drug Administration. FDA approves pembrolizumab for adults and children with TMB-H solid tumors; 2020.

149. Piha-Paul SA, Oh DY, Ueno M, et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: results from the KEYNOTE-158 and KEYNOTE-028 studies. Int J Cancer 2020;147:2190-8.

150. Food and Drug Administration US Food and Drug Administration. FDA D.I.S.C.O. burst edition: FDA approval of Imfinzi (durvalumab) for adult patients with locally advanced or metastatic biliary tract cancer. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-disco-burst-edition-fda-approval-imfinzi-durvalumab-adult-patients-locally-advanced-or [Last accessed on 28 Jul 2023].

151. NCCN National Comprehensive Cancer Network. NCCN guidelines: hepatobiliary cancer. Available from: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1438 [Last accessed on 28 Jul 2023].

152. June CH, Sadelain M. Chimeric antigen receptor therapy. N Engl J Med 2018;379:64-73.

153. Guo Y, Feng K, Liu Y, et al. Phase I study of chimeric antigen receptor-modified T Cells in patients with EGFR-Positive advanced biliary tract cancers. Clin Cancer Res 2018;24:1277-86.

154. Supimon K, Sangsuwannukul T, Sujjitjoon J, et al. Anti-mucin 1 chimeric antigen receptor T cells for adoptive T cell therapy of cholangiocarcinoma. Sci Rep 2021;11:6276.

155. Sangsuwannukul T, Supimon K, Sujjitjoon J, et al. Anti-tumour effect of the fourth-generation chimeric antigen receptor T cells targeting CD133 against cholangiocarcinoma cells. Int Immunopharmacol 2020;89:107069.

156. Greten TF, Schwabe R, Bardeesy N, et al. Immunology and immunotherapy of cholangiocarcinoma. Nat Rev Gastroenterol Hepatol 2023;20:349-65.

157. Loeuillard E, Yang J, Buckarma E, et al. Targeting tumor-associated macrophages and granulocytic myeloid-derived suppressor cells augments PD-1 blockade in cholangiocarcinoma. J Clin Invest 2020;130:5380-96.

158. Diggs LP, Ruf B, Ma C, et al. CD40-mediated immune cell activation enhances response to anti-PD-1 in murine intrahepatic cholangiocarcinoma. J Hepatol 2021;74:1145-54.

159. Zhou M, Wang C, Lu S, et al. Tumor-associated macrophages in cholangiocarcinoma: complex interplay and potential therapeutic target. EBioMedicine 2021;67:103375.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Lodetti Zangrandi G, Tirpanlar D, Pastore M, Soldani C, Lleo A, Raggi C. Tumor microenvironment highlighting tumor-associated macrophages and immune cells. Hepatoma Res 2023;9:32. http://dx.doi.org/10.20517/2394-5079.2023.32

AMA Style

Lodetti Zangrandi G, Tirpanlar D, Pastore M, Soldani C, Lleo A, Raggi C. Tumor microenvironment highlighting tumor-associated macrophages and immune cells. Hepatoma Research. 2023; 9: 32. http://dx.doi.org/10.20517/2394-5079.2023.32

Chicago/Turabian Style

Lodetti Zangrandi, Giulio, Dilruba Tirpanlar, Mirella Pastore, Cristiana Soldani, Ana Lleo, Chiara Raggi. 2023. "Tumor microenvironment highlighting tumor-associated macrophages and immune cells" Hepatoma Research. 9: 32. http://dx.doi.org/10.20517/2394-5079.2023.32

ACS Style

Lodetti Zangrandi, G.; Tirpanlar D.; Pastore M.; Soldani C.; Lleo A.; Raggi C. Tumor microenvironment highlighting tumor-associated macrophages and immune cells. Hepatoma. Res. 2023, 9, 32. http://dx.doi.org/10.20517/2394-5079.2023.32

About This Article

Special Issue

This article belongs to the Special Issue Molecular Pathogenesis and Drivers of Cholangiocarcinoma
© The Author(s) 2023. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License (https://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, sharing, adaptation, distribution and reproduction in any medium or format, for any purpose, even commercially, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Data & Comments

Data

Views
362
Downloads
105
Citations
1
Comments
0
3

Comments

Comments must be written in English. Spam, offensive content, impersonation, and private information will not be permitted. If any comment is reported and identified as inappropriate content by OAE staff, the comment will be removed without notice. If you have any queries or need any help, please contact us at support@oaepublish.com.

0
Download PDF
Cite This Article 6 clicks
Like This Article 3 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Hepatoma Research
ISSN 2454-2520 (Online) 2394-5079 (Print)

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/

Portico

All published articles are preserved here permanently:

https://www.portico.org/publishers/oae/