Next Article in Journal
Tricellulin Effect on Paracellular Water Transport
Previous Article in Journal
Upgrading the Repertoire of miRNAs in Gastric Adenocarcinoma to Provide a New Resource for Biomarker Discovery
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity

by
Juliana Hofstätter Azambuja
1,2,3,
Nils Ludwig
2,3,
Elizandra Braganhol
1 and
Theresa L. Whiteside
2,3,4,*
1
The Postgraduate Biosciences Program, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre 90050-170, RS Brazil
2
Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
3
UPMC Hillman Cancer, Pittsburgh, PA 15213, USA
4
Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2019, 20(22), 5698; https://doi.org/10.3390/ijms20225698
Submission received: 14 October 2019 / Revised: 4 November 2019 / Accepted: 12 November 2019 / Published: 14 November 2019
(This article belongs to the Section Molecular Immunology)

Abstract

:
The adenosine pathway plays a key role in modulating immune responses in physiological and pathological conditions. Physiologically, anti-inflammatory effects of adenosine balance pro-inflammatory adenosine 5’-triphosphate (ATP), protecting tissues from damage caused by activated immune cells. Pathologically, increased adenosine monophosphatase (AMPase) activity in tumors leads to increased adenosine production, generating a deeply immunosuppressed microenvironment and promoting cancer progression. Adenosine emerges as a promising target for cancer therapy. It mediates protumor activities by inducing tumor cell proliferation, angiogenesis, chemoresistance, and migration/invasion by tumor cells. It also inhibits the functions of immune cells, promoting the formation of a tumor-permissive immune microenvironment and favoriting tumor escape from the host immune system. Pharmacologic inhibitors, siRNA or antibodies specific for the components of the adenosine pathway, or antagonists of adenosine receptors have shown efficacy in pre-clinical studies in various in vitro and in vivo tumor models and are entering the clinical arena. Inhibition of the adenosine pathway alone or in combination with classic immunotherapies offers a potentially effective therapeutic strategy in cancer.

Graphical Abstract

1. Introduction

The immune system plays an important role in the prevention as well as progression of malignancy [1]. On the one hand, it benefits the host by eliminating or neutralizing the tumor mass; on the other, it may be hijacked by the tumor to promote its progression. The recruitment of immune cells to the tumor suggests the presence of anti-tumor immunity. However, in the tumor microenvironment (TME), infiltrating immune cells become re-programmed by tumor-derived factors and assume a tumor-promoting phenotype [2].
The nucleoside adenosine (ADO) is involved in the regulation of diverse physiological and pathological processes [3,4,5,6,7,8,9]. ADO has recently emerged as a powerful immune checkpoint in the TME [10,11]. ADO acting directly on tumor cells promotes growth, survival, angiogenesis, chemoresistance, and metastasis [12,13]. However, under physiological conditions, ADO acting as an immuno-regulatory molecule protects normal tissues from inflammatory damage while in pathological conditions, it can impair anti-tumor immunity [14]. ADO attenuates functions of protective immune cells, including T cells and natural killer (NK) cells. It enhances the suppressive functions of regulatory T cells (Tregs), macrophages, and myeloid-derived suppressor cells (MDSCs), inducing cancer progression [15,16,17,18]. Here, we review approaches to blocking the adenosine pathway in cancer, removing the immune function brake and restoring the host’s ability to control tumor progression.

2. The ADO Pathway

Adenosine 5’-triphosphate (ATP) is a triphosphate nucleotide, and its main function is to provide energy to cells. For decades, this was the sole known ATP function. The history of the purinergic system began in 1929, when Drury and Szent-Gyorgi described the dilation of coronary blood vessels and hypotensive actions of purines in the heart and blood vessels [19]. Their findings contributed to establishing the current view of nucleotides and nucleosides as signaling molecules of the purinergic system. As such, they are involved in the regulation of various pathophysiological processes in the extracellular environments [20].
Purinergic signaling is characterized by the activity of extracellular purines (ATP, adenosine diphosphate (ADP), and ADO) or pyrimidines (uridine-5’-triphosphate (UTP) and uridine diphosphate (UDP)), which act as signaling molecules. Purinergic signaling serves as a communication system between cells. It is involved in a variety of mechanisms, including immune responses, inflammation, pain, platelet aggregation, proliferation, cell death, development, and neurotransmission [3,5,6,7,8,9,14,21,22]. Purines or pyrimidines exert their effects through interactions with specific membrane receptors called purinergic receptors or purinoceptors [5]. They comprise a group of enzymes that are involved in nucleotide degradation or nucleoside formation and, therefore, play a key role in the regulation of purinergic signaling [23].

2.1. Enzymes

The responses mediated by nucleotides and nucleosides upon binding to their respective receptor are catalyzed by enzymes called ectonucleotidases. These enzymes are responsible for the levels of nucleotides and nucleosides present in the extracellular microenvironment. Ectonucleotidases control the binding of nucleotides and nucleosides to their cellular receptors. As shows in Figure 1, this family of enzymes includes the ecto-nucleoside triphosphate-diphosphohydrolases (E-NTPDases), which catalyze the sequential degradation of ATP to ADP and adenosine monophosphate (AMP); the ecto-pyrophosphate-phosphodiesterases (E-NPP), which catalyze hydrolysis of ADP to AMP and of AMP to ADO; alkaline phosphatases (ALPs), which catalyze the degradation of ATP to ADP, ADP to AMP, and AMP to ADO; and, finally, the ecto-5’-nucleotidase (CD73), a 70-kDa glycosyl-phosphatidyl inositol-anchored protein on the plasma membrane. CD73 can be cleaved from the cell surface and act as a soluble enzyme, which irreversibly catalyzes the hydrolysis of AMP to ADO. In addition, adenosine deaminase (ADA) degrades ADO to inosine. The bioavailability of ADO is also regulated by nucleoside equilibrative transporters (ENTs) or concentrative nucleoside transporters (CNTs), which reside in the cell membrane and transport ADO into the cells. In the intracellular microenvironment, ADO is phosphorylated by ADO kinase (AdoK) and adenylate kinases into ADP [23].
However, ADO can also be generated by the non-canonical adenosinergic pathway by nicotinamide adenine dinucleotide (NAD+)-glycohydrolase/CD38 (NAD+ to ADP-ribose (ADPR)) and CD203a (PC-1) (ADPR to AMP) that is subsequently metabolized by CD73 to ADO. Therefore, CD73 represents the common link between the two adenosinergic pathways [24].

2.2. Receptors

The release of nucleotides and/or nucleosides into the extracellular microenvironment is accompanied by interactions with their respective receptors on cells, allowing for the purinergic signaling cascade to continue. Purinergic receptors are divided into two major groups, the P1R, which has the main endogenous agonist, ADO, and the P2R, which are sensitive to di- and triphosphate nucleosides, such as ATP, ADP, UTP, and UDP.
P1R, also called ADO receptors, are divided into four subtypes, A1, A2A, A2B, and A3, and all are coupled to G protein but differ in their affinity for ADO [25]. High-affinity receptors, A1R, A2AR, and A3R, bind ADO in the nanomolar range, whereas A2BR binds ADO in the micromolar range. Thus, at physiological concentrations of ADO, signaling is primarily mediated via A2AR, A1R, and A3R. A2BR are only activated when elevated levels of ADO are generated, such as in the inflammatory TME [26]. The A1R and A3R are coupled to the Gi or Go proteins, and their activation leads to a decrease in intracellular cyclic AMP (cAMP) levels, whereas A2AR and A2BR are coupled to Gs protein, resulting in increased levels of intracellular cAMP [25,27,28]. P1R are widely distributed among various cell types. They are expressed in the heart, lung, liver, testis, muscle, spinal cord, spleen, intestine, and brain [5]. In the immune system, these receptors are present in most cells and mediate the immunosuppressive and anti-inflammatory effects of ADO [18].
P2Rs comprise two categories of receptors, P2X and P2Y. P2YR are coupled to G protein and are metabotropic. P2XR are ionotropic and are divided into seven subtypes (P2X 1–7) that respond to ATP, whereas P2YR are subdivided into eight subtypes (P2Y 1, 2, 4, 6, 11–14) and are activated by ATP, ADP, UTP, and UDP, and are also sensitive to sugar nucleotides, such as UDP-glucose and UDP-galactose [29]. P2XR are broadly distributed in various cells, such as platelets, neurons, and muscle cells [30]. P2YR are found in a wide variety of organs and tissues: airway epithelium, different regions of the kidney, pancreas, adrenal gland, heart, vascular endothelium, skin, muscle, and various components of the nervous system, such as the cortex, hippocampus, and cerebellum [5].

3. ADO in Cancer

The role of ADO as a promoter of tumor progression is dependent on the activity and expression of CD73 in tumor cells. CD73 expression is elevated in different tumor types, including breast cancer [31], glioblastoma [32], F colorectal cancer [33], ovarian cancer [34], melanoma [34], gastric cancer [35], and bladder cancer [36]. Elevated CD73 expression levels significantly correlate with shorter overall survival in breast, ovarian, lung, and gastric cancer [37], and have been linked to cancer progression, migration, invasion, metastasis, chemoresistance, and neovascularization processes [13,38,39]. More importantly, ADO is now considered to be one of the most relevant immunosuppressive regulatory molecules in the TME [15,40,41]. Due to the favorable results seen in tumor models, targeting CD73 or ADORs has become a promising therapeutic approach in different types of human cancer. CD73 expression and ADO production by tumor cells have also been associated with the tumor progression, chemoresistance, migration, and angiogenesis, and these functions are summarized in Table 1, Table 2 and Table 3.

4. ADO in the Immune System

It has been reported that ATP, ADP, and ADO play a key role in modulating immune responses [14]. In normal conditions, ATP is found mostly in the cytoplasm at the concentration of 3 to 10 mM, whereas in the extracellular compartment, ATP levels are low, ranging from 1 to 10nM. Extracellular concentrations of ATP, as well as those of other nucleotides, may increase in response to different stimuli or conditions, such as cell lysis, hypoxia, or inflammation [30]. High concentrations of ATP in extracellular fluids can be interpreted as an indicator of tissue damage, which can trigger an inflammatory response characterized by the secretion of pro-inflammatory cytokines [81]. On the other hand, ADO, which is released by tumor cells or formed by hydrolysis of ATP, generally acts contrary to extracellular ATP [82]. ADO concentrations in homeostatic situations range from 10 to 200nM, whereas in stress situations, ADO levels may be as high as 10 to 100μM [13]. Increased extracellular ADO concentrations occur in situations of ischemia, hypoxia, epithelial-to-mesenchymal transition, cytotoxic stress, or trauma [83]. ADO mediates immunosuppressive responses for the protection of tissues adjacent to the excessive inflammation against attacks by defense cells [84]. At such sites, immune cells express P1 receptors and ectonucleotidases [14]. These opposite functions of ATP/ADO in the control of the immune response are illustrated in Figure 2.
In human peripheral blood, CD73 is expressed on approximately 90% of monocytes, 50% of CD8+ T cells, 10% of CD4+ T cells, 75% of Tregs, and 2% to 5% of NK cells. While in mice, CD73 is expressed on approximately 90% of monocytes, 30% of CD8+ T cells, 10% of CD4+ T cells, 1% to 5% of Tregs, and 50% of NK cells [11]. Thus, the distinct CD73 expression profiles between human and murine immune cells may lead to distinct cellular responses. Therefore, data obtained from preclinical models should be carefully considered before making plans for the therapeutic use of CD73 inhibitors.

