Next Article in Journal
Sex Influence on Autophagy Markers and miRNAs in Basal and Angiotensin II-Treated Human Umbilical Vein Endothelial Cells
Next Article in Special Issue
A Review of CAR-T Combination Therapies for Treatment of Gynecological Cancers
Previous Article in Journal
Dysautonomia in Amyotrophic Lateral Sclerosis
Previous Article in Special Issue
Biomarkers for Immunotherapy in Driver-Gene-Negative Advanced NSCLC
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Immune Regulatory Role of Adenosine in the Tumor Microenvironment

1
Department of Immunology, School of Basic Medicine, China Medical University, Shenyang 100001, China
2
College of Life Science and Bioengineering, Beijing Jiaotong University, Beijing 100044, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(19), 14928; https://doi.org/10.3390/ijms241914928
Submission received: 6 September 2023 / Revised: 30 September 2023 / Accepted: 3 October 2023 / Published: 5 October 2023
(This article belongs to the Special Issue Immunotherapy for Cancer 2023)

Abstract

:
Adenosine, an immunosuppressive metabolite, is produced by adenosine triphosphate (ATP) released from dying or stressed cells and is found at high levels in the tumor microenvironment of most solid tumors. It mediates pro-tumor activities by inducing tumor cell proliferation, migration or invasion, tumor tissue angiogenesis, and chemoresistance. In addition, adenosine plays an important role in regulating anti-tumor immune responses and facilitating tumor immune escape. Adenosine receptors are broadly expressed by tumor-infiltrated immune cells, including suppressive tumor-associated macrophages and CD4+ regulatory T cells, as well as effector CD4+ T cells and CD8+ cytotoxic T lymphocytes. Therefore, adenosine is indispensable in down-regulating anti-tumor immune responses in the tumor microenvironment and contributes to tumor progression. This review describes the current progress on the role of adenosine/adenosine receptor pathway in regulating the tumor-infiltrating immune cells that contribute to tumor immune evasion and aims to provide insights into adenosine-targeted tumor immunotherapy.

1. Introduction

Adenosine, an endogenous purine nucleoside, plays an important role in regulating immune responses. Adenosine is mainly formed by consecutive extracellular adenosine triphosphate (eATP) dephosphorylation catalyzed by ectonucleoside triphosphate di-phosphohydrolase 1 (CD39) and ecto-5′-nucleotidase (CD73), in which CD39 initially mediates dephosphorylation of ATP to adenosine diphosphate (ADP) and adenosine monophosphate (AMP), then CD73 converts AMP to adenosine. Adenosine is degraded into inosine by adenosine deaminase (ADA), which occurs both intracellularly and extracellularly. Although the CD39/CD73 pathway is still considered to be the major source of adenosine in the tumor microenvironment (TME) [1,2], additional ectoenzymes also contribute to the metabolism of extracellular nucleotides. For instance, nicotinamide adenine dinucleotide (NAD+) can also be released in the hypoxic TME by the salvage pathway and can be hydrolyzed by CD38 to form ADP ribose. ADP is then further degraded to AMP through the CD38/CD203a/CD73 pathway [3,4,5]. Adenosine could be transported into the cell by ENTs (both directions) and CNTs (one-way transport). Intracellular adenosine can be produced by hydrolyzing AMP through cytoplasmic 5′-nucleotidase-I (cN-I) [6,7] or by hydrolyzing S-adenosine homocysteine (SAH) through SAH hydrolase. The generated adenosine is either phosphorylated to AMP by adenosine kinase (ADK) or degraded to inosine by ADA [8,9,10,11]. eATP is released by dying or stressed cells [12]. Meanwhile, tumor-derived exosomes (TEX) were shown to be associated with cellular stress, such as hypoxia, acidic pH, and many other triggers present in the TME [13]. Studies have shown that the CD39 and CD73 carried by TEX are enzymatically active and can produce adenosine [14,15] (Figure 1).
Under physiological conditions, the main role of adenosine is tissue protection and anti-injury to counteract the pro-immunogenic and pro-inflammatory activities of eATP [7]. However, under pathological conditions, increased levels of adenosine are involved in anti-inflammatory responses in the tissues and suppressive anti-tumor immunity in various cancers [16,17,18]. For example, extracellular adenosine concentrations are 10 to 20 times higher than normal levels in inflamed tissues in the context of ischemia, trauma, and inflammation [19,20,21,22]. Accumulating evidence shows that adenosine can also be produced at high levels as a metabolite in the TME of major solid tumors [23,24], serving as immune suppressive molecules that contribute to tumor immune escape by modulating various immune cells via receptor-dependent/receptor-independent mechanisms. Secondly, adenosine indirectly affects the concentration of other factors in the adenosinergic pathway, such as ATP, AMP [6], inosine [25], adenosine deaminase (ADAR) [26], and AMP-activated protein kinase (AMPK) [27]. These factors regulate the communication between tumor cells and immune cells by different mechanisms.
To restore immunosurveillance, largely by blocking adenosine-producing enzymes in the TME and adenosine receptors on immune cells [17]. There are four adenosine receptor subtypes, which belong to the family of G protein-coupled receptors (GPCRs), including A1, A2A, A2B, and A3 receptors (A1R, A2AR, A2BR, and A3R) [19]. A1R is mainly expressed in the central nervous system, whereas A2BR and A3R are mostly expressed in the peripheral tissues and participate in inflammation and immune responses. A2AR is predominantly expressed in various immune cells, both in central and peripheral lymphoid tissues. When adenosine levels are low, its interaction occurs preferentially with the A1R and/or A3R, activating the Gi/o protein and thus reducing adenylate cyclase (AC) and protein kinase A (PKA). At higher levels, adenosine activates the A2AR and/or A2BR components, activating the AC/cyclic AMP (cAMP)/PKA cascade through the Gs protein, thereby regulating intracellular cAMP levels that control the activity of various cells [28,29,30]. It has been demonstrated that A2AR is broadly expressed on several immune cells, including tumor-associated macrophages (TAMs), CD4+ regulatory T cells (Tregs), effector CD4+ T cells, and cytotoxic T lymphocytes at distant levels, in the TME of major solid cancers [8,31]. Therefore, adenosine may act prominently as a key regulator to control anti-tumor immunity and may serve as a potential target for tumor immunotherapy. Here, we describe the current progress on the role of the adenosine/adenosine receptor pathway in regulating the tumor-infiltrated immune cells that contribute to tumor immune escape and introduce the current status of targeted adenosine/adenosine receptor therapy. In addition, we evaluate the limitations of targeting this pathway, aiming to provide insights into adenosine-targeted tumor immunotherapy.

2. The Role of Adenosine on the Regulation of Various Immune Cells in the TME

Various immune cells infiltrate tumor tissues that control tumor progression. Some of these cells mediate tumor-antagonizing immune responses that suppress tumor progression, such as natural killer (NK) cells, dendritic cells (DCs), effector CD4+ T cells, and CD8+ T cells. Several immune cells may serve as suppressors that inhibit anti-tumor immunity, facilitating tumor progression. These cells include Tregs, TAMs, and myeloid-derived suppressor cells (MDSCs) [32,33]. Accumulating evidence shows that most of these cells express CD39/CD73 and/or adenosine receptors in the TME that could be redundantly regulated by adenosine, thus controlling tumor development [1,34,35,36].

2.1. Natural Killer Cells

NK cells arise from hematopoietic stem cells originating in the bone marrow and further differentiate from the common lymphoid progenitor cell. The activation of NK cells is determined by the balance of activating and inhibitory signals on their surface upon interaction with cognate ligands in potential target cells. NK cells can also be activated by cytokines alone without target cell interaction, such as interferon (IFN)-γ [37]. It has been shown that adenosine has specific immunomodulatory effects on the maturation, migration, and effector functions of NK cells [38,39,40]. In addition to inhibiting the maturation of NK cells and limiting the accumulation of cytotoxic CD56dim subsets [40], adenosine also prevents the transport of NK cells to tumor sites by changing the chemokine environment [41] and inhibits the function of NK cell effector molecules against tumor targets [42]. Tumor-infiltrating NK cells upregulate CD73 expression, and the frequency of these CD73+ NK cells correlate with larger tumor sizes in breast cancer patients. CD73+ NK cells undergo transcriptional reprogramming and upregulate interleukin (IL)-10 production via STAT3 transcriptional activity, suppressing CD4+ T cell proliferation and IFN-γ production [43].
Notably, adenosine inhibits the cytotoxic effect of NK cells mainly through A2AR signaling and causes tumor immune escape in several solid tumors, such as MethA sarcoma and 3LL Lewis lung carcinoma, through cAMP-dependent signaling that mediates PKA engagement [44,45,46]. A clinical study showed that CD56dimCD16 and CD56brightCD16 NK cells represent the predominant NK cell subpopulations in acute myeloid leukemia (AML) and that CD39+/CD38+ cells cluster on CD56brightCD16 NK cells. Combined targeting of CD39 or A2AR significantly augments the anti-TIGIT-mediated lysis of AML cells [38]. Meanwhile, blocking the activity of the CD73 enzyme shows increased NK cell killing of tumor cells and an obvious anti-tumor response [47]. In addition, A2BR antagonists rescue T and NK cell proliferation, increase IFN-γ and perforin production, and increase tumor-infiltrating lymphocyte infiltration into breast cancer spheroids [48]. Therefore, targeting inhibition of adenosine/adenosine receptors may enhance NK cell activities that positively regulate the anti-tumor immune response that inhibits tumor progression.

2.2. Dendritic Cells

DCs play an important role in the initiation of T cell-mediated anti-tumor immune responses. While immature DCs exhibit a potent capacity for taken-up antigen and antigen processing, mature DCs show efficient presentation in the context of MHC molecules to T cells, thus initiating anti-tumor immune responses. Diverse DC subsets have been identified in the tumor microenvironment and tumor-draining lymph nodes, including the CD103+ cDC1 subset, CD11b+ cDC2 subset, and B220+ plasmacytoid DCs [49]. Adenosine has been found to negatively regulate the antigen presentation process of DCs in the TME. A2AR and/or A2BR+ DCs showed decreased CD4+ T cell priming and anti-tumor immune responses in the TME [50,51]. Conversely, blocking A2ARs deprives DCs of their contact with naïve conventional T cells, which leads to suppressed T cell priming and antigen-specific T cell responses. Indeed, in colorectal cancer, melanoma, and fibrosarcoma in mice, an A2AR antagonist enhanced the antigen presentation capacity of CD103+ DCs as well as increased T cell function [52]. Furthermore, it has been shown that blocking CD73 signaling in patients with colon adenocarcinoma synergistically enhances oxaliplatin (OXP)-induced ATP release, a hallmark of immunogenic cell death, which promotes DC maturation and immune cell infiltration [53].
Adenosine also regulates the differentiation and maturation of DCs in the TME. Upon binding to adenosine, DCs preferentially differentiate into a myeloid DC population through a mechanism driven by expressing high levels of tolerogenic factors, such as COX-2, IDO, IL-6, IL-8, IL-10, TGF-β, and VEGF, thereby favoring the activation of suppressive CD4+ Tregs [51,54]. Constructed nanoparticles assembling small molecular A2AR inhibitors showed enhanced DC activation and increased infiltrating CD8+ T cells in the TME, leading to suppression of tumor progression [55].

2.3. Effector CD4+ T Helper Cells

CD4+ T cells play an important role in tumor immunity. Upon activation in response to antigen stimulation in the context of MHC class II molecules, CD4+ T cells undergo proliferation and differentiation to generate effector CD4+ T cells in the draining lymphoid tissues of the tumor. CD4+ T cells secrete different cytokine profiles that are closely associated with anti-tumor immunity in various cancers. T helper 1 (Th1), T helper 2 (Th2), and T helper 17 (Th17) cells have been found in the TME of various inflamed cancers. It is believed that Th1 cells promote CD8+ T cell immunity to tumor cells as well as enhance anti-tumor immune responses by recruiting inflammatory cells, such as macrophages, granulocytes, and NK cells, to the site of tumor tissue. In contrast, Th2 cells are required for priming B cell activation and anti-tumor antibody production [56,57]. Studies have shown that adenosine inhibits the proliferation of Th1 and Th2 cells through binding to A2AR on the surface of cells [58,59]. The role of adenosine in the differentiation and function of Th17 cells remains unclear. Some studies showed that both CD73+ Th17 cells and CD39+ Th19 cells may function as immune suppressor cells instead of effector cells, with increased IL-10 production that favors the development of cancer in various murine models, including EL4 thymoma, B16F10 melanoma, LLC lung carcinoma, and MC38 colon carcinoma. Furthermore, infiltrated CD39+ Th17 cells in the TME are proportional to poor clinical outcomes in cancer patients [60,61]. Interestingly, adenosine and an A2AR agonist (PSB0777) promoted IL-17A and IL-8 production from human peripheral blood mononuclear cells in response to Candida albicans stimulation, suggesting a role for the adenosine/A2AR pathway in Th17 cell differentiation [62]. In addition, in vivo experiments in mice showed that A2BR promotes Th17 differentiation by augmenting IL-6 production by DCs, independent of intracellular cAMP, suggesting a different mechanism for Th17 cell differentiation by A2BR [63].