4.1. ADO in Macrophages

The mononuclear phagocytic system is among the major targets of ADO, and phagocytic cells are highly susceptible to ADO effects [89]. Macrophages express all subtypes of ADO receptors, although A2AR and A2BR are expressed at higher levels compared to A1R and A3R [88].
Macrophages are classified as M1, which are classically activated, and M2, which mediate anti-inflammatory functions [89,90,91], as shown in Figure 3A.
P1R activation has been shown to suppress M1 activation and proinflammatory cytokines release [88,92,94,95,96]. In contrast, its activation induces M2 activation [85,92,98] with IL-10 [96] and vascular endothelial growth factor (VEGF) production [97]. ADO effects on macrophages are summarized in Figure 3B.
Notably, blocking CD73 activity with a specific CD73 inhibitor (adenosine 5’-α, β-methylene-diphosphate) enhanced the M1 phenotype, diminished IL-4 and IL-10 production, and promoted pro-inflammatory cytokine release [87].
We demonstrated in M1 macrophages that ATPase activity was decreased, while M2-type macrophages increased ATP/ADP/AMP hydrolysis through increased expression of CD39/CD73. This has led to rapid ADO accumulation without alteration in the purinergic receptor expression [99]. ADO generated by M2 macrophages is implicated in a decreased proliferation of CD4+ T lymphocytes [100]. We studied the purinergic signaling following co-incubation of primary mouse macrophages with a mouse glioblastoma cell line. We found that A2AR and P2X7R activation was necessary for IL-10, monocyte chemoattractant protein-1 (MCP-1), and IL-6 release by macrophages after interaction with glioblastoma. The related cytokines modulated conversion of macrophages to the M2-phenotype [101] and decreased the activity of ectonucleotidases [102]. In ovarian cancer, tumor cells use CD39/CD73 enzymes to control macrophage migration [100]. A2AR stimulation has direct myelosuppressive effects that indirectly contribute to suppression of T cells and NK cells in primary and metastatic melanoma microenvironments, indicating that the blocking of the A2AR has the potential to enhance immune killing of tumors [103,104].
Future studies should focus on the potential of blocking CD73/P1Rs to control the inflammatory microenvironment, macrophage phenotypes, and pro-tumor activities in cancer. Blocking ADO production may favor M1 (antitumor)-type polarization and may inhibit M2-type polarization. Only limited data exist on strategies that could control macrophage polarization and tumor progression.

4.2. ADO in Lymphocytes

4.2.1. CD4+ T Cells and NK

T cells can be activated as a result of antigen presentation by antigen-presenting cells (APCs). As a result, CD4+ T cells differentiate into T helper 1 (Th1) cells or T helper 2 (Th2) cells.
Human T lymphocytes were shown to express A1R, A2AR, A2BR, and A3R at different levels. A2AR were predominantly expressed and further upregulated upon stimulation [105]. Extracellular ADO inhibited T cell activation by APCs and modified T cell differentiation, cytokine production, and proliferation (Figure 4).
ADO signaling via A2AR negatively regulated the production of type 1 cytokines and enhanced the production of IL-10 [107,108,109] by cAMP/protein kinase A and caused Signal transducer and activator of transcription 5 (STAT5) dephosphorylation, which resulted in reduced IL-2R signaling in T cells [112] and inhibition of the nuclear factor kappa B (NF-kB) pathway [113]. This suggested that adenosinergic signaling via A2AR antagonism may be a promising target for activating anti-cancer immune responses. In addition, A2BR controls lymphocyte migration to the lymph nodes in mice, which facilitates the encounter of naive T cells with antigen presenting cells (APC) [114]. In this context, under activating conditions, human lymphocytes up-regulated A3 receptors [115], probably exerting protective effects against an immune attack.
A1R and A2R have the potential to regulate NK cell activity [116]. Human naive NK cells do not express significant levels of CD73 [117], but stimulated NK cells increased the expressions of A2AR, P2X7R, CD38, CD39, ENPP1, CD73, PANX1, and ENT1, and decreased ADA expression, suggesting that ADO is generated by canonical and non-canonical pathways after cellular activation [118]. The ADO effects on NK are presented in Figure 4. Mechanisms involve adenylyl cyclase, increased production of cAMP, and activation of PKA [75]. However, ADO also enhanced effector functions of NK cells in combination with IFN-alpha via A3R [119]. In addition, NK cells acted as regulatory cells, decreasing CD4+ T cell proliferation through ADO production via the ectoenzyme network, with a pivotal role for CD38 [120].
ADO suppressed metabolism and protein synthesis in NK cells, inhibiting oxidative phosphorylation and glycolysis [117]. Tumors utilize this strategy via the production of ADO to inhibit cytotoxic activity to NK cells and escape from the host immune system [121]. One strategy to circumvent NK cell suppression by the adenosinergic pathway would be to target A3R and/or A2AR because data show that A3R agonists and A2AR antagonists activate NK cells and further improve their anti-tumor effects in a melanoma mouse model in vivo [122,123].

4.2.2. T Regulatory Cells

Tregs (CD4+ Foxp3+ regulatory T cells) play a key role in maintaining the control immune responses in human health and disease. Treg co-express CD39/CD73 on the surface and generate extracellular ADO, contributing to immunosuppressive activities [124]. Recently, it has been shown that lymphocytes can also generate ADO in a non-canonical way through ectoenzymes CD38 (an NAD+ nucleosidase), NPP1, and CD73 [125].
The ADO effects on Treg are summarized in Figure 4. ADO mediated the suppression of Tregs by PGE2 receptors expressed on T cells, leading to the upregulation of adenylate cyclase and cAMP activities [126]. Tumors use the adenosinergic pathway by increasing ADO production to promote Treg activity, aiming at an immunosuppressive microenvironment to escape immune surveillance and promote cancer growth [127].

4.2.3. CD8+ T Cells

Tumors utilize ADO to suppress CD8+ T cell functions and to avoid tumor rejection. Otha et al. reported that in A2AR−/− tumor-bearing mice, tumor-infiltrating CD8+ T cells mediated tumor rejection [128]. Consistent with this, stimulation of cytotoxic T lymphocytes with a P1R agonist decreased cell proliferation, IFN-γ production, and cytotoxicity [110,111]. After A2AR stimulation, CD8+ T cells produced higher levels of cAMP and decreased IFN-γ and IL-2 expression, decreasing maturation of CD8+ T cells [110]. These data suggest that A2AR-antagonists should be considered in therapeutic protocols, since this approach may improve antitumor immunity and control tumor growth.

5. ADO in Exosomes

Exosomes are emerging as critical but poorly understood components of a complex communication network between the tumor and host cells. Exosomes are virus-sized extracellular vesicles deriving from the endocytic compartment of parent cells. They are secreted by normal and malignant cells and are present in all body fluids. Such molecules can serve as biomarkers of tumor progression and the immune response, or as predictors of response to therapies [129,130].
Exosomes derived from cancer cells were shown to carry CD39 and CD73 on their surface and exhibit potent ATP-AMP phosphohydrolytic activities. Multiple myeloma cell-derived exosomes release exosomes that are equipped with CD39/CD73 and with the enzymes that generate ADO via the non-canonical pathway (NAD+ /CD38/CD203a(PC-1)/CD73), and thus are able to generate ADO utilizing both the canonical and non-canonical pathways [131]. ADO produced by exosomes was shown to inhibit T- cell activation through A2A R [132]. The exosome-mediated increase of ADO production is not only driven by direct production of ADO by exosomes but also indirectly by inducing/upregulating the expression of ADO pathway components on recipient cells. Prostate cancer-derived exosomes induced CD73 expression on dendritic cells, which led to an inhibition of tumor necrosis factor-alpha (TNFα) and IL-12 production by T lymphocytes in an ATP-dependent manner [133]. In addition, exosomes released from head and neck squamous cell carcinoma cells expressed CD39 and CD73 and increased the ADO production in Treg cells [134]. Treg-derived exosomes were also shown to carry biologically active CD73, and their production of ADO was considered to play a regulatory role for this cell type, stimulating immunosuppressive functions [135]. Thus, tumor-derived exosomes are emerging as a new mechanism of cancer-driver immune suppression that involves the ADO pathway.

6. ADO Pathway in Cancer Therapy

6.1. Targeting ADO Receptors in Cancer Cells

6.1.1. A1R

The potential of targeting A1R for cancer treatment was mainly explored in breast cancer. In this context, estradiol, which has a critical role in breast cancer growth, increased A1R expression in breast cancer cell lines. Transfection with anti-A1R siRNA decreased cell proliferation, indicating that targeting the A1R receptor is beneficial in hormone-dependent breast cancer [136]. Furthermore, antagonism of A1R induced apoptosis of breast cancer cells by the upregulation of p53, caspase 3, 8, and 9 expression [50]. A1R antagonism also suppressed renal cell carcinoma growth/migration, induced apoptosis and cell cycle arrest in vitro, and reduced tumor volume in a nude mice subcutaneous model [137]. In addition, our data indicate that A1R is responsible for ADO-induced stimulation of gliomas proliferation in vitro [44]. However, the potential of A1R blockade still needs to be further explored, mainly in vivo in immunocompetent animal models, to prove its potential for future therapy in humans.

6.1.2. A2AR

Antagonism of A2AR is currently the most widely used pre-clinical approach to cancer therapy. The A2AR blockade impaired lung adenocarcinoma tumor cells’ growth in vitro and inhibited human tumor xenograft growth in mice [57]. The A2AR blockade also protected against tumor metastasis and enhanced NK cell functions in an in vivo melanoma model [69]. Inhibition of serine/threonine-protein kinase B-Raf (BRAF) and mitogen-activated protein kinase (MEK) in combination with A2AR provided a significant reduction of tumor progression and metastasis formation in a melanoma mouse model [58]. The A2AR-deficient mice showed an increased intratumoral CD8+ T cell frequency and number [138] and had increased frequencies of tumor-associated NK cells in melanoma models [122]. Pharmacological targeting of A2AR increased CAR T cell efficacy [139]. Pharmacological blockade of A2AR by selective antagonists decreased tumor growth in a head and neck squamous cell carcinoma mouse model by reducing the population of Tregs inside the tumor, spleen, blood, and lymph node, and enhanced the anti-tumor response of CD8+ T cells by increasing the release of IFN-γ [56]. Dual blockade of programmed cell death protein 1 (PD-1) and A2AR significantly enhanced the expression of IFN-γ and granzyme B by tumor-infiltrating CD8+ T cells and, accordingly, increased growth inhibition and the survival of mice in a breast cancer model [140] and increased the efficacy of a dendritic cell-based cancer vaccine by increasing the production of IFN-γ and reducing IL-10 [141]. This suggests the strong association between antitumor effects of the A2AR antagonist and the immune system, and the potential of its blockade for the restoration of effective anti-tumor immune responses.

6.1.3. A2BR

Selective antagonism of A2BR was shown to inhibit cell growth in prostate cancer cell lines [53]. Antagonizing A2BR increased colorectal cancer cell death in vitro via enhanced mitochondrial oxidative phosphorylation and reactive oxygen species (ROS) production [142]. A2BR antagonism in breast cancer also induced cell cycle arrest and apoptosis through the cyclin-, Bax/Bcl-2, and extracellular receptor kinase (ERK1/2) pathways [63]. In vivo, A2BR blockade also reversed immune suppression in the tumor microenvironment by reducing levels of IL-10, MCP-1, and MDSCs in melanoma lesions and increasing the frequency of CD8+ T lymphocytes and NK cells, and increasing the levels of Th1-like cytokines [42]. Enhancing anti-tumor immunity by inhibiting differentiation to Treg, which resulted in a suppression of metastasis in a melanoma mouse model in vivo, is another reported result of A2BR antagonism [59]. Cekic et al. reported that targeting A2BR the reduced growth of bladder and breast tumors in syngeneic mice, reduced metastases of breast cancer cells, increased tumor levels of IFN-γ, and enhanced dendritic cell activation, leading to an improved anti-tumor response [143].

6.1.4. A3R

As mentioned above, A3R is involved in control of tumor progression. The potential of A3R activation for cancer treatment has been explored. A3R mediated apoptosis in human bladder cancer cell lines via protein kinase C in vitro [144]. It also decreased cell growth and cell motility in breast cancer cell lines [70,145]. In addition, agonism of A3R enhanced TNF-related apoptosis-inducing ligand-mediated apoptosis (TRAIL) in thyroid carcinoma cells via NF-kB activation [146], induced cell death in glioblastoma by increasing Ca2+ and ROS, and downregulated ERK and AKT [61]. In vivo, agonism of the A3R reduced tumor growth and breast tumor-derived bone metastasis in a rat preclinical model [66], colon carcinoma [147].

6.2. CD73

Considering the tumor-promoting effects of ADO in the TME, the enzymatic activity of CD73 appears to be a promising target for the treatment of cancer. Three approaches of CD73 inhibition have been described: Neutralizing antibodies, siRNA, or pharmacologic inhibition.
Recently, we demonstrated the potential for CD73 blockade using either specific siRNA sequences or a pharmacological inhibitor in controlling glioblastoma progression in vitro and in an orthotopic immunocompetent in vivo model using Wistar rats. We showed that inhibition of CD73 with both technologies decreases the amount of ADO in the cerebrospinal fluid and correlates with decreased tumor volume and increased lymphocytic infiltrate [44]. Similar findings were reported in a breast cancer immunocompetent mice model [148]. In breast cancer, anti-CD73 monoclonal antibody (mAb) inhibited metastasis formation [149]. This strategy may also be combined with standard chemotherapy, since in vivo evidence in a mouse ovarian carcinoma model indicates that combining docetaxel with anti-CD73 antibody therapy is more effective than using both treatments individually [48]. Studies demonstrated synergism between anti-CD73 and anti-PD-1 or anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) antibodies in pre-clinical models. A selective inhibitor of CD73 enhanced anti-CTLA4 mAb efficiency by improving the anti-tumor immune response, and reduced melanoma growth in vivo [150]. However, in order to translate these technologies from the bench to the bedside, it is necessary to think about how to deliver these drugs to the tumor and avoid adverse effects. In this context, the strategy that is under consideration involves the application of nanotechnology. Zhi et al. showed that intravenous administration of CD73-siRNA-loaded nanoparticles led to reduced tumor growth and metastasis formation, with improved mice survival in a breast cancer model [151]. Jadidi-Niaragh et al. showed that CD73-siRNA encapsulated into chitosan-lactate nanoparticles suppressed CD73 expression in breast cancer cell lines and protected the oligonucleotides against serum and heparin degradation, suggesting that this system may increase siRNA performance when administered to patients by increasing the half-life of the sequences [152]. We demonstrated the potential of a cationic nanoemulsion delivering an siRNA anti-CD73 via the nasal for glioblastoma treatment in vitro and in vivo [79]. Nevertheless, the strategy of blocking CD73 requires a carefully strategy, because despite its prominent role, CD73 is not the only enzyme capable of producing ADO in the TME. Therefore, a strategy combining the blockade of CD73 enzymatic activity in combination with ADO antagonists may be of interest and may potentiate the antitumor effect of a therapy targeting the adenosinergic signaling pathway. In recent years, many small molecule drugs or antibodies targeting the adenosinergic pathway have undergone or are undergoing clinical trials. Their targets include CD73, A2AR, and A2BR. These findings are summarized in Table 4.

6.3. CD38

As cited above, ADO can also be generated by the non-canonical pathway. This pathway starts with CD38. Therefore, inhibition of the adenosinergic ectoenzyme function by the targeting of CD38 may lead to lower ADO levels in the TME. The first class of CD38-targeting antibody, daratumumab, is currently approved as a single agent and in combination with standards of care for the treatment of multiple myeloma [153]. Their antitumor effect is related to immunoregulatory activities. Daratumumab reduced the frequency of regulatory B cells, Tregs, and MDSCs, and enhanced helper and cytotoxic T cells [154].

6.4. Combined Therapies

6.4.1. Radiotherapy

Radiotherapy (RT)-induced damage leads to complex interactions between immune cells and tumors. RT has a dual effect, as it may activate innate and adaptive immune responses or increase immunosuppression by killing immune and tumor cells, inducing the accumulation of immunoregulatory mediators and cell types. Various observations from preclinical and clinical studies suggest that targeting radiation-induced immune deviation may offer promising opportunities for improvements of response to radiotherapy. In this context, the CD73-ADO axis system is currently seen as a major target [155]. However, the question about the effects of combined radiotherapy and anti-CD73/ADO antagonism is still open and awaits further studies.

6.4.2. Chemotherapy

As shown in Table 1, the CD73-ADO axis plays a role in cancer chemoresistance. It has been reported that the blockade of CD73 and ADOR enhances the effect of chemotherapy [43,45,48,49]. This indicates that inhibition of the adenosinergic pathway contributes to the enhancement of chemotherapy. It has also been elucidated that combination therapy with anti-CD73 antibodies enhances the response in vitro to vincristine [45], temozolomide [44], docetaxel [48], doxorubicin [49], TRAIL [46], and anti-human epidermal growth factor receptor 2 (HER2) antibody [47]. Moreover, a higher efficacy has been reported with A2BR antagonists [42]. The potential mechanisms associated with ADO-mediated chemoresistance are dependent on P1R activation and expression and the activity of multidrug resistance-associated protein (MRP1). However, additional in vivo studies are necessary to elucidate the real potential of the anti-CD73/ADOR antagonism to improve chemotherapy responses.

6.4.3. Immunotherapies

Checkpoint receptor blockade can lead to restoration of anti-cancer responses. However, not all patients respond to treatment, highlighting the need for further research to better understand tumor evasion mechanisms and to identify other targets that can effectively remove ‘brakes’ imposed on the immune response by the tumor. As described here, ADO is known to act through ADOR to negatively regulate T cells, macrophages, and NK cells responsible for anti-tumor responses in the TME, and thus targeting this pathway may prove to be a significant immunotherapeutic strategy. First, PD-1 blockade upregulates the expression of A2AR, making these cells more susceptible to ADO-mediated immunosuppression [140]. In this context, preclinical studies demonstrated a synergism between anti-CD73 and anti-PD-1 mAbs [156] or CTL-4 mAbs [150]. Synergism has also been observed between anti-PD-1 and A2AR antagonists [140,157], suggesting that combining CD73 and ADO as a therapeutic target might overcome immune system suppression in cancer and improve immunotherapeutic responses.

7. Conclusions

The CD73-ADO axis, among the currently investigated anti-tumor strategies, has gained much attention as a novel immune checkpoint for cancer therapy. Numerous preclinical studies showed that ADO mediates pro-tumor as well as immunosuppressive activities. The potential of CD73/ADOR inhibition for controlling tumor growth and metastasis formation and activation of the immune system increases interest in targeting the ADO pathway for cancer treatment. Several clinical trials involving antibodies/inhibitors targeting CD73 and antagonists/agonists targeting ADOR in cancer patients are currently ongoing.

Author Contributions

J.H.A. developed the concept and wrote the manuscript. N.L. was responsible for discussions and data interpretation; E.B. and T.L.W. edited the manuscript.

Funding

J.H.A. was supported by the Program PDSE 88881.188926/2018-01 from CAPES, Brazil. N.L. was supported by the Leopoldina Fellowship LPDS 2017-12 from German National Academy of Sciences Leopoldina. T.L.W. was partially supported by the NIH grant RO1 CA 168628.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ADAadenosine deaminase
ADOadenosine
AdoKAdenosine kinase
ADPadenosine diphosphate
ADPRADP-ribose
ALPsalkaline phosphatases
AMIDfactor-homologous mitochondrion-associated inducer of death
AMPadenosine monophosphate
APCsantigen presenting cells
ATPAdenosine 5’-triphosphate
cAMPintracellular cyclic AMP
CD39E-NTPDase1
CD73ecto-5’-nucleotidase
CD73 inhibitoradenosine 5’-α,β-methylene-diphosphate
CNTsconcentrative nucleoside transporters
CTLA-4anti- cytotoxic T-lymphocyte-associated protein 4
DCsdendritic cells
ECMextracellular matrix
EGFEpidermal growth factor
EGFREpidermal growth factor receptor
EMTEpithelial–mesenchymal transition
E-NPPecto-pyrophosphate-phosphodiesterases
ENTnucleoside equilibrative transporters
E-NTPDasesecto-nucleoside triphosphate-diphosphohydrolases
ErbB2Receptor Tyrosine Kinase 2
ERKextracellular signal-regulated kinase
GM-CSFGranulocyte-macrophage colony-stimulating factor
HIF-1alphaHypoxia-inducible factor 1-alpha
HNSCCHead and neck squamous cell carcinoma
IFNγInterferon gamma
IL-10Interleukin-10
IL-13Interleukin 13
IL-2Interleukin 2
IL-4Interleukin 4
IL-6Interleukin-6
IL-8Interleukin 8
JNKc-Jun N-terminal kinase
MCP-1monocyte chemoattractant protein-1
MDSCsmyeloid-derived suppressor cells
MIP-αHuman macrophage inflammatory protein alpha
NAD+adenine dinucleotide
NKNatural Killer
NOnitric oxide
NSCLCNon-small Cell Lung Cancer
PD-1programmed cell death protein 1
PIP3Phosphatidylinositol (3,4,5)-trisphosphate
Rap1Ras-proximate-1
ROSreactive oxygen species
siRNASmall interfering RNA
TGF-βTransforming growth factor beta
Th1T helper 1 cells
Th2helper 2 cells
TMEtumor microenvironment
TMZTemozolomide
TNF-αtumor necrosis factor-alpha
TRAILTNF-related apoptosis-inducing ligand mediated apoptosis
Tregregulatory T cell
UDPuridine diphosphate
UTPUridine-5’-triphosphate
VEGFEndothelial Growth Factor