2.4. Cytotoxic CD8+ T Cells

CD8+ T cells are the most potent killing cells, with the ability to specifically recognize and eradicate immunogenic cancer cells. However, tumor-reactive CD8+ T cells become dysfunctional in the context of suppressive TMEs with the development of tumor progression. Adenosine is the key suppressive molecule in the TME and may induce tumor-reactive CD8+ T cells to become dysfunctional. Studies have shown that CD8+ T central memory cells (TCM) in the TME express high levels of A2AR that are susceptible to regulation by adenosine, leading to CD8+ T cells being functionally exhausted in the TME [64,65]. In addition, memory CD8+ T cells highly expressed CD73, which contrasts with terminally differentiated effector cells that did not express CD73 [66]. Studies have shown that tumor-infiltrating CD39+CD8+ T cells show less cytotoxicity compared with those of CD39CD8+ T cells, which are characterized by the production of IFN-γ, tumor necrosis factor (TNF)-α, and granzyme B, suggesting that tumor-infiltrating CD8+ T cells with high CD39 expression exhibited features of exhaustion [67]. Indeed, CD39+CD8+ T cells exhibited a CD69+PD-1+perforinlowIFNγlow “exhausted” phenotype [68].
In addition, CD73 may control glucose uptake by CD8+ T cells through the production of adenosine, resulting in a decrease in the efficiency of T cells to control tumor growth. CD73-deficient CD8+ T cells showed increased glucose uptake and mitochondrial respiration and improved anti-tumor ability in melanoma-bearing mice [69]. The expression level of CD73 on CD8+ T cells is regulated by costimulatory signals initiated by the binding of CD28 on the surface of CD8+ T cells to the B7 molecules on target cells. In the absence of CD28 costimulation, CD73 expression levels on CD8+ T cells are upregulated in the TME in several solid cancers [70].

2.5. Tumor-Associated Macrophages

TAMs play an important role in regulating tumor progression and chemotherapy resistance. Macrophages display highly tumor environment-dependent plasticity that varies their biological function. Macrophages in the TME can be polarized into opposite functional states, known as M1 and M2 polarization [71,72,73]. M1 and M2 are classified as highly simplified models of complex functional behavior and cellular plasticity. The M1 phenotype is characterized by the expression of high levels of proinflammatory cytokines, high production of reactive nitrogen and oxygen intermediates, promotion of the Th1 response, and strong microbicidal and tumoricidal activity. In contrast, M2 macrophages are considered to be involved in parasite containment, promotion of tissue remodeling, and tumor progression and have immunoregulatory functions [74,75,76]. Many studies have shown that adenosine is a necessary element for tumor-induced macrophage proliferation. Macrophages secrete GM-CSF and enhance the expression of A2AR on macrophages in the presence of adenosine, thus initiating macrophage proliferation in hepatocellular carcinoma (HCC). Mechanistic analysis showed that tumor-derived adenosine binds to A2AR of TAMs, promoting M2-like macrophage polarization as well as proliferation via the activation of phosphatidylinositol-3-kinase (PI3K)/Akt and MEK/ERK pathways [77].
Previous studies have shown that the PI3Kγ signaling pathway of TAMs inhibits the activation of NF-κB through Akt and mTOR, which mediates immunosuppression to promote tumor growth. At the same time, the PI3Kγ signal in TAMs inhibits the recruitment of CD8+ T cells around the tumor [78,79]. High expression of A2ARs on tumor cells promotes the secretion of chemokines and polarizing factors by activating the PI3K/AKT/NF-κB pathway, thereby promoting the migration and invasion of TAMs [80]. Although the main population of TAMs is immunosuppressive M2 macrophages, TAMs can be reprogrammed into M1 helper macrophages [78,79]. PI3Kγ inhibition can reverse these effects of TAMs by polarizing macrophages into NF-κB-dependent pro-inflammatory M1 macrophages [78]. Therefore, it is of great significance to identify the key checkpoint where TAMs are reprogrammed into M1 macrophages to further promote the killing effect of M1 macrophages on tumors. A2AR knockdown increases M1 polarization in TAMs [80,81]. In ovarian cancer, inhibition of CD39 or CD73 can reverse the suppression of T cell proliferation mediated by TAMs [82]. Hypoxia can induce the expression of A2AR and A2BR, and reduce the expression of adenosine kinase and balanced nucleoside transporters. In addition, the hypoxia microenvironment and tumor mTOR signal can stabilize or induce the expression of HIF-1α, respectively, and HIF-1α can induce the expression of CD39 and CD73 [75]. Therefore, metabolic changes in the TME promote the accumulation of adenosine in the interstitial space, and hypoxia further enhances the level of adenosine in the TME, which leads to immunosuppression by promoting M2 polarization. In addition, CD73 antibody triggers a strong accumulation of M1-type macrophages in non-small-cell lung cancer [83]. The metabolic changes of adenosine are very important for the reprogramming of TAMs. Blocking the adenosine/adenosine receptor pathway to promote M1 polarization of TAMs to establish an effective anti-tumor response is expected to become another auxiliary means of anti-tumor immunotherapy.

2.6. Myeloid-Derived Suppressor Cells

Bone marrow myeloid-derived suppressor cells (MDSCs) are a heterogeneous immune suppressive population that can be granulocytic or monocytic. MDSCs may mediate immune suppression via multiple mechanisms, including the release of proangiogenic factor VEGF, matrix remodeling enzyme MMPs, and inhibitory cytokine TGF-β [84]. It has been shown that MDSCs produce extracellular adenosine by expressing CD39 and CD73 [85,86]. Genetic ablation of CD73 led to decreased MDSCs in orthotopic mouse models of pancreatic ductal adenocarcinoma (PDAC) [87]. The high expression of adenosine produced by MDSCs in the TME may promote the proliferation of MDSCs themselves and their immunosuppressive activity in the mouse model of Lewis lung cancer [85]. Adenosine also improves the survival of MDSCs in the TME. Hypoxia is the characteristic feature of HCC that contributes to tumor progression. Hypoxia induces enhanced expression of CD39 through stabilization of HIF-1 in the tumor tissue of HCC, thereby preventing differentiation but promoting the survival of MDSCs [88].
A study showed that CD73-expressing MDSCs in the TME exhibited superior T cell suppressor function compared with CD73 MDSCs in murine tumor models, including lung, colon, and melanoma. Mechanistically, tumor-derived prostaglandin E2 (PGE2), induces CD73 expression in MDSCs via both the STAT3 and CREB signaling pathways [89]. MDSCs express both A2AR and A2BR on their surface and are thus regulated by adenosine in an autocrine manner. It has been demonstrated that an A2AR inhibitor reduced the accumulation of MDSCs in the TME, improved DC activation, and increased CD8+ T lymphocyte infiltration [55]. Administration of an A2BR agonist had increased tumor growth in melanoma-bearing mice that was associated with increased accumulation of CD11b+Gr1+ MDSCs in the TME as well as higher levels of IL-10 and MCP-1. Conversely, the pharmacological blockade of A2BR reversed this suppressive effect in the TME, leading to a significant melanoma growth delay [90,91].

2.7. CD4+ Regulatory T Cells

CD4+ Tregs function as immune suppressor cells that negatively regulate anti-tumor immunity in most cancers. Tregs regulate anti-tumor immune responses through multiple mechanisms, including the production of adenosine through co-expression of CD39/CD73, granzyme B, perforin, or Fas/FasL pathways, and the development of tolerogenic DCs, leading to the formation of a regulatory T cell subset in the TME [92,93]. Studies have shown that tumor-infiltrating Tregs with higher expression of CD39 and CD73 displayed stronger immunosuppressive function compared with those in the draining lymph nodes of tumor-bearing mice [1,94]. CD39+CD73+ Tregs increase their proliferation rate and immunosuppressive function in an autocrine manner [95,96]. Furthermore, adenosine-A2AR signaling also promotes the induction of Foxp3+Treg cells from the CD4+Foxp3 T cells that suppress effector T cell-mediated anti-tumor responses, promoting tumor progression [21,97]. In addition, tumor-derived adenosine also recruits CD4+ Treg cells to the TME, causing an immunosuppressive microenvironment. Tang et al. showed that binding tumor cell-derived adenosine to A2AR enhances CD73 transcription and upregulates chemokine CCL5 through activation of the p38/STAT1 pathway, recruiting Tregs to pancreatic tumors [97]. Adenosine produced by CD4+ Tregs suppresses the effector function of CD8+ and CD4+ T cells upon binding to corresponding A2AR on the effector cells. Another study showed that CD4+ Treg cells suppress the immune response through the increase of cAMP in target cells by the expression of COX-2 and the production of PGE2. The mechanisms responsible for Treg-mediated suppression involve the binding of adenosine and PGE2 produced by Tregs to their respective receptors expressed on T effector cells, leading to the up-regulation of adenylate cyclase and cAMP activities in T effector cells and to their functional inhibition [98,99]. These studies indicate that the presence of adenosine and PGE2 in the TME may synergistically mediate a powerful suppression of anti-tumor immunity, resulting in the progressive development of tumors.
Type 1 regulatory (Tr1) T cells are Foxp3 regulatory T cells, which are characterized by the predominant production of IL-10 and the expression of inhibitory receptors, such as LAG3 and CD49b, on the surface [100,101]. CD39+CD73+ Tr1 cells are found in the tumor microenvironment that facilitates the production of adenosine by hydrolyzing exogenous ATP/ADP [102]. A study showed that CD39+CD73+ Tr1 cells produce both adenosine and PGE2 in the TME, which promotes the development of head and neck squamous cell carcinoma. The inhibitory function of Tr1 was blocked by the usage of A2AR or EP2R antagonists (inhibitors of the PGE2 pathway), which confirmed that both adenosine and PGE2 were involved in Tr1-mediated immunosuppression in several solid tumors [99,103] (Figure 2).

3. Adenosine/Adenosine Receptor Blockade

3.1. Targeting CD39 or CD73

Due to the prominent suppressive role of adenosine in the TME that contributes to tumor development and progression, targeting adenosine pathways shows great promise in tumor immunotherapy [104,105,106]. Studies showed that inhibiting CD39 on macrophages in glioblastoma significantly increases their production of TNF and IL-12 while decreasing IL-10 secretion [107,108]. Meanwhile, in mouse melanoma and colorectal tumor models, TAMs and endothelial cells with high CD39 expression were effectively depleted following anti-CD39 treatment, thereby blocking angiogenesis [109]. CD39-specific antibodies suppressed the expression of CD39 mRNA and protein in murine colon adenocarcinoma, human breast cancer, and primary human T cells. This improved CD8+ T cell proliferation and substantially reduced the frequency of intratumoral Tregs [110].
The study on the mechanism of the anti-CD39 monoclonal antibody by Li et al. shows that anti-CD39 may bind to CD39 on intratumoral macrophages and monocytes expressing the P2X7 receptor, causing the release of eATP and triggering the activation of NALP3 inflammatory bodies. The downstream activation and release of IL-18 and IL-1β may promote the proliferation of CD8+ T cells and the effect function mediated by IFN-γ. It was revealed that the eATP-P2X7-ASC-NALP3 inflammatory body-IL18 pathway plays an important role in blocking the anti-tumor activity mediated by the CD39 enzyme, rather than simply reducing adenosine as the mechanism [1]. ATP degradation mediated by CD39 and CD73 eliminates the ability of apoptotic cells to recruit monocytes. ATP can induce a change in cell membrane permeability, resulting in the flow of Na+ and Ca+ into cells, which may result in growth inhibition [2]. Silva et al. demonstrated that activation of P2RX7 by eATP can promote metabolic adaptation and survival of the most persistent and functionally related memory CD8+ T cell population [111]. Drug blocking of CD39 prevented the degradation of eATP and enhanced macrophage phagocytosis of antibody-coated lymphoma cells in a P2X7 receptor-dependent manner [112].
Studies have shown that high expression of CD73 is associated with tumor development and a poor prognosis [97,113,114]. In glioblastoma, CD73 blockade was found to induce tumor cell apoptosis. Meanwhile, the population of Tregs, microglia, and macrophages was significantly reduced in the tumor microenvironment, but IL-6, CCL17, and CCL22 increased [113]. It was shown that CD39 and CD73 expression were significantly associated with poor survival in human PDAC samples and that the favorable prognostic effect associated with the presence of tumor-infiltrating CD8+ T cells was abolished. Although inhibition of CD39 or CD73 alone significantly slowed tumor growth in vivo [115,116], targeting these two nucleotidases showed significantly better anti-tumor activity [117]. Therefore, blocking the adenosine pathway may have double immunosuppressive effects: one is to promote the anti-tumor activity of effector T, NK, and other cells by blocking the accumulation of adenosine in the TME, and the other is to inhibit the proliferation of tumor cells by increasing eATP and providing essential sensor molecules to attract antigen-presenting cells to the tumor site. Several therapies targeting CD39 or CD73 have entered clinical trials (Table 1).
Interestingly, an increasing number of studies have shown that CD39 molecules, but not CD73 molecules, are considered co-inhibitory receptor molecules [118,119]. The conversion of ATP to ADP/AMP in the TME is regulated by CD39, leading to an increase in the AMP/ATP ratio and subsequent activation of AMPK [120]. AMPK serves as a central guardian for maintaining energy homeostasis by orchestrating diverse cellular processes, such as lipogenesis [121], glycolysis [122], the tricarboxylic acid cycle (TCA cycle) [123], cell cycle progression [124], and mitochondrial dynamics [125]. At the start, AMPK serves as a suppressor of tumors, potentially by working against the metabolic and signaling shifts that arise in cancer cells, such as heightened lipogenesis and the activation of mTORC1. In the event that tumorigenesis does take place, however, AMPK transitions to promoting the growth of the tumor, as it shields the tumor cells from the stresses that arise from their speedy proliferation [126]. A study has shown that in mouse models of colon cancer and fibrosarcoma, AMPK promotes lipid peroxidation by mediating phosphorylation of BECN1 and leads to ferroptosis of tumor cells [127]. In addition, in mouse melanomas, AMPK activates p38 MAPK, which inhibits PD-1 expression in Tregs by phosphorylating GSK-3β [128]. However, Cai Z et al. demonstrated that AMPK, activated in mouse metastasis models, drives pyruvate dehydrogenase complex (PDHc) activation to maintain the TCA cycle and promote breast cancer metastasis by adapting cancer cells to metabolic and oxidative stresses [123]. It is well documented that AMPK possesses double-edged sword characteristics in the context of tumor development and progression by modulating the inflammatory and metabolic pathways [129].
Otherwise, the CD39 molecule can also influence the level of eATP in the TME. Continuously, eATP continuously regulates the polarization of macrophages and the antigen presentation of DCs [1,119]. Therefore, the significant effects of CD39 may be due not only to its role as a rate-limiting enzyme in the adenosinergic pathway [19,119], but also to its indirect regulation of eATP or AMPK in the TME.