References

  1. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
  2. Ladanyi, A.; Timar, J. Immunologic and immunogenomic aspects of tumor progression. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
  3. Burnstock, G. Purinergic Signalling and Endothelium. Curr. Vasc. Pharmacol. 2016, 14, 130–145. [Google Scholar] [CrossRef]
  4. Burnstock, G.; Di Virgilio, F. Purinergic signalling and cancer. Purinergic Signal. 2013, 9, 491–540. [Google Scholar] [CrossRef]
  5. Burnstock, G. Purinergic signalling: Past, present and future. Braz. J. Med. Biol. Res. 2009, 42, 3–8. [Google Scholar] [CrossRef]
  6. Burnstock, G. Purinergic Signalling and Neurological Diseases: An Update. CNS Neurol. Disord. Drug Targets 2017, 16, 257–265. [Google Scholar] [CrossRef]
  7. Burnstock, G. Purinergic Mechanisms and Pain. Adv. Pharmacol. 2016, 75, 91–137. [Google Scholar] [CrossRef]
  8. Burnstock, G. Purinergic Signaling in the Cardiovascular System. Circ. Res. 2017, 120, 207–228. [Google Scholar] [CrossRef]
  9. Burnstock, G.; Dale, N. Purinergic signalling during development and ageing. Purinergic Signal. 2015, 11, 277–305. [Google Scholar] [CrossRef]
  10. Passos, D.F.; Bernardes, V.M.; da Silva, J.L.G.; Schetinger, M.R.C.; Leal, D.B.R. Adenosine signaling and adenosine deaminase regulation of immune responses: Impact on the immunopathogenesis of HIV infection. Purinergic Signal. 2018, 14, 309–320. [Google Scholar] [CrossRef]
  11. Allard, D.; Chrobak, P.; Allard, B.; Messaoudi, N.; Stagg, J. Targeting the CD73-adenosine axis in immuno-oncology. Immunol. Lett. 2019, 205, 31–39. [Google Scholar] [CrossRef] [PubMed]
  12. Allard, B.; Turcotte, M.; Stagg, J. CD73-generated adenosine: Orchestrating the tumor-stroma interplay to promote cancer growth. J. Biomed. Biotechnol. 2012, 2012, 485156. [Google Scholar] [CrossRef] [PubMed]
  13. Young, A.; Mittal, D.; Stagg, J.; Smyth, M.J. Targeting cancer-derived adenosine: New therapeutic approaches. Cancer Discov. 2014, 4, 879–888. [Google Scholar] [CrossRef]
  14. Burnstock, G.; Boeynaems, J.M. Purinergic signalling and immune cells. Purinergic Signal. 2014, 10, 529–564. [Google Scholar] [CrossRef]
  15. Leone, R.D.; Emens, L.A. Targeting adenosine for cancer immunotherapy. J Immunother Cancer 2018, 6, 57. [Google Scholar] [CrossRef]
  16. Jin, D.; Fan, J.; Wang, L.; Thompson, L.F.; Liu, A.; Daniel, B.J.; Shin, T.; Curiel, T.J.; Zhang, B. CD73 on tumor cells impairs antitumor T-cell responses: A novel mechanism of tumor-induced immune suppression. Cancer Res. 2010, 70, 2245–2255. [Google Scholar] [CrossRef]
  17. Vigano, S.; Alatzoglou, D.; Irving, M.; Menetrier-Caux, C.; Caux, C.; Romero, P.; Coukos, G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front. Immunol. 2019, 10, 925. [Google Scholar] [CrossRef]
  18. Sek, K.; Molck, 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]
  19. Drury, A.N.; Szent-Gyorgyi, A. The physiological activity of adenine compounds with especial reference to their action upon the mammalian heart. J. Physiol. 1929, 68, 213–237. [Google Scholar] [CrossRef]
  20. Giuliani, A.L.; Sarti, A.C.; Di Virgilio, F. Extracellular nucleotides and nucleosides as signalling molecules. Immunol. Lett. 2019, 205, 16–24. [Google Scholar] [CrossRef]
  21. Burnstock, G. Purinergic signalling in the reproductive system in health and disease. Purinergic Signal. 2014, 10, 157–187. [Google Scholar] [CrossRef] [PubMed]
  22. Burnstock, G. Blood cells: An historical account of the roles of purinergic signalling. Purinergic Signal. 2015, 11, 411–434. [Google Scholar] [CrossRef] [PubMed]
  23. Zimmermann, H.; Zebisch, M.; Strater, N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal. 2012, 8, 437–502. [Google Scholar] [CrossRef] [PubMed]
  24. Ferretti, E.; Horenstein, A.L.; Canzonetta, C.; Costa, F.; Morandi, F. Canonical and non-canonical adenosinergic pathways. Immunol. Lett. 2019, 205, 25–30. [Google Scholar] [CrossRef]
  25. 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]
  26. Gessi, S.; Merighi, S.; Varani, K.; Borea, P.A. Adenosine receptors in health and disease. Adv. Pharmacol. 2011, 61, 41–75. [Google Scholar] [CrossRef]
  27. Borea, P.A.; Varani, K.; Vincenzi, F.; Baraldi, P.G.; Tabrizi, M.A.; Merighi, S.; Gessi, S. The A3 adenosine receptor: History and perspectives. Pharmacol. Rev. 2015, 67, 74–102. [Google Scholar] [CrossRef]
  28. Borea, P.A.; Dalpiaz, A.; Varani, K.; Gessi, S.; Gilli, G. Binding thermodynamics at A1 and A2A adenosine receptors. Life Sci. 1996, 59, 1373–1388. [Google Scholar] [CrossRef]
  29. Rayah, A.; Kanellopoulos, J.M.; Di Virgilio, F. P2 receptors and immunity. Microbes Infect. 2012, 14, 1254–1262. [Google Scholar] [CrossRef]
  30. Di Virgilio, F. P2X receptors and inflammation. Curr. Med. Chem. 2015, 22, 866–877. [Google Scholar] [CrossRef]
  31. Kruger, K.H.; Thompson, L.F.; Kaufmann, M.; Moller, P. Expression of ecto-5’-nucleotidase (CD73) in normal mammary gland and in breast carcinoma. Br. J. Cancer 1991, 63, 114–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Bavaresco, L.; Bernardi, A.; Braganhol, E.; Cappellari, A.R.; Rockenbach, L.; Farias, P.F.; Wink, M.R.; Delgado-Canedo, A.; Battastini, A.M. The role of ecto-5’-nucleotidase/CD73 in glioma cell line proliferation. Mol. Cell. Biochem. 2008, 319, 61–68. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, B.; Song, B.; Wang, X.; Chang, X.S.; Pang, T.; Zhang, X.; Yin, K.; Fang, G.E. The expression and clinical significance of CD73 molecule in human rectal adenocarcinoma. Tumour Biol. 2015, 36, 5459–5466. [Google Scholar] [CrossRef] [PubMed]
  34. Sadej, R.; Skladanowski, A.C. Dual, enzymatic and non-enzymatic, function of ecto-5’-nucleotidase (eN, CD73) in migration and invasion of A375 melanoma cells. Acta Biochim. Pol. 2012, 59, 647–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lu, X.X.; Chen, Y.T.; Feng, B.; Mao, X.B.; Yu, B.; Chu, X.Y. Expression and clinical significance of CD73 and hypoxia-inducible factor-1alpha in gastric carcinoma. World J. Gastroenterol. 2013, 19, 1912–1918. [Google Scholar] [CrossRef]
  36. Stella, J.; Bavaresco, L.; Braganhol, E.; Rockenbach, L.; Farias, P.F.; Wink, M.R.; Azambuja, A.A.; Barrios, C.H.; Morrone, F.B.; Oliveira Battastini, A.M. Differential ectonucleotidase expression in human bladder cancer cell lines. Urol. Oncol. 2010, 28, 260–267. [Google Scholar] [CrossRef]
  37. Jiang, T.; Xu, X.; Qiao, M.; Li, X.; Zhao, C.; Zhou, F.; Gao, G.; Wu, F.; Chen, X.; Su, C.; et al. Comprehensive evaluation of NT5E/CD73 expression and its prognostic significance in distinct types of cancers. BMC Cancer 2018, 18, 267. [Google Scholar] [CrossRef] [Green Version]
  38. Di Virgilio, F.; Adinolfi, E. Extracellular purines, purinergic receptors and tumor growth. Oncogene 2017, 36, 293–303. [Google Scholar] [CrossRef]
  39. Antonioli, L.; Novitskiy, S.V.; Sachsenmeier, K.F.; Fornai, M.; Blandizzi, C.; Hasko, G. Switching off CD73: A way to boost the activity of conventional and targeted antineoplastic therapies. Drug Discov. Today 2017, 22, 1686–1696. [Google Scholar] [CrossRef]
  40. 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]
  41. 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]
  42. 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] [PubMed] [Green Version]
  43. Yan, A.; Joachims, M.L.; Thompson, L.F.; Miller, A.D.; Canoll, P.D.; Bynoe, M.S. CD73 Promotes Glioblastoma Pathogenesis and Enhances Its Chemoresistance via A2B Adenosine Receptor Signaling. J. Neurosci. 2019, 39, 4387–4402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Azambuja, J.H.; Gelsleichter, N.E.; Beckenkamp, L.R.; Iser, I.C.; Fernandes, M.C.; Figueiro, F.; Battastini, A.M.O.; Scholl, J.N.; de Oliveira, F.H.; Spanevello, R.M.; et al. CD73 Downregulation Decreases In Vitro and In Vivo Glioblastoma Growth. Mol. Neurobiol. 2019, 56, 3260–3279. [Google Scholar] [CrossRef] [PubMed]
  45. Quezada, C.; Garrido, W.; Oyarzun, C.; Fernandez, K.; Segura, R.; Melo, R.; Casanello, P.; Sobrevia, L.; San Martin, R. 5’-ectonucleotidase mediates multiple-drug resistance in glioblastoma multiforme cells. J. Cell. Physiol. 2013, 228, 602–608. [Google Scholar] [CrossRef]
  46. Mikhailov, A.; Sokolovskaya, A.; Yegutkin, G.G.; Amdahl, H.; West, A.; Yagita, H.; Lahesmaa, R.; Thompson, L.F.; Jalkanen, S.; Blokhin, D.; et al. CD73 participates in cellular multiresistance program and protects against TRAIL-induced apoptosis. J. Immunol. 2008, 181, 464–475. [Google Scholar] [CrossRef] [Green Version]
  47. Turcotte, M.; Allard, D.; Mittal, D.; Bareche, Y.; Buisseret, L.; Jose, V.; Pommey, S.; Delisle, V.; Loi, S.; Joensuu, H.; et al. CD73 Promotes Resistance to HER2/ErbB2 Antibody Therapy. Cancer Res. 2017, 77, 5652–5663. [Google Scholar] [CrossRef]
  48. Li, H.; Lv, M.; Qiao, B.; Li, X. Blockade pf CD73/adenosine axis improves the therapeutic efficacy of docetaxel in epithelial ovarian cancer. Arch. Gynecol. Obstet. 2019, 299, 1737–1746. [Google Scholar] [CrossRef]
  49. Loi, S.; Pommey, S.; Haibe-Kains, B.; Beavis, P.A.; Darcy, P.K.; Smyth, M.J.; Stagg, J. CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 11091–11096. [Google Scholar] [CrossRef] [Green Version]
  50. Dastjerdi, M.N.; Rarani, M.Z.; Valiani, A.; Mahmoudieh, M. The effect of adenosine A1 receptor agonist and antagonist on p53 and caspase 3, 8, and 9 expression and apoptosis rate in MCF-7 breast cancer cell line. Res. Pharm. Sci. 2016, 11, 303–310. [Google Scholar] [CrossRef] [Green Version]
  51. Ma, D.F.; Kondo, T.; Nakazawa, T.; Niu, D.F.; Mochizuki, K.; Kawasaki, T.; Yamane, T.; Katoh, R. Hypoxia-inducible adenosine A2B receptor modulates proliferation of colon carcinoma cells. Hum. Pathol. 2010, 41, 1550–1557. [Google Scholar] [CrossRef] [PubMed]
  52. Vecchio, E.A.; Tan, C.Y.; Gregory, K.J.; Christopoulos, A.; White, P.J.; May, L.T. Ligand-Independent Adenosine A2B Receptor Constitutive Activity as a Promoter of Prostate Cancer Cell Proliferation. J. Pharmacol. Exp. Ther. 2016, 357, 36–44. [Google Scholar] [CrossRef] [PubMed]
  53. Wei, Q.; Costanzi, S.; Balasubramanian, R.; Gao, Z.G.; Jacobson, K.A. A2B adenosine receptor blockade inhibits growth of prostate cancer cells. Purinergic Signal. 2013, 9, 271–280. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kasama, H.; Sakamoto, Y.; Kasamatsu, A.; Okamoto, A.; Koyama, T.; Minakawa, Y.; Ogawara, K.; Yokoe, H.; Shiiba, M.; Tanzawa, H.; et al. Adenosine A2b receptor promotes progression of human oral cancer. BMC Cancer 2015, 15, 563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Koszalka, P.; Golunska, M.; Urban, A.; Stasilojc, G.; Stanislawowski, M.; Majewski, M.; Skladanowski, A.C.; Bigda, J. Specific Activation of A3, A2A and A1 Adenosine Receptors in CD73-Knockout Mice Affects B16F10 Melanoma Growth, Neovascularization, Angiogenesis and Macrophage Infiltration. PLoS ONE 2016, 11, e0151420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ma, S.R.; Deng, W.W.; Liu, J.F.; Mao, L.; Yu, G.T.; Bu, L.L.; Kulkarni, A.B.; Zhang, W.F.; Sun, Z.J. Blockade of adenosine A2A receptor enhances CD8(+) T cells response and decreases regulatory T cells in head and neck squamous cell carcinoma. Mol. Cancer 2017, 16, 99. [Google Scholar] [CrossRef]
  57. Mediavilla-Varela, M.; Luddy, K.; Noyes, D.; Khalil, F.K.; Neuger, A.M.; Soliman, H.; Antonia, S.J. Antagonism of adenosine A2A receptor expressed by lung adenocarcinoma tumor cells and cancer associated fibroblasts inhibits their growth. Cancer Biol. Ther. 2013, 14, 860–868. [Google Scholar] [CrossRef] [Green Version]
  58. Young, A.; Ngiow, S.F.; Madore, J.; Reinhardt, J.; Landsberg, J.; Chitsazan, A.; Rautela, J.; Bald, T.; Barkauskas, D.S.; Ahern, E.; et al. Targeting Adenosine in BRAF-Mutant Melanoma Reduces Tumor Growth and Metastasis. Cancer Res. 2017, 77, 4684–4696. [Google Scholar] [CrossRef] [Green Version]
  59. Kaji, W.; Tanaka, S.; Tsukimoto, M.; Kojima, S. Adenosine A(2B) receptor antagonist PSB603 suppresses tumor growth and metastasis by inhibiting induction of regulatory T cells. J. Toxicol. Sci. 2014, 39, 191–198. [Google Scholar] [CrossRef] [Green Version]
  60. Lee, E.J.; Min, H.Y.; Chung, H.J.; Park, E.J.; Shin, D.H.; Jeong, L.S.; Lee, S.K. A novel adenosine analog, thio-Cl-IB-MECA, induces G0/G1 cell cycle arrest and apoptosis in human promyelocytic leukemia HL-60 cells. Biochem. Pharmacol. 2005, 70, 918–924. [Google Scholar] [CrossRef]
  61. Kim, T.H.; Kim, Y.K.; Woo, J.S. The adenosine A3 receptor agonist Cl-IB-MECA induces cell death through Ca(2)(+)/ROS-dependent down regulation of ERK and Akt in A172 human glioma cells. Neurochem. Res. 2012, 37, 2667–2677. [Google Scholar] [CrossRef] [PubMed]
  62. Aghaei, M.; Panjehpour, M.; Karami-Tehrani, F.; Salami, S. Molecular mechanisms of A3 adenosine receptor-induced G1 cell cycle arrest and apoptosis in androgen-dependent and independent prostate cancer cell lines: Involvement of intrinsic pathway. J. Cancer Res. Clin. Oncol. 2011, 137, 1511–1523. [Google Scholar] [CrossRef] [PubMed]
  63. Jafari, S.M.; Joshaghani, H.R.; Panjehpour, M.; Aghaei, M. A2B adenosine receptor agonist induces cell cycle arrest and apoptosis in breast cancer stem cells via ERK1/2 phosphorylation. Cell. Oncol. (Dordr.) 2018, 41, 61–72. [Google Scholar] [CrossRef] [PubMed]
  64. Nagaya, H.; Gotoh, A.; Kanno, T.; Nishizaki, T. A3 adenosine receptor mediates apoptosis in in vitro RCC4-VHL human renal cancer cells by up-regulating AMID expression. J. Urol. 2013, 189, 321–328. [Google Scholar] [CrossRef]
  65. Kanno, T.; Nakano, T.; Fujita, Y.; Gotoh, A.; Nishizaki, T. Adenosine induces apoptosis in SBC-3 human lung cancer cells through A(3) adenosine receptor-dependent AMID upregulation. Cell. Physiol. Biochem. 2012, 30, 666–677. [Google Scholar] [CrossRef]
  66. Varani, K.; Vincenzi, F.; Targa, M.; Paradiso, B.; Parrilli, A.; Fini, M.; Lanza, G.; Borea, P.A. The stimulation of A(3) adenosine receptors reduces bone-residing breast cancer in a rat preclinical model. Eur. J. Cancer 2013, 49, 482–491. [Google Scholar] [CrossRef]
  67. Cappellari, A.R.; Pillat, M.M.; Souza, H.D.; Dietrich, F.; Oliveira, F.H.; Figueiro, F.; Abujamra, A.L.; Roesler, R.; Lecka, J.; Sevigny, J.; et al. Ecto-5’-Nucleotidase Overexpression Reduces Tumor Growth in a Xenograph Medulloblastoma Model. PLoS ONE 2015, 10, e0140996. [Google Scholar] [CrossRef]
  68. Merighi, S.; Simioni, C.; Gessi, S.; Varani, K.; Mirandola, P.; Tabrizi, M.A.; Baraldi, P.G.; Borea, P.A. A(2B) and A(3) adenosine receptors modulate vascular endothelial growth factor and interleukin-8 expression in human melanoma cells treated with etoposide and doxorubicin. Neoplasia 2009, 11, 1064–1073. [Google Scholar] [CrossRef] [Green Version]
  69. Beavis, P.A.; Divisekera, U.; Paget, C.; Chow, M.T.; John, L.B.; Devaud, C.; Dwyer, K.; Stagg, J.; Smyth, M.J.; Darcy, P.K. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc. Natl. Acad. Sci. USA 2013, 110, 14711–14716. [Google Scholar] [CrossRef] [Green Version]
  70. Ledderose, C.; Hefti, M.M.; Chen, Y.; Bao, Y.; Seier, T.; Li, L.; Woehrle, T.; Zhang, J.; Junger, W.G. Adenosine arrests breast cancer cell motility by A3 receptor stimulation. Purinergic Signal. 2016, 12, 673–685. [Google Scholar] [CrossRef] [Green Version]
  71. Merighi, S.; Benini, A.; Mirandola, P.; Gessi, S.; Varani, K.; Simioni, C.; Leung, E.; Maclennan, S.; Baraldi, P.G.; Borea, P.A. Caffeine inhibits adenosine-induced accumulation of hypoxia-inducible factor-1alpha, vascular endothelial growth factor, and interleukin-8 expression in hypoxic human colon cancer cells. Mol. Pharmacol. 2007, 72, 395–406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Martinez-Ramirez, A.S.; Diaz-Munoz, M.; Battastini, A.M.; Campos-Contreras, A.; Olvera, A.; Bergamin, L.; Glaser, T.; Jacintho Moritz, C.E.; Ulrich, H.; Vazquez-Cuevas, F.G. Cellular Migration Ability Is Modulated by Extracellular Purines in Ovarian Carcinoma SKOV-3 Cells. J. Cell. Biochem. 2017, 118, 4468–4478. [Google Scholar] [CrossRef] [PubMed]
  73. Zhou, P.; Zhi, X.; Zhou, T.; Chen, S.; Li, X.; Wang, L.; Yin, L.; Shao, Z.; Ou, Z. Overexpression of Ecto-5’-nucleotidase (CD73) promotes T-47D human breast cancer cells invasion and adhesion to extracellular matrix. Cancer Biol. Ther. 2007, 6, 426–431. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Stagg, J.; Divisekera, U.; McLaughlin, N.; Sharkey, J.; Pommey, S.; Denoyer, D.; Dwyer, K.M.; Smyth, M.J. Anti-CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2010, 107, 1547–1552. [Google Scholar] [CrossRef] [Green Version]
  75. 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]
  76. Koszalka, P.; Pryszlak, A.; Golunska, M.; Kolasa, J.; Stasilojc, G.; Skladanowski, A.C.; Bigda, J.J. Inhibition of CD73 stimulates the migration and invasion of B16F10 melanoma cells in vitro, but results in impaired angiogenesis and reduced melanoma growth in vivo. Oncol. Rep. 2014, 31, 819–827. [Google Scholar] [CrossRef] [Green Version]
  77. Qiao, Z.; Li, X.; Kang, N.; Yang, Y.; Chen, C.; Wu, T.; Zhao, M.; Liu, Y.; Ji, X. A Novel Specific Anti-CD73 Antibody Inhibits Triple-Negative Breast Cancer Cell Motility by Regulating Autophagy. Int. J. Mol. Sci. 2019, 20, 1057. [Google Scholar] [CrossRef] [Green Version]
  78. Ma, X.L.; Shen, M.N.; Hu, B.; Wang, B.L.; Yang, W.J.; Lv, L.H.; Wang, H.; Zhou, Y.; Jin, A.L.; Sun, Y.F.; et al. CD73 promotes hepatocellular carcinoma progression and metastasis via activating PI3K/AKT signaling by inducing Rap1-mediated membrane localization of P110beta and predicts poor prognosis. J. Hematol. Oncol. 2019, 12, 37. [Google Scholar] [CrossRef]
  79. Azambuja, J.H.; Schuh, R.S.; Michels, L.R.; Gelsleichter, N.E.; Beckenkamp, L.R.; Iser, I.C.; Lenz, G.S.; de Oliveira, F.H.; Venturin, G.; Greggio, S.; et al. Nasal Administration of Cationic Nanoemulsions as CD73-siRNA Delivery System for Glioblastoma Treatment: A New Therapeutical Approach. Mol. Neurobiol. 2019. [Google Scholar] [CrossRef]
  80. Gao, Z.W.; Wang, H.P.; Lin, F.; Wang, X.; Long, M.; Zhang, H.Z.; Dong, K. CD73 promotes proliferation and migration of human cervical cancer cells independent of its enzyme activity. BMC Cancer 2017, 17, 135. [Google Scholar] [CrossRef] [Green Version]
  81. Di Virgilio, F.; Sarti, A.C.; Grassi, F. Modulation of innate and adaptive immunity by P2X ion channels. Curr. Opin. Immunol. 2018, 52, 51–59. [Google Scholar] [CrossRef] [PubMed]
  82. Antonioli, L.; Fornai, M.; Blandizzi, C.; Pacher, P.; Hasko, G. Adenosine signaling and the immune system: When a lot could be too much. Immunol. Lett. 2019, 205, 9–15. [Google Scholar] [CrossRef] [PubMed]
  83. Antonioli, L.; Blandizzi, C.; Pacher, P.; Hasko, G. Immunity, inflammation and cancer: A leading role for adenosine. Nat. Rev. Cancer 2013, 13, 842–857. [Google Scholar] [CrossRef] [PubMed]
  84. Kazemi, M.H.; Raoofi Mohseni, S.; Hojjat-Farsangi, M.; Anvari, E.; Ghalamfarsa, G.; Mohammadi, H.; Jadidi-Niaragh, F. Adenosine and adenosine receptors in the immunopathogenesis and treatment of cancer. J. Cell. Physiol. 2018, 233, 2032–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Kreckler, L.M.; Wan, T.C.; Ge, Z.D.; Auchampach, J.A. Adenosine inhibits tumor necrosis factor-alpha release from mouse peritoneal macrophages via A2A and A2B but not the A3 adenosine receptor. J. Pharmacol. Exp. Ther. 2006, 317, 172–180. [Google Scholar] [CrossRef] [PubMed]
  86. Regateiro, F.S.; Cobbold, S.P.; Waldmann, H. CD73 and adenosine generation in the creation of regulatory microenvironments. Clin. Exp. Immunol. 2013, 171, 1–7. [Google Scholar] [CrossRef]
  87. Ponce, N.E.; Sanmarco, L.M.; Eberhardt, N.; Garcia, M.C.; Rivarola, H.W.; Cano, R.C.; Aoki, M.P. CD73 Inhibition Shifts Cardiac Macrophage Polarization toward a Microbicidal Phenotype and Ameliorates the Outcome of Experimental Chagas Cardiomyopathy. J. Immunol. 2016, 197, 814–823. [Google Scholar] [CrossRef]