3.2. Targeting A2AR or A2BR

Adenosine plays a pro-tumor role in the TME mainly by interacting with A2AR on immune cells. Therefore, an increasing number of studies have focused on targeting A2AR. In the mouse model of chronic lymphoblastic leukemia, targeting A2AR had no effect on the size and weight of the tumor but saved the dysfunction of immune cells by reducing the accumulation of Tregs, restoring the expression of CD107a on T cells, and increasing the secretion of IL-2 and IFN-γ, indicating that anti-A2AR affects the function of immune cells rather than tumor cells [130,131,132]. Studies showed that targeting A2AR may enhance T cell activation and effector function in several murine cancer models, including MC38, CT26, RENCA, and B16. Notably, anti-A2AR could induce systemic anti-tumor immunity and increase memory formation that prevents tumor recurrence [133]. However, Festag et al. showed that T cell proliferation was inhibited by adenosine not through binding to adenosine receptors, but by intracellular downstream metabolites of adenosine, further blocking the synthesis of DNA, thereby inhibiting T cell proliferation and promoting T cell apoptosis in human T cells [134]. This may indicate that blocking CD39 or CD73 is more effective than blocking adenosine receptors. Adenosine inhibits T cell proliferation, activation, and apoptosis in different ways, but whether CD39 or CD73 blockers are superior to adenosine receptor blockers remains to be further verified [131,135,136].
Of the two major adenosine receptors responsible for immunosuppressive activity in the TME, A2BR has received much less attention than the high-affinity A2AR. A2AR is mainly expressed on the surface of T cells and NK cells. In contrast, A2BR is a low-affinity receptor mainly expressed on myeloid cells, including DCs, macrophages, and MDSCs, as well as cancer-associated fibroblasts (CAFs) [137]. A study showed that high expression of A2BR leads to worse outcomes in lung cancer patients. Mechanistically, inhibition of A2BR increases the glycolysis of DCs and promotes metabolic reprogramming of DCs to a more immunogenic state [138]. In a melanoma model, blocking A2BR stimulated T cell-mediated immunosurveillance by impairing the influx of MDSCs into the TME [139]. Due to the high concentration of adenosine in the TME, A2BR has a role that cannot be ignored, so the combination of A2AR and A2BR may provide a more comprehensive effect. However, there are currently only a few therapies that single-target CD39 or CD73 in clinical trials (Table 2).

3.3. Targeting CD38

As a bypass pathway for adenosine production, CD38 is the main NAD-degrading enzyme in several mammalian tissues. Studies have shown that CD38 is highly and uniformly expressed at the cell surface of multiple myeloma (MM) cells, and monoclonal antibodies against CD38 are highly efficacious in the treatment of MM [140,141,142]. In non-small-cell lung cancer, CXCR4 inhibitors can reduce the expression of CD73, CD38, and IL-10 on metastasis-initiating cells, thereby rescuing the cytotoxic activity of T cells and preventing TAM polarization, possibly by causing a decrease in adenosine and IL-10 production. This can effectively control the migration or invasion of lung tumor cells in vitro [143]. Interestingly, in HCC, patients with high CD38+CD68+ macrophage density had a better median overall survival of 34.43 months compared with 9.66 months in patients with low CD38+CD68+ macrophage density. CD38hi macrophages produce more IFN-γ and related cytokines, which may explain their predictive value when treated with immune checkpoint inhibitors [144]. CD38 has also been identified as a cell surface marker in hematologic cancers such as MM, but the effects of CD38 on different immune cells and other cancers are still being explored [142]. More than 100 therapies targeting CD38 are being investigated clinically, of which eight have entered phase 3 clinical trials (Table 3).

3.4. Combination Therapy

Cancer immunotherapy has made great progress and shown better efficacy than conventional chemical therapies for several malignancies in these decades. Immunotherapy, including immune checkpoint inhibitors and adoptive cell therapy, has demonstrated objective clinical responses in several cancers. For example, monoclonal antibodies against PD-1 showed satisfactory clinical advantages and are recommended for first- or second-line treatments in some chemical drug-resistant cancers [145,146,147]. Anti-tumor efficacy was significantly enhanced when used in combination with other immunotherapies, including the recovery of immune responses in models with incomplete responses to anti-PD-L1 or anti-CTLA-4 monotherapy [148].
Several studies have shown that the combination of anti-CD39 and anti-PD1 is more effective in the treatment of tumors than a single treatment in the MC38 tumor model. Furthermore, anti-CD39 can transform anti-PD1-resistant tumors into sensitive tumors, thereby transforming “cold” tumors into “hot” tumors. Mechanistically, anti-CD39 increases the proliferation of tumor-infiltrating lymphocytes, while anti-PD-1 may restore the dysfunctional phenotype of these lymphocytes [1,119,149]. The combinational blockade of CD73 and PD-1 also showed promising tumor suppression. The immunomodulatory mechanism of CD73 blockers is different from that of PD-1 blockers in a mouse colorectal cancer model. Anti-CD73 enhanced the anticancer function of immunosuppressive Tregs and depleted T cells, while PD-1 blockers quantitatively decreased Malat1high Treg and M2 macrophages. PD-1 blocking induces Treg deletion, and anti-CD73 therapy increases the activation of CD8+ T cells [137,150].
Anti-A2AR combined with either anti-PD-L1 or anti-CTLA-4 therapy showed efficacious tumor inhibition and led to up to 90% of tumors being eliminated in MC38 tumor-bearing mice that did not respond fully to anti-PD-L1 or anti-CTLA-4 monoclonal antibodies alone. Therefore, prioritizing strategies that offer combination therapy and target the adenosine pathway and immunotherapy in cancer is of great importance. Reportedly, more than 50 combination therapies targeting the ATP-adenosine pathway through CD73 or A2AR/A2BR antagonists are being explored clinically [151]. For example, several adenosine/adenosine receptor pathway blockers combined with PD-1 blockers have been used in clinical trials (Table 4).

4. Limitation and Prospect

A variety of metabolic regulation methods of adenosine levels have been developed, and several clinical studies have been carried out to evaluate the initial efficacy of new inhibitors of CD39, CD73, or A2aR signaling pathways in cancer therapy. It’s worth noting that blocking the adenosine/adenosine receptor pathway combined with immune checkpoint inhibitors such as anti-PD-1 or anti-CTLA-4 can significantly enhance the efficacy of anti-PD-1 or anti-CTLA-4, including in anti-PD-1-/anti-PD-L1-tolerant tumor types. In summary, the adenosine/adenosine receptor pathway is expected to be another important target for tumor immunotherapy (Figure 3).
The high-affinity A2AR adenosine signaling pathway in tumor tissues is the mainstream research direction because it is considered to effectively inhibit the immune response in tumor and normal tissues. However, it must be considered that other adenosine receptors may be more important; as mentioned earlier, the affinities of the adenosine receptors are different, and the level of extracellular adenosine depends on tissue, treatment, and intensity in time and space. In addition, it is important to consider that, as a whole, the TME is composed of tumor cells, immune cells, stromal cells, and metabolites. The other metabolites in the adenosinergic pathway, such as ATP, inosine, and AMPK, should be fully considered. Therefore, clarifying the specific mechanism of each adenosinergic pathway-related factor and the results of its combined action will provide strong theoretical support for us to combine adenosine and its receptor pathways and metabolic regulation in the future.