  88. Hasko, G.; Pacher, P. Regulation of macrophage function by adenosine. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 865–869. [Google Scholar] [CrossRef] [Green Version]
  89. Murray, P.J.; Allen, J.E.; Biswas, S.K.; Fisher, E.A.; Gilroy, D.W.; Goerdt, S.; Gordon, S.; Hamilton, J.A.; Ivashkiv, L.B.; Lawrence, T.; et al. Macrophage activation and polarization: Nomenclature and experimental guidelines. Immunity 2014, 41, 14–20. [Google Scholar] [CrossRef] [Green Version]
  90. Gordon, S.; Pluddemann, A.; Martinez Estrada, F. Macrophage heterogeneity in tissues: Phenotypic diversity and functions. Immunol. Rev. 2014, 262, 36–55. [Google Scholar] [CrossRef] [Green Version]
  91. Martinez, F.O.; Helming, L.; Gordon, S. Alternative activation of macrophages: An immunologic functional perspective. Annu. Rev. Immunol. 2009, 27, 451–483. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Hasko, G.; Kuhel, D.G.; Chen, J.F.; Schwarzschild, M.A.; Deitch, E.A.; Mabley, J.G.; Marton, A.; Szabo, C. Adenosine inhibits IL-12 and TNF-[alpha] production via adenosine A2a receptor-dependent and independent mechanisms. FASEB J. 2000, 14, 2065–2074. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Barnholt, K.E.; Kota, R.S.; Aung, H.H.; Rutledge, J.C. Adenosine blocks IFN-gamma-induced phosphorylation of STAT1 on serine 727 to reduce macrophage activation. J. Immunol. 2009, 183, 6767–6777. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Szabo, C.; Scott, G.S.; Virag, L.; Egnaczyk, G.; Salzman, A.L.; Shanley, T.P.; Hasko, G. Suppression of macrophage inflammatory protein (MIP)-1alpha production and collagen-induced arthritis by adenosine receptor agonists. Br. J. Pharmacol. 1998, 125, 379–387. [Google Scholar] [CrossRef] [Green Version]
  95. Hasko, G.; Pacher, P.; Deitch, E.A.; Vizi, E.S. Shaping of monocyte and macrophage function by adenosine receptors. Pharmacol. Ther. 2007, 113, 264–275. [Google Scholar] [CrossRef] [Green Version]
  96. Hasko, G.; Szabo, C.; Nemeth, Z.H.; Kvetan, V.; Pastores, S.M.; Vizi, E.S. Adenosine receptor agonists differentially regulate IL-10, TNF-alpha, and nitric oxide production in RAW 264.7 macrophages and in endotoxemic mice. J. Immunol. 1996, 157, 4634–4640. [Google Scholar]
  97. Ferrante, C.J.; Pinhal-Enfield, G.; Elson, G.; Cronstein, B.N.; Hasko, G.; Outram, S.; Leibovich, S.J. The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL-4Ralpha) signaling. Inflammation 2013, 36, 921–931. [Google Scholar] [CrossRef]
  98. Sciaraffia, E.; Riccomi, A.; Lindstedt, R.; Gesa, V.; Cirelli, E.; Patrizio, M.; De Magistris, M.T.; Vendetti, S. Human monocytes respond to extracellular cAMP through A2A and A2B adenosine receptors. J. Leukoc. Biol. 2014, 96, 113–122. [Google Scholar] [CrossRef] [Green Version]
  99. Zanin, R.F.; Braganhol, E.; Bergamin, L.S.; Campesato, L.F.; Filho, A.Z.; Moreira, J.C.; Morrone, F.B.; Sevigny, J.; Schetinger, M.R.; de Souza Wyse, A.T.; et al. Differential macrophage activation alters the expression profile of NTPDase and ecto-5’-nucleotidase. PLoS ONE 2012, 7, e31205. [Google Scholar] [CrossRef] [Green Version]
  100. 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] [Green Version]
  101. Bergamin, L.S.; Braganhol, E.; Figueiro, F.; Casali, E.A.; Zanin, R.F.; Sevigny, J.; Battastini, A.M. Involvement of purinergic system in the release of cytokines by macrophages exposed to glioma-conditioned medium. J. Cell. Biochem. 2015, 116, 721–729. [Google Scholar] [CrossRef] [PubMed]
  102. Azambuja, J.H.; da Silveira, E.F.; de Carvalho, T.R.; Oliveira, P.S.; Pacheco, S.; do Couto, C.T.; Beira, F.T.; Stefanello, F.M.; Spanevello, R.M.; Braganhol, E. Glioma sensitive or chemoresistant to temozolomide differentially modulate macrophage protumor activities. Biochim. Biophys. Acta Gen. Subj. 2017, 1861, 2652–2662. [Google Scholar] [CrossRef] [PubMed]
  103. 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] [Green Version]
  104. Cekic, C.; Linden, J. Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res. 2014, 74, 7239–7249. [Google Scholar] [CrossRef] [Green Version]
  105. Schiedel, A.C.; Lacher, S.K.; Linnemann, C.; Knolle, P.A.; Muller, C.E. Antiproliferative effects of selective adenosine receptor agonists and antagonists on human lymphocytes: Evidence for receptor-independent mechanisms. Purinergic Signal. 2013, 9, 351–365. [Google Scholar] [CrossRef] [Green Version]
  106. Ohta, A.; Kini, R.; Ohta, A.; Subramanian, M.; Madasu, M.; Sitkovsky, M. The development and immunosuppressive functions of CD4(+) CD25(+) FoxP3(+) regulatory T cells are under influence of the adenosine-A2A adenosine receptor pathway. Front. Immunol. 2012, 3, 190. [Google Scholar] [CrossRef] [Green Version]
  107. Erdmann, A.A.; Gao, Z.G.; Jung, U.; Foley, J.; Borenstein, T.; Jacobson, K.A.; Fowler, D.H. Activation of Th1 and Tc1 cell adenosine A2A receptors directly inhibits IL-2 secretion in vitro and IL-2-driven expansion in vivo. Blood 2005, 105, 4707–4714. [Google Scholar] [CrossRef]
  108. Munoz, E.; Zubiaga, A.M.; Merrow, M.; Sauter, N.P.; Huber, B.T. Cholera toxin discriminates between T helper 1 and 2 cells in T cell receptor-mediated activation: Role of cAMP in T cell proliferation. J. Exp. Med. 1990, 172, 95–103. [Google Scholar] [CrossRef] [Green Version]
  109. Lappas, C.M.; Day, Y.J.; Marshall, M.A.; Engelhard, V.H.; Linden, J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J. Exp. Med. 2006, 203, 2639–2648. [Google Scholar] [CrossRef] [Green Version]
  110. Ohta, A.; Kjaergaard, J.; Sharma, S.; Mohsin, M.; Goel, N.; Madasu, M.; Fradkov, E.; Ohta, A.; Sitkovsky, M. In vitro induction of T cells that are resistant to A 2 adenosine receptor-mediated immunosuppression. Br. J. Pharmacol. 2009, 156, 297–306. [Google Scholar] [CrossRef] [Green Version]
  111. Kjaergaard, J.; Hatfield, S.; Jones, G.; Ohta, A.; Sitkovsky, M. A2A adenosine receptor gene-deletion or synthetic A2A antagonist liberate tumor-reactive CD8+ T-cells from tumor-induced immunosuppression. J. Immunol. 2018, 201, 782–791. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Zhang, H.; Conrad, D.M.; Butler, J.J.; Zhao, C.; Blay, J.; Hoskin, D.W. Adenosine acts through A2 receptors to inhibit IL-2-induced tyrosine phosphorylation of STAT5 in T lymphocytes: Role of cyclic adenosine 3’,5’-monophosphate and phosphatases. J. Immunol. 2004, 173, 932–944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Minguet, S.; Huber, M.; Rosenkranz, L.; Schamel, W.W.; Reth, M.; Brummer, T. Adenosine and cAMP are potent inhibitors of the NF-kappa B pathway downstream of immunoreceptors. Eur. J. Immunol. 2005, 35, 31–41. [Google Scholar] [CrossRef] [PubMed]
  114. Diab, D.L.; Yerian, L.; Schauer, P.; Kashyap, S.R.; Lopez, R.; Hazen, S.L.; Feldstein, A.E. Cytokeratin 18 fragment levels as a noninvasive biomarker for nonalcoholic steatohepatitis in bariatric surgery patients. Clin. Gastroenterol. Hepatol. 2008, 6, 1249–1254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Gessi, S.; Varani, K.; Merighi, S.; Cattabriga, E.; Avitabile, A.; Gavioli, R.; Fortini, C.; Leung, E.; Mac Lennan, S.; Borea, P.A. Expression of A3 adenosine receptors in human lymphocytes: Up-regulation in T cell activation. Mol. Pharmacol. 2004, 65, 711–719. [Google Scholar] [CrossRef]
  116. Priebe, T.; Platsoucas, C.D.; Nelson, J.A. Adenosine receptors and modulation of natural killer cell activity by purine nucleosides. Cancer Res. 1990, 50, 4328–4331. [Google Scholar]
  117. Chambers, A.M.; Wang, J.; Lupo, K.B.; Yu, H.; Atallah Lanman, N.M.; Matosevic, S. Adenosinergic Signaling Alters Natural Killer Cell Functional Responses. Front. Immunol. 2018, 9, 2533. [Google Scholar] [CrossRef]
  118. Yu, J.C.; Lin, G.; Field, J.J.; Linden, J. Induction of antiinflammatory purinergic signaling in activated human iNKT cells. JCI Insight 2018, 3. [Google Scholar] [CrossRef]
  119. Jeffe, F.; Stegmann, K.A.; Broelsch, F.; Manns, M.P.; Cornberg, M.; Wedemeyer, H. Adenosine and IFN-{alpha} synergistically increase IFN-gamma production of human NK cells. J. Leukoc. Biol. 2009, 85, 452–461. [Google Scholar] [CrossRef]
  120. Morandi, F.; Horenstein, A.L.; Chillemi, A.; Quarona, V.; Chiesa, S.; Imperatori, A.; Zanellato, S.; Mortara, L.; Gattorno, M.; Pistoia, V.; et al. CD56brightCD16- NK Cells Produce Adenosine through a CD38-Mediated Pathway and Act as Regulatory Cells Inhibiting Autologous CD4+ T Cell Proliferation. J. Immunol. 2015, 195, 965–972. [Google Scholar] [CrossRef] [Green Version]
  121. Kuldova, M.; Svoboda, J.; Kovaru, F.; Vannucci, L.; Kovaru, H.; Fiserova, A. NK cell-mediated cytotoxicity modulation by A(2) adenosine receptor agonist in different mammalian species. Folia Microbiol. (Praha) 2009, 54, 364–368. [Google Scholar] [CrossRef] [PubMed]
  122. 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] [PubMed] [Green Version]
  123. Harish, A.; Hohana, G.; Fishman, P.; Arnon, O.; Bar-Yehuda, S. A3 adenosine receptor agonist potentiates natural killer cell activity. Int. J. Oncol. 2003, 23, 1245–1249. [Google Scholar] [CrossRef] [PubMed]
  124. Dwyer, K.M.; Hanidziar, D.; Putheti, P.; Hill, P.A.; Pommey, S.; McRae, J.L.; Winterhalter, A.; Doherty, G.; Deaglio, S.; Koulmanda, M.; et al. Expression of CD39 by human peripheral blood CD4+ CD25+ T cells denotes a regulatory memory phenotype. Am. J. Transplant. 2010, 10, 2410–2420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Horenstein, A.L.; Chillemi, A.; Zaccarello, G.; Bruzzone, S.; Quarona, V.; Zito, A.; Serra, S.; Malavasi, F. A CD38/CD203a/CD73 ectoenzymatic pathway independent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2013, 2, e26246. [Google Scholar] [CrossRef] [Green Version]
  126. 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] [Green Version]
  127. Di Gennaro, P.; Gerlini, G.; Caporale, R.; Sestini, S.; Brandani, P.; Urso, C.; Pimpinelli, N.; Borgognoni, L. T regulatory cells mediate immunosuppresion by adenosine in peripheral blood, sentinel lymph node and TILs from melanoma patients. Cancer Lett. 2018, 417, 124–130. [Google Scholar] [CrossRef]
  128. Ohta, A.; Gorelik, E.; Prasad, S.J.; Ronchese, F.; Lukashev, D.; Wong, M.K.K.; Huang, X.; Caldwell, S.; Liu, K.; Smith, P.; et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl. Acad. Sci. USA 2006, 103, 13132–13137. [Google Scholar] [CrossRef] [Green Version]
  129. Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene 2008, 27, 5904–5912. [Google Scholar] [CrossRef] [Green Version]
  130. Whiteside, T.L. Exosomes and tumor-mediated immune suppression. J. Clin. Invest. 2016, 126, 1216–1223. [Google Scholar] [CrossRef] [Green Version]
  131. Morandi, F.; Marimpietri, D.; Horenstein, A.L.; Bolzoni, M.; Toscani, D.; Costa, F.; Castella, B.; Faini, A.C.; Massaia, M.; Pistoia, V.; et al. Microvesicles released from multiple myeloma cells are equipped with ectoenzymes belonging to canonical and non-canonical adenosinergic pathways and produce adenosine from ATP and NAD\r+\r. Oncoimmunology 2018, 7, e1458809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. 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]
  133. Salimu, J.; Webber, J.; Gurney, M.; Al-Taei, S.; Clayton, A.; Tabi, Z. Dominant immunosuppression of dendritic cell function by prostate-cancer-derived exosomes. J. Extracell. Vesicles 2017, 6, 1368823. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Theodoraki, M.N.; Hoffmann, T.K.; Jackson, E.K.; Whiteside, T.L. Exosomes in HNSCC plasma as surrogate markers of tumour progression and immune competence. Clin. Exp. Immunol. 2018, 194, 67–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Smyth, L.A.; Ratnasothy, K.; Tsang, J.Y.; Boardman, D.; Warley, A.; Lechler, R.; Lombardi, G. CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur. J. Immunol. 2013, 43, 2430–2440. [Google Scholar] [CrossRef] [PubMed]
  136. Puhan, M.A.; Chandra, D.; Mosenifar, Z.; Ries, A.; Make, B.; Hansel, N.N.; Wise, R.A.; Sciurba, F. National Emphysema Treatment Trial Research, G., The minimal important difference of exercise tests in severe COPD. Eur. Respir. J. 2011, 37, 784–790. [Google Scholar] [CrossRef]
  137. Zhou, Y.; Tong, L.; Chu, X.; Deng, F.; Tang, J.; Tang, Y.; Dai, Y. The Adenosine A1 Receptor Antagonist DPCPX Inhibits Tumor Progression via the ERK/JNK Pathway in Renal Cell Carcinoma. Cell. Physiol. Biochem. 2017, 43, 733–742. [Google Scholar] [CrossRef]
  138. Young, A.; Ngiow, S.F.; Barkauskas, D.S.; Sult, E.; Hay, C.; Blake, S.J.; Huang, Q.; Liu, J.; Takeda, K.; Teng, M.W.L.; et al. Co-inhibition of CD73 and A2AR Adenosine Signaling Improves Anti-tumor Immune Responses. Cancer Cell 2016, 30, 391–403. [Google Scholar] [CrossRef] [Green Version]
  139. 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. Invest. 2017, 127, 929–941. [Google Scholar] [CrossRef] [Green Version]
  140. Beavis, P.A.; Milenkovski, N.; Henderson, M.A.; John, L.B.; Allard, B.; Loi, S.; Kershaw, M.H.; Stagg, J.; Darcy, P.K. Adenosine Receptor 2A Blockade Increases the Efficacy of Anti-PD-1 through Enhanced Antitumor T-cell Responses. Cancer Immunol. Res. 2015, 3, 506–517. [Google Scholar] [CrossRef]
  141. Arab, S.; Kheshtchin, N.; Ajami, M.; Ashurpoor, M.; Safvati, A.; Namdar, A.; Mirzaei, R.; Mousavi Niri, N.; Jadidi-Niaragh, F.; Ghahremani, M.H.; et al. Increased efficacy of a dendritic cell-based therapeutic cancer vaccine with adenosine receptor antagonist and CD73 inhibitor. Tumour Biol. 2017, 39, 1010428317695021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Molck, C.; Ryall, J.; Failla, L.M.; Coates, J.L.; Pascussi, J.M.; Heath, J.K.; Stewart, G.; Hollande, F. The A2b adenosine receptor antagonist PSB-603 promotes oxidative phosphorylation and ROS production in colorectal cancer cells via adenosine receptor-independent mechanism. Cancer Lett. 2016, 383, 135–143. [Google Scholar] [CrossRef] [PubMed]
  143. 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] [PubMed] [Green Version]
  144. Kanno, T.; Gotoh, A.; Fujita, Y.; Nakano, T.; Nishizaki, T. A(3) adenosine receptor mediates apoptosis in 5637 human bladder cancer cells by G(q) protein/PKC-dependent AIF upregulation. Cell. Physiol. Biochem. 2012, 30, 1159–1168. [Google Scholar] [CrossRef] [PubMed]
  145. Panjehpour, M.; Karami-Tehrani, F. An adenosine analog (IB-MECA) inhibits anchorage-dependent cell growth of various human breast cancer cell lines. Int. J. Biochem. Cell Biol. 2004, 36, 1502–1509. [Google Scholar] [CrossRef]
  146. Morello, S.; Sorrentino, R.; Porta, A.; Forte, G.; Popolo, A.; Petrella, A.; Pinto, A. Cl-IB-MECA enhances TRAIL-induced apoptosis via the modulation of NF-kappaB signalling pathway in thyroid cancer cells. J. Cell. Physiol. 2009, 221, 378–386. [Google Scholar] [CrossRef]
  147. Fishman, P.; Bar-Yehuda, S.; Ohana, G.; Barer, F.; Ochaion, A.; Erlanger, A.; Madi, L. An agonist to the A3 adenosine receptor inhibits colon carcinoma growth in mice via modulation of GSK-3 beta and NF-kappa B. Oncogene 2004, 23, 2465–2471. [Google Scholar] [CrossRef] [Green Version]
  148. Zhi, X.; Chen, S.; Zhou, P.; Shao, Z.; Wang, L.; Ou, Z.; Yin, L. RNA interference of ecto-5’-nucleotidase (CD73) inhibits human breast cancer cell growth and invasion. Clin. Exp. Metastasis 2007, 24, 439–448. [Google Scholar] [CrossRef]
  149. Terp, M.G.; Olesen, K.A.; Arnspang, E.C.; Lund, R.R.; Lagerholm, B.C.; Ditzel, H.J.; Leth-Larsen, R. Anti-human CD73 monoclonal antibody inhibits metastasis formation in human breast cancer by inducing clustering and internalization of CD73 expressed on the surface of cancer cells. J. Immunol. 2013, 191, 4165–4173. [Google Scholar] [CrossRef] [Green Version]
  150. Iannone, R.; Miele, L.; Maiolino, P.; Pinto, A.; Morello, S. Adenosine limits the therapeutic effectiveness of anti-CTLA4 mAb in a mouse melanoma model. Am. J. Cancer Res. 2014, 4, 172–181. [Google Scholar]
  151. Jadidi-Niaragh, F.; Atyabi, F.; Rastegari, A.; Kheshtchin, N.; Arab, S.; Hassannia, H.; Ajami, M.; Mirsanei, Z.; Habibi, S.; Masoumi, F.; et al. CD73 specific siRNA loaded chitosan lactate nanoparticles potentiate the antitumor effect of a dendritic cell vaccine in 4T1 breast cancer bearing mice. J. Control. Release 2017, 246, 46–59. [Google Scholar] [CrossRef] [PubMed]
  152. Jadidi-Niaragh, F.; Atyabi, F.; Rastegari, A.; Mollarazi, E.; Kiani, M.; Razavi, A.; Yousefi, M.; Kheshtchin, N.; Hassannia, H.; Hadjati, J.; et al. Downregulation of CD73 in 4T1 breast cancer cells through siRNA-loaded chitosan-lactate nanoparticles. Tumour Biol. 2016, 37, 8403–8412. [Google Scholar] [CrossRef] [PubMed]
  153. van de Donk, N. Reprint of “Immunomodulatory effects of CD38-targeting antibodies”. Immunol. Lett. 2019, 205, 71–77. [Google Scholar] [CrossRef] [PubMed]
  154. Krejcik, J.; Casneuf, T.; Nijhof, I.S.; Verbist, B.; Bald, J.; Plesner, T.; Syed, K.; Liu, K.; van de Donk, N.W.; Weiss, B.M.; et al. Daratumumab depletes CD38+ immune regulatory cells, promotes T-cell expansion, and skews T-cell repertoire in multiple myeloma. Blood 2016, 128, 384–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. de Leve, S.; Wirsdorfer, F.; Jendrossek, V. Targeting the Immunomodulatory CD73/Adenosine System to Improve the Therapeutic Gain of Radiotherapy. Front. Immunol. 2019, 10, 698. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Hay, C.M.; Sult, E.; Huang, Q.; Mulgrew, K.; Fuhrmann, S.R.; McGlinchey, K.A.; Hammond, S.A.; Rothstein, R.; Rios-Doria, J.; Poon, E.; et al. Targeting CD73 in the tumor microenvironment with MEDI9447. Oncoimmunology 2016, 5, e1208875. [Google Scholar] [CrossRef] [PubMed]
  157. Mittal, D.; Young, A.; Stannard, K.; Yong, M.; Teng, M.W.; Allard, B.; Stagg, J.; Smyth, M.J. Antimetastatic effects of blocking PD-1 and the adenosine A2A receptor. Cancer Res. 2014, 74, 3652–3658. [Google Scholar] [CrossRef]
Figure 1. Extracellular and intracellular (canonical and non-canonical) adenosinergic pathways. In (A) biochemical interactions in the adenosine (ADO) pathway and in (B) cellular receptors in the ADO pathway.
Figure 1. Extracellular and intracellular (canonical and non-canonical) adenosinergic pathways. In (A) biochemical interactions in the adenosine (ADO) pathway and in (B) cellular receptors in the ADO pathway.
Ijms 20 05698 g001
Figure 2. The opposite effects of ATP (pro-inflammatory) and ADO (anti-inflammatory) on immune cells (T cells, NK cells, and macrophages). In (A), ATP in extracellular fluids can be considered as a damage-associated molecular pattern (DAMP), which can trigger an inflammatory response characterized by proliferation, cytotoxicity, and the secretion of pro-inflammatory cytokines, such as interleukin-2 (IL-2), interferon-γ (INF-γ), and interleukin-β (IL-1β) [81]. In (B), ADO mediates immunosuppressive responses for the protection of tissues adjacent to inflammation from attacks by immune cells. In this case, ADO induces secretion of anti-inflammatory cytokines, such IL-10, and reduces secretion of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), IL-12, and IL-2 [42,85,86,87,88].
Figure 2. The opposite effects of ATP (pro-inflammatory) and ADO (anti-inflammatory) on immune cells (T cells, NK cells, and macrophages). In (A), ATP in extracellular fluids can be considered as a damage-associated molecular pattern (DAMP), which can trigger an inflammatory response characterized by proliferation, cytotoxicity, and the secretion of pro-inflammatory cytokines, such as interleukin-2 (IL-2), interferon-γ (INF-γ), and interleukin-β (IL-1β) [81]. In (B), ADO mediates immunosuppressive responses for the protection of tissues adjacent to inflammation from attacks by immune cells. In this case, ADO induces secretion of anti-inflammatory cytokines, such IL-10, and reduces secretion of pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), IL-12, and IL-2 [42,85,86,87,88].