Author Contributions

Writing—original draft preparation, J.X. and J.W.; writing—review and editing, J.X., J.Z. and J.W.; supervision and funding acquisition, J.Z. and J.W. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Key Project of the Liaoning Provincial Department of Education (ZD2020001) and the National Natural Science Foundation of China (81972689).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Li, X.-Y.; Moesta, A.K.; Xiao, C.; Nakamura, K.; Casey, M.; Zhang, H.; Madore, J.; Lepletier, A.; Aguilera, A.R.; Sundarrajan, A.; et al. Targeting CD39 in Cancer Reveals an Extracellular ATP- and Inflammasome-Driven Tumor Immunity. Cancer Discov. 2019, 9, 1754–1773. [Google Scholar] [CrossRef] [PubMed]
  2. Bastid, J.; Cottalorda-Regairaz, A.; Alberici, G.; Bonnefoy, N.; Eliaou, J.F.; Bensussan, A. Entpd1/Cd39 Is a Promising Therapeutic Target in Oncology. Oncogene 2013, 32, 1743–1751. [Google Scholar] [CrossRef] [PubMed]
  3. Chini, E.N.; Chini, C.C.; Netto, J.M.E.; de Oliveira, G.C.; van Schooten, W. The Pharmacology of Cd38/Nadase: An Emerging Target in Cancer and Diseases of Aging. Trends Pharmacol. Sci. 2018, 39, 424–436. [Google Scholar] [CrossRef] [PubMed]
  4. Horenstein, A.L.; Chillemi, A.; Quarona, V.; Zito, A.; Roato, I.; Morandi, F.; Marimpietri, D.; Bolzoni, M.; Toscani, D.; Oldham, R.J.; et al. NAD+-Metabolizing Ectoenzymes in Remodeling Tumor–Host Interactions: The Human Myeloma Model. Cells 2015, 4, 520–537. [Google Scholar] [CrossRef] [PubMed]
  5. Vaisitti, T.; Audrito, V.; Serra, S.; Bologna, C.; Brusa, D.; Malavasi, F.; Deaglio, S. NAD+-metabolizing ecto-enzymes shape tumor-host interactions: The chronic lymphocytic leukemia model. FEBS Lett. 2011, 585, 1514–1520. [Google Scholar] [CrossRef]
  6. Azambuja, J.H.; Ludwig, N.; Braganhol, E.; Whiteside, T.L. Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity. Int. J. Mol. Sci. 2019, 20, 5698. [Google Scholar] [CrossRef]
  7. Kotulova, J.; Hajduch, M.; Dzubak, P. Current Adenosinergic Therapies: What Do Cancer Cells Stand to Gain and Lose? Int. J. Mol. Sci. 2021, 22, 12569. [Google Scholar] [CrossRef]
  8. Zhulai, G.; Oleinik, E.; Shibaev, M.; Ignatev, K. Adenosine-Metabolizing Enzymes, Adenosine Kinase and Adenosine Deaminase, in Cancer. Biomolecules 2022, 12, 418. [Google Scholar] [CrossRef] [PubMed]
  9. Park, J.; Gupta, R.S. Adenosine kinase and ribokinase—The RK family of proteins. Cell Mol. Life Sci. 2008, 65, 2875–2896. [Google Scholar] [CrossRef]
  10. Dutta, N.; Deb, I.; Sarzynska, J.; Lahiri, A. Inosine and Its Methyl Derivatives: Occurrence, Biogenesis, and Function in Rna. Prog. Biophys. Mol. Biol. 2022, 169–170, 21–52. [Google Scholar] [CrossRef]
  11. Gatsiou, A.; Vlachogiannis, N.; Lunella, F.F.; Sachse, M.; Stellos, K. Adenosine-to-Inosine RNA Editing in Health and Disease. Antioxid. Redox Signal. 2018, 29, 846–863. [Google Scholar] [CrossRef] [PubMed]
  12. Xia, C.; Yin, S.; To, K.K.W.; Fu, L. Cd39/Cd73/A2ar Pathway and Cancer Immunotherapy. Mol. Cancer 2023, 22, 44. [Google Scholar] [CrossRef] [PubMed]
  13. Ludwig, N.; Rubenich, D.S.; Zaręba, Ł.; Siewiera, J.; Pieper, J.; Braganhol, E.; Reichert, T.E.; Szczepański, M.J. Potential Roles of Tumor Cell- and Stroma Cell-Derived Small Extracellular Vesicles in Promoting a Pro-Angiogenic Tumor Microenvironment. Cancers 2020, 12, 3599. [Google Scholar] [CrossRef] [PubMed]
  14. Ludwig, N.; Yerneni, S.S.; Azambuja, J.H.; Gillespie, D.G.; Menshikova, E.V.; Jackson, E.K.; Whiteside, T.L. Tumor-derived exosomes promote angiogenesis via adenosine A2B receptor signaling. Angiogenesis 2020, 23, 599–610. [Google Scholar] [CrossRef]
  15. Clayton, A.; Al-Taei, S.; Webber, J.; Mason, M.D.; Tabi, Z. Cancer Exosomes Express CD39 and CD73, Which Suppress T Cells through Adenosine Production. J. Immunol. 2011, 187, 676–683. [Google Scholar] [CrossRef] [PubMed]
  16. Yegutkin, G.G.; Boison, D. ATP and Adenosine Metabolism in Cancer: Exploitation for Therapeutic Gain. Pharmacol. Rev. 2022, 74, 799–824. [Google Scholar] [CrossRef] [PubMed]
  17. Allard, B.; Allard, D.; Buisseret, L.; Stagg, J. The Adenosine Pathway in Immuno-Oncology. Nat. Rev. Clin. Oncol. 2020, 17, 611–629. [Google Scholar] [CrossRef] [PubMed]
  18. Beavis, P.A.; Stagg, J.; Darcy, P.K.; Smyth, M.J. CD73: A potent suppressor of antitumor immune responses. Trends Immunol. 2012, 33, 231–237. [Google Scholar] [CrossRef]
  19. Moesta, A.K.; Li, X.-Y.; Smyth, M.J. Targeting CD39 in cancer. Nat. Rev. Immunol. 2020, 20, 739–755. [Google Scholar] [CrossRef]
  20. De Marchi, E.; Orioli, E.; Pegoraro, A.; Sangaletti, S.; Portararo, P.; Curti, A.; Colombo, M.P.; Di Virgilio, F.; Adinolfi, E. The P2X7 receptor modulates immune cells infiltration, ectonucleotidases expression and extracellular ATP levels in the tumor microenvironment. Oncogene 2019, 38, 3636–3650. [Google Scholar] [CrossRef]
  21. Di Virgilio, F.; Sarti, A.C.; Falzoni, S.; De Marchi, E.; Adinolfi, E. Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat. Rev. Cancer 2018, 18, 601–618. [Google Scholar] [CrossRef] [PubMed]
  22. Blay, J.; White, T.D.; Hoskin, D.W. The Extracellular Fluid of Solid Carcinomas Contains Immunosuppressive Concentrations of Adenosine. Cancer Res. 1997, 57, 2602–2605. [Google Scholar] [PubMed]
  23. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol. Rev. 2017, 276, 121–144. [Google Scholar] [CrossRef] [PubMed]
  24. Allard, B.; Beavis, P.A.; Darcy, P.K.; Stagg, J. Immunosuppressive activities of adenosine in cancer. Curr. Opin. Pharmacol. 2016, 29, 7–16. [Google Scholar] [CrossRef] [PubMed]
  25. Samami, E.; Aleebrahim-Dehkordi, E.; Mohebalizadeh, M.; Yaribash, S.; Saghazadeh, A.; Rezaei, N. Inosine, Gut Microbiota, and Cancer Immunometabolism. Am. J. Physiol. Endocrinol. Metab. 2023, 324, E1–E8. [Google Scholar] [CrossRef] [PubMed]
  26. Liu, J.; Wang, F.; Zhang, Y.; Liu, J.; Zhao, B. ADAR1-Mediated RNA Editing and Its Role in Cancer. Front. Cell Dev. Biol. 2022, 10, 956649. [Google Scholar] [CrossRef]
  27. Dehnavi, S.; Kiani, A.; Sadeghi, M.; Biregani, A.F.; Banach, M.; Atkin, S.L.; Jamialahmadi, T.; Sahebkar, A. Targeting AMPK by Statins: A Potential Therapeutic Approach. Drugs 2021, 81, 923–933. [Google Scholar] [CrossRef]
  28. Guo, S.; Han, F.; Zhu, W. CD39—A bright target for cancer immunotherapy. BioMedicine 2022, 151, 113066. [Google Scholar] [CrossRef]
  29. Zhang, T.; Yu-Jing, L.; Ma, T. The immunomodulatory function of adenosine in sepsis. Front. Immunol. 2022, 13, 936547. [Google Scholar] [CrossRef]
  30. Borea, P.A.; Gessi, S.; Merighi, S.; Vincenzi, F.; Varani, K. Pharmacology of Adenosine Receptors: The State of the Art. Physiol. Rev. 2018, 98, 1591–1625. [Google Scholar] [CrossRef]
  31. Sun, C.; Wang, B.; Hao, S. Adenosine-A2A Receptor Pathway in Cancer Immunotherapy. Front. Immunol. 2022, 13, 837230. [Google Scholar] [CrossRef] [PubMed]
  32. Hinshaw, D.C.; Shevde, L.A. The tumor microenvironment innately modulates cancer progression. Cancer Res. 2019, 79, 4557–4566. [Google Scholar] [CrossRef] [PubMed]
  33. Lei, X.; Lei, Y.; Li, J.-K.; Du, W.-X.; Li, R.-G.; Yang, J.; Li, J.; Li, F.; Tan, H.-B. Immune cells within the tumor microenvironment: Biological functions and roles in cancer immunotherapy. Cancer Lett. 2020, 470, 126–133. [Google Scholar] [CrossRef] [PubMed]
  34. Guerra, L.; Bonetti, L.; Brenner, D. Metabolic Modulation of Immunity: A New Concept in Cancer Immunotherapy. Cell Rep. 2020, 32, 107848. [Google Scholar] [CrossRef] [PubMed]
  35. Sek, K.; Mølck, C.; Stewart, G.D.; Kats, L.; Darcy, P.K.; Beavis, P.A. Targeting Adenosine Receptor Signaling in Cancer Immunotherapy. Int. J. Mol. Sci. 2018, 19, 3837. [Google Scholar] [CrossRef] [PubMed]
  36. Borea, P.A.; Gessi, S.S.; Merighi, S.; Varani, K. Adenosine as a Multi-Signalling Guardian Angel in Human Diseases: When, Where and How Does It Exert Its Protective Effects? Trends Pharmacol. Sci. 2016, 37, 419–434. [Google Scholar] [CrossRef] [PubMed]
  37. Shimasaki, N.; Jain, A.; Campana, D. Nk Cells for Cancer Immunotherapy. Nat. Rev. Drug Discov. 2020, 19, 200–218. [Google Scholar] [CrossRef] [PubMed]
  38. Brauneck, F.; Seubert, E.; Wellbrock, J.; Wiesch, J.S.Z.; Duan, Y.; Magnus, T.; Bokemeyer, C.; Koch-Nolte, F.; Menzel, S.; Fiedler, W. Combined Blockade of Tigit and Cd39 or A2ar Enhances Nk-92 Cell-Mediated Cytotoxicity in Aml. Int. J. Mol. Sci. 2021, 22, 12919. [Google Scholar] [CrossRef]
  39. Young, A.; Ngiow, S.F.; Gao, Y.; Patch, A.-M.; Barkauskas, D.S.; Messaoudene, M.; Lin, G.; Coudert, J.D.; Stannard, K.A.; Zitvogel, L.; et al. A2ar Adenosine Signaling Suppresses Natural Killer Cell Maturation in the Tumor Microenvironment. Cancer Res. 2018, 78, 1003–1016. [Google Scholar] [CrossRef]
  40. Wang, J.; Matosevic, S. Adenosinergic signaling as a target for natural killer cell immunotherapy. J. Mol. Med. 2018, 96, 903–913. [Google Scholar] [CrossRef]
  41. Zhang, N.; Yang, D.; Dong, H.; Chen, Q.; Dimitrova, D.I.; Rogers, T.J.; Sitkovsky, M.; Oppenheim, J.J. Adenosine A2a Receptors Induce Heterologous Desensitization of Chemokine Receptors. Blood 2006, 108, 38–44. [Google Scholar] [CrossRef] [PubMed]
  42. Raskovalova, T.; Lokshin, A.; Huang, X.; Jackson, E.K.; Gorelik, E. Adenosine-Mediated Inhibition of Cytotoxic Activity and Cytokine Production by Il-2/Nkp46-Activated Nk Cells: Involvement of Protein Kinase a Isozyme I (Pka I). Immunol. Res. 2006, 36, 91–99. [Google Scholar] [CrossRef] [PubMed]
  43. Neo, S.Y.; Yang, Y.; Record, J.; Ma, R.; Chen, X.; Chen, Z.; Tobin, N.P.; Blake, E.; Seitz, C.; Thomas, R.; et al. Cd73 Immune Checkpoint Defines Regulatory Nk Cells within the Tumor Microenvironment. J. Clin. Investig. 2020, 130, 1185–1198. [Google Scholar] [CrossRef] [PubMed]
  44. Tong, L.; Jiménez-Cortegana, C.; Tay, A.H.; Wickström, S.; Galluzzi, L.; Lundqvist, A. NK cells and solid tumors: Therapeutic potential and persisting obstacles. Mol. Cancer 2022, 21, 206. [Google Scholar] [CrossRef] [PubMed]
  45. Lokshin, A.; Raskovalova, T.; Huang, X.; Zacharia, L.C.; Jackson, E.K.; Gorelik, E. Adenosine-Mediated Inhibition of the Cytotoxic Activity and Cytokine Production by Activated Natural Killer Cells. Cancer Res. 2006, 66, 7758–7765. [Google Scholar] [CrossRef] [PubMed]
  46. Raskovalova, T.; Huang, X.; Sitkovsky, M.; Zacharia, L.C.; Jackson, E.K.; Gorelik, E. Gs Protein-Coupled Adenosine Receptor Signaling and Lytic Function of Activated NK Cells. J. Immunol. 2005, 175, 4383–4391. [Google Scholar] [CrossRef] [PubMed]
  47. Chambers, A.M.; Lupo, K.B.; Wang, J.; Cao, J.; Utturkar, S.; Lanman, N.; Bernal-Crespo, V.; Jalal, S.; Pine, S.R.; Torregrosa-Allen, S.; et al. Engineered Natural Killer Cells Impede the Immunometabolic Cd73-Adenosine Axis in Solid Tumors. Elife 2022, 11, e73699. [Google Scholar] [CrossRef]
  48. Tay, A.H.M.; Prieto-Díaz, R.; Neo, S.; Tong, L.; Chen, X.; Carannante, V.; Önfelt, B.; Hartman, J.; Haglund, F.; Majellaro, M.; et al. A2B adenosine receptor antagonists rescue lymphocyte activity in adenosine-producing patient-derived cancer models. J. Immunother. Cancer 2022, 10, e004592. [Google Scholar] [CrossRef]
  49. 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]
  50. Cekic, C.; Sag, D.; Li, Y.; Theodorescu, D.; Strieter, R.M.; Linden, J. Adenosine A2B Receptor Blockade Slows Growth of Bladder and Breast Tumors. J. Immunol. 2012, 188, 198–205. [Google Scholar] [CrossRef]
  51. Novitskiy, S.V.; Ryzhov, S.; Zaynagetdinov, R.; Goldstein, A.E.; Huang, Y.; Tikhomirov, O.Y.; Blackburn, M.R.; Biaggioni, I.; Carbone, D.P.; Feoktistov, I.; et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 2008, 112, 1822–1831. [Google Scholar] [CrossRef] [PubMed]
  52. Borodovsky, A.; Barbon, C.M.; Wang, Y.; Ye, M.; Prickett, L.; Chandra, D.; Shaw, J.; Deng, N.; Sachsenmeier, K.; Clarke, J.D.; et al. Small Molecule Azd4635 Inhibitor of a(2a)R Signaling Rescues Immune Cell Function Including Cd103(+) Dendritic Cells Enhancing Anti-Tumor Immunity. J. Immunother. Cancer 2020, 8, e000417. [Google Scholar] [CrossRef] [PubMed]
  53. Lin, Y.-S.; Chiang, S.-F.; Chen, C.-Y.; Hong, W.-Z.; Chen, T.-W.; Chen, W.T.-L.; Ke, T.-W.; Yang, P.-C.; Liang, J.-A.; Shiau, A.; et al. Targeting CD73 increases therapeutic response to immunogenic chemotherapy by promoting dendritic cell maturation. Cancer Immunol. Immunother. 2023, 72, 2283–2297. [Google Scholar] [CrossRef] [PubMed]
  54. Ben Addi, A.; Lefort, A.; Hua, X.; Libert, F.; Communi, D.; Ledent, C.; Macours, P.; Tilley, S.L.; Boeynaems, J.-M.; Robaye, B. Modulation of murine dendritic cell function by adenine nucleotides and adenosine: Involvement of the A2B receptor. Eur. J. Immunol. 2008, 38, 1610–1620. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, Y.; Liu, Y.; Xu, D.; Zang, J.; Zheng, X.; Zhao, Y.; Li, Y.; He, R.; Ruan, S.; Dong, H.; et al. Targeting the Negative Feedback of Adenosine-A2AR Metabolic Pathway by a Tailored Nanoinhibitor for Photothermal Immunotherapy. Adv. Sci. 2022, 9, e2104182. [Google Scholar] [CrossRef] [PubMed]
  56. Borst, J.; Ahrends, T.; Bąbała, N.; Melief, C.J.M.; Kastenmüller, W. CD4+ T cell help in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2018, 18, 635–647. [Google Scholar] [CrossRef] [PubMed]
  57. Speiser, D.E.; Chijioke, O.; Schaeuble, K.; Munz, C. Cd4(+) T Cells in Cancer. Nat. Cancer 2023, 4, 317–329. [Google Scholar] [CrossRef] [PubMed]
  58. Bai, Y.; Zhang, X.; Zheng, J.; Liu, Z.; Yang, Z.; Zhang, X. Overcoming High Level Adenosine-Mediated Immunosuppression by Dzd2269, a Potent and Selective A2ar Antagonist. J. Exp. Clin. Cancer Res. 2022, 41, 302. [Google Scholar] [CrossRef]
  59. Kawano, M.; Takagi, R.; Tokano, M.; Matsushita, S. Adenosine Induces Il-31 Secretion by T-Helper 2 Cells: Implication for the Effect of Adenosine on Atopic Dermatitis and Its Therapeutic Strategy. Biochem. Biophys. Res. Commun. 2023, 645, 47–54. [Google Scholar] [CrossRef]
  60. Lee, J.; Shin, K.-O.; Kim, Y.; Cho, J.; Lim, H.W.; Yoon, S.-I.; Lee, G.-S.; Ko, H.-J.; Kim, P.-H.; Uchida, Y.; et al. Cathelicidin-Related Antimicrobial Peptide Regulates CD73 Expression in Mouse Th17 Cells via p38. Cells 2020, 9, 1561. [Google Scholar] [CrossRef]
  61. Chalmin, F.; Mignot, G.; Bruchard, M.; Chevriaux, A.; Végran, F.; Hichami, A.; Ladoire, S.; Derangère, V.; Vincent, J.; Masson, D.; et al. Stat3 and Gfi-1 Transcription Factors Control Th17 Cell Immunosuppressive Activity via the Regulation of Ectonucleotidase Expression. Immunity 2012, 36, 362–373. [Google Scholar] [CrossRef] [PubMed]
  62. Tokano, M.; Kawano, M.; Takagi, R.; Matsushita, S. Istradefylline, an Adenosine A2a Receptor Antagonist, Inhibits the Cd4(+) T-Cell Hypersecretion of Il-17a and Il-8 in Humans. Immunol. Med. 2022, 45, 244–250. [Google Scholar] [CrossRef] [PubMed]
  63. Wilson, J.M.; Kurtz, C.C.; Black, S.G.; Ross, W.G.; Alam, M.S.; Linden, J.; Ernst, P.B. The A2B Adenosine Receptor Promotes Th17 Differentiation via Stimulation of Dendritic Cell IL-6. J. Immunol. 2011, 186, 6746–6752. [Google Scholar] [CrossRef] [PubMed]
  64. Mastelic-Gavillet, B.; Rodrigo, B.N.; Décombaz, L.; Wang, H.; Ercolano, G.; Ahmed, R.; Lozano, L.E.; Ianaro, A.; Derré, L.; Valerio, M.; et al. Adenosine mediates functional and metabolic suppression of peripheral and tumor-infiltrating CD8+ T cells. J. Immunother. Cancer 2019, 7, 257. [Google Scholar] [CrossRef] [PubMed]
  65. Shi, L.; Feng, M.; Du, S.; Wei, X.; Song, H.; Yixin, X.; Song, J.; Wenxian, G. Adenosine Generated by Regulatory T Cells Induces CD8+T Cell Exhaustion in Gastric Cancer through A2aR Pathway. BioMed Res. Int. 2019, 2019, 4093214. [Google Scholar] [CrossRef] [PubMed]
  66. Heng, T.S.P.; The Immunological Genome Project Consortium; Painter, M.W.; Elpek, K.; Lukacs-Kornek, V.; Mauermann, N.; Turley, S.J.; Koller, D.; Kim, F.S.; Wagers, A.J.; et al. The Immunological Genome Project: Networks of gene expression in immune cells. Nat. Immunol. 2008, 9, 1091–1094. [Google Scholar] [CrossRef]
  67. Canale, F.P.; Ramello, M.C.; Núñez, N.; Furlan, C.L.A.; Bossio, S.N.; Serrán, M.G.; Boari, J.T.; del Castillo, A.; Ledesma, M.; Sedlik, C.; et al. CD39 Expression Defines Cell Exhaustion in Tumor-Infiltrating CD8+ T Cells. Cancer Res 2018, 78, 115–128. [Google Scholar] [CrossRef] [PubMed]
  68. Liao, J.; Zeng, D.-N.; Li, J.-Z.; Hua, Q.-M.; Xiao, Z.; He, C.; Mao, K.; Zhu, L.-Y.; Chu, Y.; Wen, W.-P.; et al. Targeting adenosinergic pathway enhances the anti-tumor efficacy of sorafenib in hepatocellular carcinoma. Hepatol. Int. 2020, 14, 80–95. [Google Scholar] [CrossRef]
  69. Briceño, P.; Rivas-Yañez, E.; Rosemblatt, M.V.; Parra-Tello, B.; Farías, P.; Vargas, L.; Simon, V.; Cárdenas, C.; Lladser, A.; Salazar-Onfray, F.; et al. CD73 Ectonucleotidase Restrains CD8+ T Cell Metabolic Fitness and Anti-tumoral Activity. Front. Cell Dev. Biol. 2021, 9, 638037. [Google Scholar] [CrossRef]
  70. Lai, Y.-P.; Kuo, L.-C.; Lin, B.-R.; Lin, H.-J.; Lin, C.-Y.; Chen, Y.-T.; Hsiao, P.-W.; Chang, H.-T.; Ko, P.C.-I.; Chen, H.-C.; et al. CD28 engagement inhibits CD73-mediated regulatory activity of CD8+ T cells. Commun. Biol. 2021, 4, 595. [Google Scholar] [CrossRef]
  71. Boutilier, A.J.; Elsawa, S.F. Macrophage Polarization States in the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 6995. [Google Scholar] [CrossRef] [PubMed]
  72. Pan, Y.; Yu, Y.; Wang, X.; Zhang, T. Tumor-Associated Macrophages in Tumor Immunity. Front. Immunol. 2020, 11, 583084. [Google Scholar] [CrossRef] [PubMed]
  73. Stout, R.D.; Suttles, J. Functional plasticity of macrophages: Reversible adaptation to changing microenvironments. J. Leukoc. Biol. 2004, 76, 509–513. [Google Scholar] [CrossRef] [PubMed]
  74. Xia, Y.; Rao, L.; Yao, H.; Wang, Z.; Ning, P.; Chen, X. Engineering Macrophages for Cancer Immunotherapy and Drug Delivery. Adv. Mater. 2020, 32, e2002054. [Google Scholar] [CrossRef] [PubMed]
  75. Mehla, K.; Singh, P.K. Metabolic Regulation of Macrophage Polarization in Cancer. Trends Cancer 2019, 5, 822–834. [Google Scholar] [CrossRef] [PubMed]
  76. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, J.; Wang, Y.; Chu, Y.; Li, Z.; Yu, X.; Huang, Z.; Xu, J.; Zheng, L. Tumor-derived adenosine promotes macrophage proliferation in human hepatocellular carcinoma. J. Hepatol. 2021, 74, 627–637. [Google Scholar] [CrossRef] [PubMed]
  78. Anderson, K.; Ryan, N.; Alkhimovitch, A.; Siddiqui, A.; Oghumu, S. Inhibition of Pi3k Isoform P110gamma Increases Both Anti-Tumor and Immunosuppressive Responses to Aggressive Murine Head and Neck Squamous Cell Carcinoma with Low Immunogenicity. Cancers 2021, 13, 953. [Google Scholar] [CrossRef]
  79. Kaneda, M.M.; Messer, K.S.; Ralainirina, N.; Li, H.; Leem, C.J.; Gorjestani, S.; Woo, G.; Nguyen, A.V.; Figueiredo, C.C.; Foubert, P.; et al. Pi3kgamma Is a Molecular Switch That Controls Immune Suppression. Nature 2016, 539, 437–442. [Google Scholar] [CrossRef]
  80. Bai, Y.; Zhang, X.; Zhou, J.; Guo, J.; Liu, Y.; Liang, C.; Wang, W.; Xing, Y.; Wu, J.; Hu, D. A2aR on lung adenocarcinoma cells: A novel target for cancer therapy via recruiting and regulating tumor-associated macrophages. Chem. Interact. 2023, 382, 110543. [Google Scholar] [CrossRef]
  81. Cekic, C.; Day, Y.-J.; Sag, D.; Linden, J. Myeloid Expression of Adenosine A2A Receptor Suppresses T and NK Cell Responses in the Solid Tumor Microenvironment. Cancer Res. 2014, 74, 7250–7259. [Google Scholar] [CrossRef] [PubMed]
  82. Montalban Del Barrio, I.; Penski, C.; Schlahsa, L.; Stein, R.G.; Diessner, J.; Wockel, A.; Dietl, J.; Lutz, M.B.; Mittelbronn, M.; Wischhusen, J.; et al. Adenosine-Generating Ovarian Cancer Cells Attract Myeloid Cells Which Differentiate into Adenosine-Generating Tumor Associated Macrophages—A Self-Amplifying, Cd39- and Cd73-Dependent Mechanism for Tumor Immune Escape. J. Immunother. Cancer 2016, 4, 49. [Google Scholar] [CrossRef]
  83. Jin, R.; Liu, L.; Xing, Y.; Meng, T.; Ma, L.; Pei, J.; Cong, Y.; Zhang, X.; Ren, Z.; Wang, X.; et al. Dual Mechanisms of Novel Cd73-Targeted Antibody and Antibody-Drug Conjugate in Inhibiting Lung Tumor Growth and Promoting Antitumor Immune-Effector Function. Mol. Cancer Ther. 2020, 19, 2340–2352. [Google Scholar] [CrossRef] [PubMed]
  84. Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef]
  85. Ryzhov, S.; Novitskiy, S.V.; Goldstein, A.E.; Biktasova, A.; Blackburn, M.R.; Biaggioni, I.; Dikov, M.M.; Feoktistov, I. Adenosinergic Regulation of the Expansion and Immunosuppressive Activity of CD11b+Gr1+ Cells. J. Immunol. 2011, 187, 6120–6129. [Google Scholar] [CrossRef]
  86. Groth, C.; Hu, X.; Weber, R.; Fleming, V.; Altevogt, P.; Utikal, J.; Umansky, V. Immunosuppression mediated by myeloid-derived suppressor cells (MDSCs) during tumour progression. Br. J. Cancer 2019, 120, 16–25. [Google Scholar] [CrossRef]
  87. King, R.J.; Shukla, S.K.; He, C.; Vernucci, E.; Thakur, R.; Attri, K.S.; Dasgupta, A.; Chaika, N.V.; Mulder, S.E.; Abrego, J.; et al. CD73 induces GM-CSF/MDSC-mediated suppression of T cells to accelerate pancreatic cancer pathogenesis. Oncogene 2022, 41, 971–982. [Google Scholar] [CrossRef] [PubMed]
  88. Chiu, D.K.; Tse, A.P.; Xu, I.M.; Di Cui, J.; Lai, R.K.; Li, L.L.; Koh, H.Y.; Tsang, F.H.; Wei, L.L.; Wong, C.M.; et al. Hypoxia Inducible Factor Hif-1 Promotes Myeloid-Derived Suppressor Cells Accumulation through Entpd2/Cd39l1 in Hepatocellular Carcinoma. Nat. Commun. 2017, 8, 517. [Google Scholar] [CrossRef]