Ijms 20 05698 g002
Figure 3. Effects of extracellular ADO on subsets of macrophages. In (A), differentiation of M1 and M2 and their characteristics is shown, including functions they mediate. In (B), the activation of the P1Rs on macrophages suppresses the M1 phenotype [88]. Activation of A2AR and A2BR inhibits production of interleukin-12 (IL-12) [92], interferon-γ (IFN-γ) [93], macrophage inflammatory protein-1 (MIP-1α) [94], tumor necrosis factor-α (TNF-α) [95], and nitric oxide (NO) [96] and induces IL-10 [96] and vascular endothelial growth factor (VEGF) production (left side). In contrast, suppression of CD73 activity enhances the M1 phenotype and blocks IL-4 and IL-10 production [87,97] (right side).
Figure 3. Effects of extracellular ADO on subsets of macrophages. In (A), differentiation of M1 and M2 and their characteristics is shown, including functions they mediate. In (B), the activation of the P1Rs on macrophages suppresses the M1 phenotype [88]. Activation of A2AR and A2BR inhibits production of interleukin-12 (IL-12) [92], interferon-γ (IFN-γ) [93], macrophage inflammatory protein-1 (MIP-1α) [94], tumor necrosis factor-α (TNF-α) [95], and nitric oxide (NO) [96] and induces IL-10 [96] and vascular endothelial growth factor (VEGF) production (left side). In contrast, suppression of CD73 activity enhances the M1 phenotype and blocks IL-4 and IL-10 production [87,97] (right side).
Ijms 20 05698 g003
Figure 4. The cellular effects of extracellular ADO in the regulation of lymphocyte functions. After A2AR stimulation, regulatory T cells (Tregs) increased proliferation and expression of CTLA-4, CD39, and CD73, and inhibited CD8+ T cell proliferation [106]. CD4+ T cells decreased the production of IL-2, TNFα, IL-6, and IFNγ, and enhanced the production of IL-10 [107,108,109]. In NK cells, ADO suppresses the production of IL-2, TNF-α, IFN-γ, and granulocyte macrophage colony-stimulating factor (GM-CSF) and blocks their cytolytic activity [75]. In CD8+ T cells, ADO suppresses the production of IL-2 and IFN-γ and blocks their cytolytic activity [110,111].
Figure 4. The cellular effects of extracellular ADO in the regulation of lymphocyte functions. After A2AR stimulation, regulatory T cells (Tregs) increased proliferation and expression of CTLA-4, CD39, and CD73, and inhibited CD8+ T cell proliferation [106]. CD4+ T cells decreased the production of IL-2, TNFα, IL-6, and IFNγ, and enhanced the production of IL-10 [107,108,109]. In NK cells, ADO suppresses the production of IL-2, TNF-α, IFN-γ, and granulocyte macrophage colony-stimulating factor (GM-CSF) and blocks their cytolytic activity [75]. In CD8+ T cells, ADO suppresses the production of IL-2 and IFN-γ and blocks their cytolytic activity [110,111].
Ijms 20 05698 g004
Table 1. In vitro and in vivo studies of ADO chemoresistance activities reported in the literature.
Table 1. In vitro and in vivo studies of ADO chemoresistance activities reported in the literature.
ModelMain ResultMechanism
P1R antagonism
Melanoma in vivoInhibition of A2BR enhanced efficacy of dacarbazineReversed immune suppression in the TME [42]
Glioblastoma in vivoInhibition of A2BR enhanced efficacy of TMZA2BR [43]
CD73 inhibition
Glioblastoma in vitro CD73 KO increased efficacy of TMZADO production [44]
Glioblastoma in vitroCD73 KO reversed the MDR phenotype A3R [45]
Leukemia in vitroCD73 KO restored TRAIL sensitivityIndependent of CD73enzymatic activity [46]
Breast cancer in vivoAnti-CD73 mab therapy enhanced efficacy of anti-ErbB2 mAb Association of CD73 expression with TGF-β, EMT and HIF-1 [47]
Ovarian cancer
in vitro and in vivo
Anti-CD73 mab therapy enhanced docetaxel responseReverse the immunosuppression [48]
Breast cancer in vivoCD73 inhibitor therapy enhanced efficacy of doxorubicinActivation of immune response mediated by A2AR [49]
Table 2. In vitro and in vivo studies of pro and anti-tumor activities of ADO reported in the literature.
Table 2. In vitro and in vivo studies of pro and anti-tumor activities of ADO reported in the literature.
ModelMain ResultMechanism
P1R antagonism
Breast cancer in vitroInhibition of A1R induced apoptosisUpregulation of p53 and caspases [50]
Colon carcinoma in vitroInhibition of A2BR suppressed tumor growthA2BR [51]
Prostate cancer in vitroInhibition of A2BR suppressed tumor growthA2BR [52,53]
Oral squamous cell carcinoma in vitroInhibition of A2BR suppressed tumor growthA2BR [54]
Melanoma in vivoActivation of P1R inhibited melanoma growthEnhance immune killing of tumors [55]
HNSCC in vivoInhibition of A2AR suppressed tumor growthReduced Tregs population and enhanced the anti-tumor response of CD8+ T cells [56]
Lung adenocarcinoma in vivoInhibition of A2AR suppressed tumor growthPrevented negative signaling in T cells and inhibited angiogenesis [57]
Melanoma in vivoInhibition of A2AR suppressed tumor growthNK activation [58]
Melanoma in vivoInhibition of A2BR suppressed tumor growthReduced Tregs population and increased in CD4+ and CD8+ T cells [59]
P1R agonism
Leukemia in vitroActivation of A3R induced cell cycle arrest and apoptosisModulation of Wnt, β-catenin, GSK-β and AKT [60]
Bladder cancer in vitroActivation of A3R induced cell cycle arrest and apoptosisERK and JNK activation [61]
Cancer cell linesActivation of A3R induced cell cycle arrest and apoptosisDownregulation of CDK4, cyclin D1 and upregulation of p53 [62]
Ovarian cancer in vitroActivation of A3R reduced cell viability and induced cell cycle arrestDownregulation of Cyclin D1 and CDK4 [63]
Renal cancer in vitroActivation of A3R induced apoptosisAMID upregulation [64]
Glioblastoma in vitroActivation of A3R induced cell deathERK and AKT downregulation [62]
Lung cancer in vitroActivation of A3R induced cell deathMediated by caspases upregulation [65]
Breast cancer in vivoActivation of A3R inhibited tumor proliferationNot reported [66]
CD73 inhibition
Glioblastoma in vitro and in vivoKnockdown of CD73 decreased glioma growthStimulation of AKT/NF-kB pathways [44]
CD73 overexpression
Medulloblastoma
in vitro and in vivo
Reduced proliferation and vascularizationMediated by A1R [67]
Table 3. In vitro and in vivo studies of the ADO role in tumor migration, invasiveness, and angiogenesis as reported in the literature.
Table 3. In vitro and in vivo studies of the ADO role in tumor migration, invasiveness, and angiogenesis as reported in the literature.
ModelMain ResultMechanism
P1R antagonism
Melanoma in vitroReduced angiogenesisA2BR blockade impairs IL-8 production, whereas blocking A3R decreases VEGF [68]
Breast cancer and melanoma in vivoA2AR blockade reduced metastasisEnhanced NK cell maturation and cytotoxicity [69]
P1R agonism
Breast cancer in vitroActivation of A3R induced migrationNot reported [70]
Colon cancer in vitroEnhanced migrationA2BR and A3R activation and regulation HIF-1alpha/VEGF/IL-8 via ERK1/2, p38, and AKT [71]
CD73 inhibition
Ovarian Carcinoma in vitroCD73 inhibitor blocked migrationNot reported [72]
Glioblastoma in vitro CD73 KO decreased migration and invasionAltered MMP-2 and Vimentin expression [44]
Breast cancer in vitroAnti-CD73 mab therapy inhibited migration, invasion and adhesion EGFR and IL-8 [73]
Breast cancer in vivoAnti-CD73 mab therapy decreased lung metastasesActivation of NK cells, CD8+ T and IFNγ by A2BR [74,75]
Melanoma in vitro
and in vivo
CD73 inhibitor decreased adherence of cells and enhanced migration and invasionVia P1R [76]
Breast cancer in vitro
and in vivo
Anti-CD73 mab therapy inhibited migration metastasis in vivoCD73 expression promoted autophagy [77]
Hepatocellular cancer in vitro and in vivoCD73 KO inhibited migration, invasion and metastasis A2AR activates Rap1, P110β, and PIP3 production by AKT [78]
Glioblastoma in vivoCD73 KO inhibited angiogenesisNot reported [79]
CD73 overexpression
Cervical cancer in vitroPromoted migration; and high concentration inhibited migration.Upregulation of EGFR, VEGF, and AKT [80]
Table 4. Clinical trials a.
Table 4. Clinical trials a.
NCT NumberPhaseYearType of CancerDrug NameTarget
NCT00879775Phase 22009CancerCaffeineP1R antagonist
NCT024031093Phase 1/22015Non-small Cell Lung Cancer (NSCLC)PBF-509A2AR antagonist
NCT02655822Phase 12016Advanced CancersCPI-444A2AR antagonist
NCT03274479Phase 12018Locally Advanced or Metastatic NSCLCPBF-1129A2BR antagonist
NCT00790218Phase 1/22009Hepatocellular CarcinomaCF102A3R antagonist
NCT01987999Phase 22013Prostate CancerAcetogeninsATP inhibitor
NCT02503774Phase 12015Solid TumorsMEDI9447CD73
NCT03267589Phase 22017Relapsed Ovarian CancerMEDI9447CD73
NCT03616886Phase 1/22018Triple Negative Breast CancerMEDI9447CD73
NCT03549000Phase 12018Advanced MalignanciesNZV930CD73
NCT03381274Phase 1/22018NSCLCMEDI9447CD73
NCT03454451Phase 12018CancerCPI-006CD73
NCT03835949Phase 12019Advanced or Metastatic CancerTJ004309CD73
NCT03875573Phase 22019Luminal B Breast CanceroleclumabCD73
a A current list of clinical trials investigating the role of targeting purinergic signaling in cancer.

Share and Cite

MDPI and ACS Style

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. https://doi.org/10.3390/ijms20225698

AMA Style

Azambuja JH, Ludwig N, Braganhol E, Whiteside TL. Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity. International Journal of Molecular Sciences. 2019; 20(22):5698. https://doi.org/10.3390/ijms20225698

Chicago/Turabian Style

Azambuja, Juliana Hofstätter, Nils Ludwig, Elizandra Braganhol, and Theresa L. Whiteside. 2019. "Inhibition of the Adenosinergic Pathway in Cancer Rejuvenates Innate and Adaptive Immunity" International Journal of Molecular Sciences 20, no. 22: 5698. https://doi.org/10.3390/ijms20225698

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