  89. Sarkar, O.S.; Donninger, H.; Al Rayyan, N.; Chew, L.C.; Stamp, B.; Zhang, X.; Whitt, A.; Li, C.; Hall, M.; Mitchell, R.A.; et al. Monocytic MDSCs exhibit superior immune suppression via adenosine and depletion of adenosine improves efficacy of immunotherapy. Sci. Adv. 2023, 9, eadg3736. [Google Scholar] [CrossRef]
  90. Iannone, R.; Miele, L.; Maiolino, P.; Pinto, A.; Morello, S. Blockade of A2b Adenosine Receptor Reduces Tumor Growth and Immune Suppression Mediated by Myeloid-Derived Suppressor Cells in a Mouse Model of Melanoma. Neoplasia 2013, 15, 1400–1409. [Google Scholar] [CrossRef]
  91. Sorrentino, C.; Miele, L.; Porta, A.; Pinto, A.; Morello, S. Myeloid-derived suppressor cells contribute to A2B adenosine receptor-induced VEGF production and angiogenesis in a mouse melanoma model. Oncotarget 2015, 6, 27478–27489. [Google Scholar] [CrossRef] [PubMed]
  92. Whiteside, T.L. What Are Regulatory T Cells (Treg) Regulating in Cancer and Why? In Seminars in Cancer Biology; Academic Press: Cambridge, MA, USA, 2012; Volume 22, pp. 327–334. [Google Scholar]
  93. Ohue, Y.; Nishikawa, H. Regulatory T (Treg) Cells in Cancer: Can Treg Cells Be a New Therapeutic Target? Cancer Sci. 2019, 110, 2080–2089. [Google Scholar] [CrossRef] [PubMed]
  94. Kim, H.R.; Park, H.J.; Son, J.; Lee, J.G.; Chung, K.Y.; Cho, N.H.; Shim, H.S.; Park, S.; Kim, G.; Yoon, H.I.; et al. Tumor microenvironment dictates regulatory T cell phenotype: Upregulated immune checkpoints reinforce suppressive function. J. Immunother. Cancer 2019, 7, 339. [Google Scholar] [CrossRef] [PubMed]
  95. Beavis, P.A.; Henderson, M.A.; Giuffrida, L.; Mills, J.K.; Sek, K.; Cross, R.S.; Davenport, A.J.; John, L.B.; Mardiana, S.; Slaney, C.Y.; et al. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J. Clin. Investig. 2017, 127, 929–941. [Google Scholar] [CrossRef] [PubMed]
  96. Maj, T.; Wang, W.; Crespo, J.; Zhang, H.; Wang, W.; Wei, S.; Zhao, L.; Vatan, L.; Shao, I.; Szeliga, W.; et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat. Immunol. 2017, 18, 1332–1341. [Google Scholar] [CrossRef] [PubMed]
  97. Tang, T.; Huang, X.; Lu, M.; Zhang, G.; Han, X.; Liang, T. Transcriptional control of pancreatic cancer immunosuppression by metabolic enzyme CD73 in a tumor-autonomous and -autocrine manner. Nat. Commun. 2023, 14, 3364. [Google Scholar] [CrossRef]
  98. Su, Y.; Jackson, E.K.; Gorelik, E. Receptor desensitization and blockade of the suppressive effects of prostaglandin E2 and adenosine on the cytotoxic activity of human melanoma-infiltrating T lymphocytes. Cancer Immunol. Immunother. 2011, 60, 111–122. [Google Scholar] [CrossRef] [PubMed]
  99. Whiteside, T.L.; Jackson, E.K. Adenosine and Prostaglandin E2 Production by Human Inducible Regulatory T Cells in Health and Disease. Front. Immunol. 2013, 4, 212. [Google Scholar] [CrossRef]
  100. Apetoh, L.; Quintana, F.J.; Pot, C.; Joller, N.; Xiao, S.; Kumar, D.; Burns, E.J.; Sherr, D.H.; Weiner, H.L.; Kuchroo, V.K. The aryl hydrocarbon receptor interacts with c-Maf to promote the differentiation of type 1 regulatory T cells induced by IL-27. Nat. Immunol. 2010, 11, 854–861. [Google Scholar] [CrossRef]
  101. Chihara, N.; Madi, A.; Karwacz, K.; Awasthi, A.; Kuchroo, V.K. Differentiation and Characterization of Tr1 Cells. Curr. Protoc. Immunol. 2016, 113, 3.27.1–3.27.10. [Google Scholar] [CrossRef]
  102. Churov, A.; Zhulai, G. Targeting adenosine and regulatory T cells in cancer immunotherapy. Hum. Immunol. 2021, 82, 270–278. [Google Scholar] [CrossRef] [PubMed]
  103. Mandapathil, M.; Hilldorfer, B.; Szczepanski, M.J.; Czystowska, M.; Szajnik, M.; Ren, J.; Lang, S.; Jackson, E.K.; Gorelik, E.; Whiteside, T.L. Generation and Accumulation of Immunosuppressive Adenosine by Human CD4+CD25highFOXP3+ Regulatory T Cells. J. Biol. Chem. 2010, 285, 7176–7186. [Google Scholar] [CrossRef] [PubMed]
  104. Aroua, N.; Boet, E.; Ghisi, M.; Nicolau-Travers, M.L.; Saland, E.; Gwilliam, R.; de Toni, F.; Hosseini, M.; Mouchel, P.L.; Farge, T.; et al. Extracellular Atp and Cd39 Activate Camp-Mediated Mitochondrial Stress Response to Promote Cytarabine Resistance in Acute Myeloid Leukemia. Cancer Discov. 2020, 10, 1544–1565. [Google Scholar] [CrossRef] [PubMed]
  105. Feng, L.-L.; Cai, Y.-Q.; Zhu, M.-C.; Xing, L.-J.; Wang, X. The yin and yang functions of extracellular ATP and adenosine in tumor immunity. Cancer Cell Int. 2020, 20, 110. [Google Scholar] [CrossRef] [PubMed]
  106. Schakel, L.; Schmies, C.C.; Idris, R.M.; Luo, X.; Lee, S.Y.; Lopez, V.; Mirza, S.; Vu, T.H.; Pelletier, J.; Sevigny, J.; et al. Nucleotide Analog Arl67156 as a Lead Structure for the Development of Cd39 and Dual Cd39/Cd73 Ectonucleotidase Inhibitors. Front. Pharmacol. 2020, 11, 1294. [Google Scholar] [CrossRef] [PubMed]
  107. Savio, L.E.B.; de Andrade Mello, P.; Figliuolo, V.R.; de Avelar Almeida, T.F.; Santana, P.T.; Oliveira, S.D.S.; Silva, C.L.M.; Feldbrugge, L.; Csizmadia, E.; Minshall, R.D.; et al. Cd39 Limits P2x7 Receptor Inflammatory Signaling and Attenuates Sepsis-Induced Liver Injury. J. Hepatol. 2017, 67, 716–726. [Google Scholar] [CrossRef] [PubMed]
  108. Takenaka, M.C.; Gabriely, G.; Rothhammer, V.; Mascanfroni, I.D.; Wheeler, M.A.; Chao, C.-C.; Gutiérrez-Vázquez, C.; Kenison, J.; Tjon, E.C.; Barroso, A.; et al. Control of tumor-associated macrophages and T cells in glioblastoma via AHR and CD39. Nat. Neurosci. 2019, 22, 729–740. [Google Scholar] [CrossRef] [PubMed]
  109. Zhang, H.; Feng, L.; Mello, P.d.A.; Mao, C.; Near, R.; Csizmadia, E.; Chan, L.L.-Y.; Enjyoji, K.; Gao, W.; Zhao, H.; et al. Glycoengineered anti-CD39 promotes anticancer responses by depleting suppressive cells and inhibiting angiogenesis in tumor models. J. Clin. Investig. 2022, 132, e157431. [Google Scholar] [CrossRef]
  110. Kashyap, A.S.; Thelemann, T.; Klar, R.; Kallert, S.M.; Festag, J.; Buchi, M.; Hinterwimmer, L.; Schell, M.; Michel, S.; Jaschinski, F.; et al. Antisense oligonucleotide targeting CD39 improves anti-tumor T cell immunity. J. Immunother. Cancer 2019, 7, 67. [Google Scholar] [CrossRef]
  111. Da Silva, H.B.; Beura, L.K.; Wang, H.; Hanse, E.A.; Gore, R.; Scott, M.C.; Walsh, D.A.; Block, K.E.; Fonseca, R.; Yan, Y.; et al. The purinergic receptor P2RX7 directs metabolic fitness of long-lived memory CD8+ T cells. Nature 2018, 559, 264–268. [Google Scholar] [CrossRef]
  112. Casey, M.; Segawa, K.; Law, S.C.; Sabdia, M.B.; Nowlan, B.; Salik, B.; Lee, C.; Winterford, C.; Pearson, S.; Madore, J.; et al. Inhibition of CD39 unleashes macrophage antibody-dependent cellular phagocytosis against B-cell lymphoma. Leukemia 2023, 37, 379–387. [Google Scholar] [CrossRef] [PubMed]
  113. Azambuja, J.H.; Schuh, R.S.; Michels, L.R.; Iser, I.C.; Beckenkamp, L.R.; Roliano, G.G.; Lenz, G.S.; Scholl, J.N.; Sévigny, J.; Wink, M.R.; et al. Blockade of CD73 delays glioblastoma growth by modulating the immune environment. Cancer Immunol. Immunother. 2020, 69, 1801–1812. [Google Scholar] [CrossRef] [PubMed]
  114. Tripathi, A.; Lin, E.; Xie, W.; Flaifel, A.; Steinharter, J.A.; Gatof, E.N.S.; Bouchard, G.; Fleischer, J.H.; Martinez-Chanza, N.; Gray, C.; et al. Prognostic significance and immune correlates of CD73 expression in renal cell carcinoma. J. Immunother. Cancer 2020, 8, e001467. [Google Scholar] [CrossRef] [PubMed]
  115. Hammami, A.; Allard, D.; Allard, B.; Stagg, J. Targeting the adenosine pathway for cancer immunotherapy. Semin. Immunol. 2019, 42, 101304. [Google Scholar] [CrossRef] [PubMed]
  116. Battastini, A.M.O.; Figueiro, F.; Leal, D.B.R.; Doleski, P.H.; Schetinger, M.R.C. Cd39 and Cd73 as Promising Therapeutic Targets: What Could Be the Limitations? Front. Pharmacol. 2021, 12, 633603. [Google Scholar] [CrossRef] [PubMed]
  117. Jacoberger-Foissac, C.; Cousineau, I.; Bareche, Y.; Allard, D.; Chrobak, P.; Allard, B.; Pommey, S.; Messaoudi, N.; McNicoll, Y.; Soucy, G.; et al. Cd73 Inhibits Cgas-Sting and Cooperates with Cd39 to Promote Pancreatic Cancer. Cancer Immunol. Res. 2023, 22, 56–71. [Google Scholar] [CrossRef]
  118. Ferris, R.L.; Moskovitz, J.; Kunning, S.; Ruffin, A.T.; Reeder, C.; Ohr, J.; Gooding, W.E.; Kim, S.; Karlovits, B.J.; Vignali, D.A.; et al. Phase I Trial of Cetuximab, Radiotherapy, and Ipilimumab in Locally Advanced Head and Neck Cancer. Clin. Cancer Res. 2022, 28, 1335–1344. [Google Scholar] [CrossRef]
  119. Allard, D.; Allard, B.; Stagg, J. On the mechanism of anti-CD39 immune checkpoint therapy. J. Immunother. Cancer 2020, 8, e000186. [Google Scholar] [CrossRef]
  120. Keerthana, C.K.; Rayginia, T.P.; Shifana, S.C.; Anto, N.P.; Kalimuthu, K.; Isakov, N.; Anto, R.J. The role of AMPK in cancer metabolism and its impact on the immunomodulation of the tumor microenvironment. Front. Immunol. 2023, 14, 1114582. [Google Scholar] [CrossRef]
  121. Fullerton, M.D.; Galic, S.; Marcinko, K.; Sikkema, S.; Pulinilkunnil, T.; Chen, Z.-P.; O’Neill, H.M.; Ford, R.J.; Palanivel, R.; O’Brien, M.; et al. Single phosphorylation sites in Acc1 and Acc2 regulate lipid homeostasis and the insulin-sensitizing effects of metformin. Nat. Med. 2013, 19, 1649–1654. [Google Scholar] [CrossRef]
  122. Marsin, A.-S.; Bertrand, L.; Rider, M.H.; Deprez, J.; Beauloye, C.; Vincent, M.F.; Van den Berghe, G.; Carling, D.; Hue, L. Phosphorylation and activation of heart PFK-2 by AMPK has a role in the stimulation of glycolysis during ischaemia. Curr. Biol. 2000, 10, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  123. Cai, Z.; Li, C.-F.; Han, F.; Liu, C.; Zhang, A.; Hsu, C.-C.; Peng, D.; Zhang, X.; Jin, G.; Rezaeian, A.-H.; et al. Phosphorylation of PDHA by AMPK Drives TCA Cycle to Promote Cancer Metastasis. Mol. Cell 2020, 80, 263–278.e7. [Google Scholar] [CrossRef] [PubMed]
  124. Bodmer, D.; Levano-Huaman, S. Sesn2/Ampk/Mtor Signaling Mediates Balance between Survival and Apoptosis in Sensory Hair Cells under Stress. Cell Death Dis. 2017, 8, e3068. [Google Scholar] [CrossRef] [PubMed]
  125. Toyama, E.Q.; Herzig, S.; Courchet, J.; Lewis, T.L., Jr.; Loson, O.C.; Hellberg, K.; Young, N.P.; Chen, H.; Polleux, F.; Chan, D.C.; et al. Metabolism. Amp-Activated Protein Kinase Mediates Mitochondrial Fission in Response to Energy Stress. Science 2016, 351, 275–281. [Google Scholar] [CrossRef] [PubMed]
  126. Steinberg, G.R.; Hardie, D.G. New insights into activation and function of the AMPK. Nat. Rev. Mol. Cell Biol. 2023, 24, 255–272. [Google Scholar] [CrossRef] [PubMed]
  127. Song, X.; Zhu, S.; Chen, P.; Hou, W.; Wen, Q.; Liu, J.; Xie, Y.; Liu, J.; Klionsky, D.J.; Kroemer, G.; et al. Ampk-Mediated Becn1 Phosphorylation Promotes Ferroptosis by Directly Blocking System X(C)(-) Activity. Curr. Biol. 2018, 28, 2388–2399.e5. [Google Scholar] [CrossRef] [PubMed]
  128. Pokhrel, R.H.; Acharya, S.; Ahn, J.-H.; Gu, Y.; Pandit, M.; Kim, J.-O.; Park, Y.-Y.; Kang, B.; Ko, H.-J.; Chang, J.-H. AMPK promotes antitumor immunity by downregulating PD-1 in regulatory T cells via the HMGCR/p38 signaling pathway. Mol. Cancer 2021, 20, 133. [Google Scholar] [CrossRef]
  129. Hsu, C.-C.; Peng, D.; Cai, Z.; Lin, H.-K. AMPK signaling and its targeting in cancer progression and treatment. Semin. Cancer Biol. 2022, 85, 52–68. [Google Scholar] [CrossRef]
  130. Arruga, F.; Serra, S.; Vitale, N.; Guerra, G.; Papait, A.; Gyau, B.B.; Tito, F.; Efremov, D.; Vaisitti, T.; Deaglio, S. Targeting of the A2A adenosine receptor counteracts immunosuppression in vivo in a mouse model of chronic lymphocytic leukemia. Haematologica 2021, 106, 1343–1353. [Google Scholar] [CrossRef]
  131. Sidders, B.; Zhang, P.; Goodwin, K.; O’Connor, G.; Russell, D.L.; Borodovsky, A.; Armenia, J.; McEwen, R.; Linghu, B.; Bendell, J.C.; et al. Adenosine Signaling Is Prognostic for Cancer Outcome and Has Predictive Utility for Immunotherapeutic Response. Clin. Cancer Res. 2020, 26, 2176–2187. [Google Scholar] [CrossRef]
  132. Yang, R.; Elsaadi, S.; Misund, K.; Abdollahi, P.; Vandsemb, E.N.; Moen, S.H.; Kusnierczyk, A.; Slupphaug, G.; Standal, T.; Waage, A.; et al. Conversion of ATP to adenosine by CD39 and CD73 in multiple myeloma can be successfully targeted together with adenosine receptor A2A blockade. J. Immunother. Cancer 2020, 8, e000610. [Google Scholar] [CrossRef] [PubMed]
  133. Willingham, S.B.; Ho, P.Y.; Hotson, A.; Hill, C.M.; Piccione, E.C.; Hsieh, J.; Liu, L.; Buggy, J.J.; McCaffery, I.; Miller, R.A. A2AR Antagonism with CPI-444 Induces Antitumor Responses and Augments Efficacy to Anti–PD-(L)1 and Anti–CTLA-4 in Preclinical Models. Cancer Immunol. Res. 2018, 6, 1136–1149. [Google Scholar] [CrossRef] [PubMed]
  134. Festag, J.; Thelemann, T.; Schell, M.; Raith, S.; Michel, S.; Jaschinski, F.; Klar, R. Preventing Atp Degradation by Aso-Mediated Knockdown of Cd39 and Cd73 Results in A2ar-Independent Rescue of T Cell Proliferation. Mol. Ther. Nucleic Acids 2020, 21, 656–669. [Google Scholar] [CrossRef] [PubMed]
  135. Bareche, Y.; Pommey, S.; Carneiro, M.; Buisseret, L.; Cousineau, I.; Thebault, P.; Chrobak, P.; Communal, L.; Allard, D.; Robson, S.C.; et al. High-dimensional analysis of the adenosine pathway in high-grade serous ovarian cancer. J. Immunother. Cancer 2021, 9, e001965. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, J.; Zou, Y.; Du, B.; Li, W.; Yu, G.; Li, L.; Zhou, L.; Gu, X.; Song, S.; Liu, Y.; et al. Snp-Mediated Lncrna-Entpd3-As1 Upregulation Suppresses Renal Cell Carcinoma Via Mir-155/Hif-1alpha Signaling. Cell Death Dis. 2021, 12, 672. [Google Scholar] [CrossRef] [PubMed]
  137. Thompson, E.A.; Powell, J.D. Inhibition of the Adenosine Pathway to Potentiate Cancer Immunotherapy: Potential for Combinatorial Approaches. Annu. Rev. Med. 2021, 72, 331–348. [Google Scholar] [CrossRef]
  138. Guan, S.; Suman, S.; Amann, J.M.; Wu, R.; Carbone, D.P.; Wang, J.; Dikov, M.M. Metabolic reprogramming by adenosine antagonism and implications in non-small cell lung cancer therapy. Neoplasia 2022, 32, 100824. [Google Scholar] [CrossRef] [PubMed]
  139. Morello, S.; Miele, L. Targeting the adenosine A2b receptor in the tumor microenvironment overcomes local immunosuppression by myeloid-derived suppressor cells. OncoImmunology 2014, 3, e27989. [Google Scholar] [CrossRef]
  140. Lokhorst, H.M.; Plesner, T.; Laubach, J.P.; Nahi, H.; Gimsing, P.; Hansson, M.; Minnema, M.C.; Lassen, U.; Krejcik, J.; Palumbo, A.; et al. Targeting CD38 with Daratumumab Monotherapy in Multiple Myeloma. N. Engl. J. Med. 2015, 373, 1207–1219. [Google Scholar] [CrossRef]
  141. Krejcik, J.; Frerichs, K.A.; Nijhof, I.S.; van Kessel, B.; van Velzen, J.F.; Bloem, A.C.; Broekmans, M.E.; Zweegman, S.; van Meerloo, J.; Musters, R.J.; et al. Monocytes and Granulocytes Reduce CD38 Expression Levels on Myeloma Cells in Patients Treated with Daratumumab. Clin. Cancer Res. 2017, 23, 7498–7511. [Google Scholar] [CrossRef]
  142. Van de Donk, N.W.; Richardson, P.G.; Malavasi, F. Cd38 Antibodies in Multiple Myeloma: Back to the Future. Blood 2018, 131, 13–29. [Google Scholar] [CrossRef] [PubMed]
  143. Fortunato, O.; Belisario, D.C.; Compagno, M.; Giovinazzo, F.; Bracci, C.; Pastorino, U.; Horenstein, A.; Malavasi, F.; Ferracini, R.; Scala, S.; et al. CXCR4 Inhibition Counteracts Immunosuppressive Properties of Metastatic NSCLC Stem Cells. Front. Immunol. 2020, 11, 02168. [Google Scholar] [CrossRef] [PubMed]
  144. Ng, H.H.M.; Lee, R.Y.; Goh, S.; Tay, I.S.Y.; Lim, X.; Lee, B.; Chew, V.; Li, H.; Tan, B.; Lim, S.; et al. Immunohistochemical Scoring of Cd38 in the Tumor Microenvironment Predicts Responsiveness to Anti-Pd-1/Pd-L1 Immunotherapy in Hepatocellular Carcinoma. J. Immunother Cancer 2020, 8, 02168. [Google Scholar] [CrossRef] [PubMed]
  145. Tewari, K.S.; Monk, B.J.; Vergote, I.; Miller, A.; de Melo, A.C.; Kim, H.-S.; Kim, Y.M.; Lisyanskaya, A.; Samouëlian, V.; Lorusso, D.; et al. Protocol Investigators for, and Engot Protocol En-Cx. Survival with Cemiplimab in Recurrent Cervical Cancer. N. Engl. J. Med. 2022, 386, 544–555. [Google Scholar] [CrossRef] [PubMed]
  146. Boydell, E.; Sandoval, J.L.; Michielin, O.; Obeid, M.; Addeo, A.; Friedlaender, A. Neoadjuvant Immunotherapy: A Promising New Standard of Care. Int. J. Mol. Sci. 2023, 24, 11849. [Google Scholar] [CrossRef] [PubMed]
  147. Topalian, S.L.; Taube, J.M.; Pardoll, D.M. Neoadjuvant checkpoint blockade for cancer immunotherapy. Science 2020, 367, eaax0182. [Google Scholar] [CrossRef] [PubMed]
  148. Kamai, T.; Kijima, T.; Tsuzuki, T.; Nukui, A.; Abe, H.; Arai, K.; Yoshida, K.I. Increased Expression of Adenosine 2a Receptors in Metastatic Renal Cell Carcinoma Is Associated with Poorer Response to Anti-Vascular Endothelial Growth Factor Agents and Anti-Pd-1/Anti-Ctla4 Antibodies and Shorter Survival. Cancer Immunol. Immunother. 2021, 70, 2009–2021. [Google Scholar] [CrossRef] [PubMed]
  149. Perrot, I.; Michaud, H.A.; Giraudon-Paoli, M.; Augier, S.; Docquier, A.; Gros, L.; Courtois, R.; Dejou, C.; Jecko, D.; Becquart, O.; et al. Blocking Antibodies Targeting the Cd39/Cd73 Immunosuppressive Pathway Unleash Immune Responses in Combination Cancer Therapies. Cell Rep. 2019, 27, 2411–2425.e9. [Google Scholar] [CrossRef]
  150. Kim, M.; Min, Y.K.; Jang, J.; Park, H.; Lee, S.; Lee, C.H. Single-cell RNA sequencing reveals distinct cellular factors for response to immunotherapy targeting CD73 and PD-1 in colorectal cancer. J. Immunother. Cancer 2021, 9, e002503. [Google Scholar] [CrossRef]
  151. Liu, Y.; Li, Z.; Zhao, X.; Xiao, J.; Bi, J.; Li, X.-Y.; Chen, G.; Lu, L. Review immune response of targeting CD39 in cancer. Biomark. Res. 2023, 11, 63. [Google Scholar] [CrossRef]
Figure 1. Adenosine production and degradation. Adenosine is mainly formed by consecutive extracellular adenosine triphosphate (eATP) dephosphorylation catalyzed by CD39 and CD73. NAD+ is released by the salvage pathway and hydrolyzed by CD38 to form ADP ribose. This is further degraded to AMP through CD203a. Following this, CD73 dephosphorylates AMP to adenosine. Intracellular adenosine can be produced by hydrolyzing AMP through cytoplasmic 5′-nucleotidase-I (cN-I) or by hydrolyzing S-adenosine homocysteine (SAH) through SAH hydrolase. The bioavailability of adenosine depends on its conversion to inosine via adenosine deaminase (ADA), which comes in both intracellular and extracellular forms, and adenosine could be transported by ENTs (both directions) and CNTs (one-way transport). Once inside the cell, adenosine is phosphorylated to AMP by adenosine kinase (ADK) or degraded to inosine by ADA. In addition, tumor-derived exosomes (TEX) could carry CD39, CD73 molecules and adenosine secreted outside the cell. eATP, extracellular adenosine triphosphate; AMP, adenosine monophosphate; ADP, adenosine diphosphate; cN-I, cytoplasmic 5′-nucleotidase-I; SAH, S-adenosine homocysteine; ADA, adenosine deaminase; ADK, adenosine kinase; NAD+, nicotinamide adenine dinucleotide; ENTs, equilibrative nucleoside transporters; CNTs, concentrative nucleoside transporters; P, phosphate group (the different colors are used to distinguish between ATP, AMP, ADP, and adenosine).
Figure 1. Adenosine production and degradation. Adenosine is mainly formed by consecutive extracellular adenosine triphosphate (eATP) dephosphorylation catalyzed by CD39 and CD73. NAD+ is released by the salvage pathway and hydrolyzed by CD38 to form ADP ribose. This is further degraded to AMP through CD203a. Following this, CD73 dephosphorylates AMP to adenosine. Intracellular adenosine can be produced by hydrolyzing AMP through cytoplasmic 5′-nucleotidase-I (cN-I) or by hydrolyzing S-adenosine homocysteine (SAH) through SAH hydrolase. The bioavailability of adenosine depends on its conversion to inosine via adenosine deaminase (ADA), which comes in both intracellular and extracellular forms, and adenosine could be transported by ENTs (both directions) and CNTs (one-way transport). Once inside the cell, adenosine is phosphorylated to AMP by adenosine kinase (ADK) or degraded to inosine by ADA. In addition, tumor-derived exosomes (TEX) could carry CD39, CD73 molecules and adenosine secreted outside the cell. eATP, extracellular adenosine triphosphate; AMP, adenosine monophosphate; ADP, adenosine diphosphate; cN-I, cytoplasmic 5′-nucleotidase-I; SAH, S-adenosine homocysteine; ADA, adenosine deaminase; ADK, adenosine kinase; NAD+, nicotinamide adenine dinucleotide; ENTs, equilibrative nucleoside transporters; CNTs, concentrative nucleoside transporters; P, phosphate group (the different colors are used to distinguish between ATP, AMP, ADP, and adenosine).
Ijms 24 14928 g001
Figure 2. Adenosine plays an important role in tumor immunosuppression by interacting with different receptors of different immune cells. ↑, upregulate; ↓, downregulate; →, adenosine acts on different immune cells.
Figure 2. Adenosine plays an important role in tumor immunosuppression by interacting with different receptors of different immune cells. ↑, upregulate; ↓, downregulate; →, adenosine acts on different immune cells.
Ijms 24 14928 g002
Figure 3. Targeting adenosine/adenosine receptor pathways in tumor and immune cells. Adenosine/adenosine receptor blocking works collaboratively with immune checkpoint blockers like PD-1 and CTLA-4 inhibitors to promote anti-tumor immunity and inhibit pro-tumor immune responses through different mechanisms. ↑, upregulate; ↓, downregulate; dotted arrow, pro-tumor signals; red arrow, anti-tumor signals; dashed lines around cells, the schematic structure of the cell membrane.
Figure 3. Targeting adenosine/adenosine receptor pathways in tumor and immune cells. Adenosine/adenosine receptor blocking works collaboratively with immune checkpoint blockers like PD-1 and CTLA-4 inhibitors to promote anti-tumor immunity and inhibit pro-tumor immune responses through different mechanisms. ↑, upregulate; ↓, downregulate; dotted arrow, pro-tumor signals; red arrow, anti-tumor signals; dashed lines around cells, the schematic structure of the cell membrane.
Ijms 24 14928 g003
Table 1. Studies targeting CD39 or CD73 in cancer.
Table 1. Studies targeting CD39 or CD73 in cancer.
Study IDConditionsInterventionsPhaseStart Date
NCT04306900Solid Tumor, AdultCOMBINATION_PRODUCT: TTX-030, budigalimab and mFOLFOX6|COMBINATION_PRODUCT: TTX-030, budigalimab and docetaxel|COMBINATION_PRODUCT: TTX-030 and mFOLFOX6|COMBINATION_PRODUCT: TTX-030 and budigalimab|COMBINATION_PRODUCT: TTX-030, budigalimab, nab-paclitaxel and gemcitabine|COMBINATION_PRODUCT: TTX-030 and pembrolizumab|COMBINATION_PRODUCT: TTX-030, nab-paclitaxel and gemcitabine|COMBINATION_PRODUCT: Budigalimab and mFOLFOX6PHASE130 March 2020
NCT05374226Advanced Solid Tumors or LymphomasBIOLOGICAL: JS019PHASE131 March 2022
NCT05508373Advanced Solid TumorsBIOLOGICAL: JS019PHASE129 March 2022
NCT05075564Advanced Solid TumorDRUG: Part 1 ES002023|DRUG: Part 2 ES002023PHASE123 December 2021
NCT05381935Advanced Solid TumorDRUG: ES014|DRUG: ES014PHASE121 April 2023
NCT04668300Metastatic Angiosarcoma|Metastatic Dedifferentiated Liposarcoma|Metastatic Osteosarcoma|Recurrent Angiosarcoma|Recurrent Dedifferentiated Liposarcoma|Recurrent Osteosarcoma|Refractory Dedifferentiated Liposarcoma|Refractory OsteosarcomaBIOLOGICAL: Durvalumab|BIOLOGICAL: OleclumabPHASE226 November 2020
NCT04148937Advanced CancerDRUG: LY3475070|DRUG: PembrolizumabPHASE116 January 2020
NCT05174585Solid TumorBIOLOGICAL: JAB-BX102 (anti-CD73 monoclonal antibody)|BIOLOGICAL: pembrolizumab (anti-PD-1 monoclonal antibody)PHASE1|PHASE218 August 2022
NCT05329766Gastrointestinal Tract MalignanciesDRUG: Domvanalimab|DRUG: Quemliclustat|DRUG: Zimberelimab|DRUG: Fluorouracil|DRUG: Leucovorin|DRUG: OxaliplatinPHASE210 June 2022
NCT04797468Advanced Solid TumorDRUG: HLX23PHASE118 July 2022
NCT05688215Borderline Resectable Pancreatic Adenocarcinoma|Locally Advanced Pancreatic Ductal AdenocarcinomaPROCEDURE: Biospecimen Collection|PROCEDURE: Computed Tomography|PROCEDURE: Core Biopsy|DRUG: Fluorouracil|DRUG: Irinotecan|DRUG: Leucovorin|DRUG: Leucovorin Calcium|DRUG: Oxaliplatin|DRUG: Quemliclustat|DRUG: ZimberelimabPHASE1|PHASE27 March 2023
NCT04572152Advanced or Metastatic Solid TumorsBIOLOGICAL: AK119|BIOLOGICAL: AK104PHASE118 January 2021
NCT03954704Advanced Solid TumorsDRUG: Dalutrafusp alfa|DRUG: mFOLFOX6 Regimen|DRUG: dalutrafusp alfaPHASE13 June 2019
NCT04672434Metastatic Cancer|Solid TumorDRUG: Sym021|DRUG: Sym024PHASE119 November 2020
NCT03875573Luminal BDRUG: Durvalumab|RADIATION: Stereotactic Body Radiotherapy|DRUG: OleclumabPHASE26 November 2019
NCT03454451Non-Small-Cell Lung Cancer|Renal Cell Cancer|Colorectal Cancer|Triple Negative Breast Cancer|Cervical Cancer|Ovarian Cancer|Pancreatic Cancer|Endometrial Cancer|Sarcoma|Squamous Cell Carcinoma of the Head and Neck|Bladder Cancer|Metastatic Castration Resistant Prostate Cancer|Non-hodgkin LymphomaDRUG: CPI-006|DRUG: CPI-006 + ciforadenant|DRUG: CPI-006 + pembrolizumab|DRUG: CPI-006|DRUG: CPI-006 + ciforadenant|DRUG: CPI-006 + pembrolizumabPHASE125 April 2018
NCT05632328Advanced Pancreatic Ductal Adenocarcinoma|Pancreatic Ductal Adenocarcinoma|Pancreatic CancerDRUG: AGEN1423|DRUG: Balstilimab|DRUG: Gemcitabine|DRUG: Nab-paclitaxelPHASE223 April 2023
NCT05559541Solid Tumor, AdultDRUG: AK119|DRUG: AK104PHASE1|PHASE215 December 2022
NCT05689853Solid Tumor, AdultDRUG: AK119|DRUG: AK112PHASE1|PHASE214 April 2023
NCT04940286Borderline Resectable Pancreatic Adenocarcinoma|Resectable Pancreatic Adenocarcinoma|Stage IA Pancreatic Cancer AJCC v8|Stage IB Pancreatic Cancer AJCC v8|Stage IIA Pancreatic Cancer AJCC v8|Stage IIB Pancreatic Cancer AJCC v8BIOLOGICAL: Durvalumab|DRUG: Gemcitabine|DRUG: Nab-paclitaxel|BIOLOGICAL: OleclumabPHASE228 September 2021
NCT04660812Metastatic Colorectal CancerDRUG: AB680|DRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Bevacizumab|DRUG: m-FOLFOX-6 regimen|DRUG: RegorafenibPHASE1|PHASE210 May 2021
NCT05143970Metastatic Cancer|Metastatic Breast Cancer|Metastatic Pancreatic Cancer|Metastatic Gastric Cancer|Metastatic Lung Cancer|Metastatic Ovary Cancer|Oesophageal Cancer|Endometrial Cancer|Advanced Solid TumorDRUG: IPH5301 ALONE OR IN COMBINATION WITH CHEMOTHERAPY AND TRASTUZUMABPHASE121 January 2022
NCT05431270Metastatic Cancer|Refractory Cancer|Non-Small-Cell Lung Cancer|Pancreatic Adenocarcinoma|Pancreatic Neoplasms|Lung CancerDRUG: PT199|DRUG: Anti-PD-1 monoclonal antibodyPHASE123 June 2022
NCT05119998Solid TumorDRUG: IBI325 + sintilimab|DRUG: IBI325PHASE18 February 2022
NCT05246995Solid TumorDRUG: IBI325 + SintilimabPHASE123 March 2022
Source of information: ClinicalTrials.gov listings. All information is accessed on 2 September 2023.
Table 2. Studies targeting A2aR or A2bR in cancer.
Table 2. Studies targeting A2aR or A2bR in cancer.
Study IDConditionsInterventionsPhaseStart Date
NCT04969315Renal Cell Cancer|Castrate Resistant Prostate Cancer|Non-Small-Cell Lung CancerDRUG: TT-10PHASE1|PHASE21 May 2023
NCT04976660Colorectal Cancer|Gastric Cancer|Hepatocellular Carcinoma|Pancreatic CancerDRUG: TT-4PHASE1|PHASE215 December 2022
Source of information: ClinicalTrials.gov listings. All information is accessed on 2 September 2023.
Table 3. Studies targeting CD38 in cancer.
Table 3. Studies targeting CD38 in cancer.
Study IDConditionsInterventionsPhaseStart Date
NCT03319667Plasma Cell MyelomaDRUG: Isatuximab SAR650984|DRUG: Bortezomib|DRUG: Lenalidomide|DRUG: DexamethasonePHASE37 December 2017
NCT03275285Plasma Cell MyelomaDRUG: isatuximab SAR650984|DRUG: carfilzomib|DRUG: dexamethasonePHASE325 October 2017
NCT05461209Relapsed/Refractory Multiple MyelomaDRUG: Talquetamab|DRUG: Belantamab MafodotinPHASE320 October 2022
NCT05572515Relapsed or Refractory Multiple MyelomaDRUG: Teclistamab|DRUG: Pomalidomide|DRUG: Bortezomib|DRUG: Dexamethasone|DRUG: CarfilzomibPHASE329 March 2023
NCT04270409Plasma Cell MyelomaDRUG: Isatuximab SAR650984|DRUG: Lenalidomide|DRUG: DexamethasonePHASE316 June 2020
NCT02419118Multiple MyelomaDRUG: Daratumumab|DRUG: Lenalidomide|DRUG: DexamethasonePHASE2|PHASE315 January 2023
NCT02990338Plasma Cell MyelomaDRUG: Isatuximab|DRUG: Pomalidomide|DRUG: DexamethasonePHASE322 December 2016
NCT03937635Smoldering Plasma Cell MyelomaBIOLOGICAL: Daratumumab|DRUG: Dexamethasone|DRUG: Lenalidomide|OTHER: Quality-of-Life Assessment|OTHER: Questionnaire AdministrationPHASE316 September 2019
Source of information: ClinicalTrials.gov listings. All information is accessed on 2 September 2023.
Table 4. Studies targeting multiple sites in cancer.
Table 4. Studies targeting multiple sites in cancer.
Study IDConditionsInterventionsPhaseStart Date
NCT04306900Solid Tumor, AdultCOMBINATION_PRODUCT: TTX-030, budigalimab and mFOLFOX6|COMBINATION_PRODUCT: TTX-030, budigalimab and docetaxel|COMBINATION_PRODUCT: TTX-030 and mFOLFOX6|COMBINATION_PRODUCT: TTX-030 and budigalimab|COMBINATION_PRODUCT: TTX-030, budigalimab, nab-paclitaxel and gemcitabine|COMBINATION_PRODUCT: TTX-030 and pembrolizumab|COMBINATION_PRODUCT: TTX-030, nab-paclitaxel and gemcitabine|COMBINATION_PRODUCT: Budigalimab and mFOLFOX6PHASE130 March 2020
NCT03884556Solid Tumor|LymphomaDRUG: TTX-030|DRUG: Pembrolizumab|DRUG: Gemcitabine|DRUG: nab paclitaxelPHASE110 April 2019
NCT05177770Metastatic Castration-resistant Prostate Cancer|Prostate CancerDRUG: SRF617|DRUG: etrumadenant|DRUG: zimberelimabPHASE217 January 2022
NCT04381832Prostatic Neoplasms, Castration-Resistant|Androgen-Resistant Prostatic Neoplasms|Castration Resistant Prostatic Neoplasms|Prostatic Cancer, Castration-ResistantDRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Quemliclustat|DRUG: Enzalutamide|DRUG: Docetaxel|DRUG: SGPHASE1|PHASE27 July 2020
NCT05329766Gastrointestinal Tract MalignanciesDRUG: Domvanalimab|DRUG: Quemliclustat|DRUG: Zimberelimab|DRUG: Fluorouracil|DRUG: Leucovorin|DRUG: OxaliplatinPHASE210 June 2022
NCT05688215Borderline Resectable Pancreatic Adenocarcinoma|Locally Advanced Pancreatic Ductal AdenocarcinomaPROCEDURE: Biospecimen Collection|PROCEDURE: Computed Tomography|PROCEDURE: Core Biopsy|DRUG: Fluorouracil|DRUG: Irinotecan|DRUG: Leucovorin|DRUG: Leucovorin Calcium|DRUG: Oxaliplatin|DRUG: Quemliclustat|DRUG: ZimberelimabPHASE1|PHASE27 March 2023
NCT04660812Metastatic Colorectal CancerDRUG: AB680|DRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Bevacizumab|DRUG: m-FOLFOX-6 regimen|DRUG: RegorafenibPHASE1|PHASE210 May 2021
NCT04381832Prostatic Neoplasms, Castration-Resistant|Androgen-Resistant Prostatic Neoplasms|Castration Resistant Prostatic Neoplasms|Prostatic Cancer, Castration-ResistantDRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Quemliclustat|DRUG: Enzalutamide|DRUG: Docetaxel|DRUG: SGPHASE1|PHASE27 July 2020
NCT04660812Metastatic Colorectal CancerDRUG: AB680|DRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Bevacizumab|DRUG: m-FOLFOX-6 regimen|DRUG: RegorafenibPHASE1|PHASE210 May 2021
NCT05177770Metastatic Castration-resistant Prostate Cancer|Prostate CancerDRUG: SRF617|DRUG: etrumadenant|DRUG: zimberelimabPHASE217 January 2022
NCT04381832Prostatic Neoplasms, Castration-Resistant|Androgen-Resistant Prostatic Neoplasms|Castration Resistant Prostatic Neoplasms|Prostatic Cancer, Castration-ResistantDRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Quemliclustat|DRUG: Enzalutamide|DRUG: Docetaxel|DRUG: SGPHASE1|PHASE27 July 2020
NCT04660812Metastatic Colorectal CancerDRUG: AB680|DRUG: Etrumadenant|DRUG: Zimberelimab|DRUG: Bevacizumab|DRUG: m-FOLFOX-6 regimen|DRUG: RegorafenibPHASE1|PHASE210 May 2021
NCT05177770Metastatic Castration-resistant Prostate Cancer|Prostate CancerDRUG: SRF617|DRUG: etrumadenant|DRUG: zimberelimabPHASE217 January 2022
Source of information: ClinicalTrials.gov listings. All information is accessed on 2 September 2023.
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

Xing, J.; Zhang, J.; Wang, J. The Immune Regulatory Role of Adenosine in the Tumor Microenvironment. Int. J. Mol. Sci. 2023, 24, 14928. https://doi.org/10.3390/ijms241914928

AMA Style

Xing J, Zhang J, Wang J. The Immune Regulatory Role of Adenosine in the Tumor Microenvironment. International Journal of Molecular Sciences. 2023; 24(19):14928. https://doi.org/10.3390/ijms241914928

Chicago/Turabian Style

Xing, Jianlei, Jinhua Zhang, and Jinyan Wang. 2023. "The Immune Regulatory Role of Adenosine in the Tumor Microenvironment" International Journal of Molecular Sciences 24, no. 19: 14928. https://doi.org/10.3390/ijms241914928

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