Download PDF
Review  |  Open Access  |  24 Oct 2023

Unlocking antitumor immunity with adenosine receptor blockers

Views: 971 |  Downloads: 241 |  Cited:   0
Cancer Drug Resist 2023;6:748-67.
10.20517/cdr.2023.63 |  © The Author(s) 2023.
Author Information
Article Notes
Cite This Article

Abstract

Tumors survive by creating a tumor microenvironment (TME) that suppresses antitumor immunity. The TME suppresses the immune system by limiting antigen presentation, inhibiting lymphocyte and natural killer (NK) cell activation, and facilitating T cell exhaustion. Checkpoint inhibitors like anti-PD-1 and anti-CTLA4 are immunostimulatory antibodies, and their blockade extends the survival of some but not all cancer patients. Extracellular adenosine triphosphate (ATP) is abundant in inflamed tumors, and its metabolite, adenosine (ADO), is a driver of immunosuppression mediated by adenosine A2A receptors (A2AR) and adenosine A2B receptors (A2BR) found on tumor-associated lymphoid and myeloid cells. This review will focus on adenosine as a key checkpoint inhibitor-like immunosuppressive player in the TME and how reducing adenosine production or blocking A2AR and A2BR enhances antitumor immunity.

Keywords

Immunotherapy, adenosine, adenosine receptors, adenosine A2A receptors (A2AR), adenosine A2B receptors (A2BR), tumor cells, immune cells, tumor microenvironment

INTRODUCTION

Deadly tumors have the ability to resist the body’s formidable immune defenses. They create protective micro-environments that limit antigen presentation, inhibit T and natural killer (NK) cell responses, and induce T cell exhaustion, effectively escaping immune surveillance. These mechanisms allow some tumors to grow unchecked and resist conventional cancer therapies.

Checkpoint inhibitors that block immunosuppressive signaling molecules such as PD-1, T lymphocyte antigen 4 (CTLA4), and lymphocytic activation gene 3 protein (LAG-3) have ushered in a new era of cancer immunotherapy, offering hope for prolonged survival and enhanced quality of life for many patients. However, the beneficial effects of these therapies are not universal due to the ability of some tumors to maintain an immunosuppressive environment. The interplay between cancer cells and immune cells within the tumor microenvironment (TME) is a critical determinant of the therapeutic response.

A key driver of immunosuppression within the TME is extracellular adenosine (ADO), an adenosine triphosphate (ATP) metabolite. ADO formation and its immunosuppressive signaling play a pivotal role in maintaining the immunosuppressive state of the TME, promoting tumor growth, and facilitating resistance to other therapies. This review explores the role of ADO signaling in the TME. Inhibiting ADO receptors on immune cells reduces immunosuppression and, in some cases, has an additive antitumor effect when combined with other cancer treatments.

UNDERSTANDING NON-RESPONDERS TO IMMUNOTHERAPY IN SOLID TUMORS

Despite having antitumor effects, cancer immunotherapy often fails. One prominent reason is that most tumor proteins are recognized as self-proteins and fail to activate T cells, which serve as the frontline warriors of the adaptive immune response[1]. To the extent that tumors are recognized by the immune system, their activation is muted by immunosuppressive signals like adenosine.

Antigen-presenting cells (APCs) such as macrophages and dendritic cells (DCs) also regulate the immune response to tumors[2]. The function of APC is to recognize, engulf, and present tumor antigens on their surface. Their failure to optimally phagocytose and present antigens can undermine the initiation of adaptive immunity. The host’s baseline immune response also significantly influences the success or failure of cancer immunotherapy. The baseline immune response is crucial in determining treatment efficacy, represented by the association between increased T cell infiltration into tumors and improved patient survival and immunotherapy response rates[3]. However, the factors that dictate the extent of T cell infiltration into tumors, whereby an extensively infiltrated tumor is considered “hot”, and a sparsely infiltrated tumor is considered “cold”, are just beginning to be elucidated. The factors that influence infiltration vary across tumor types and subtypes due to immune cell heterogeneity[4]. The complexity and dynamics of the immune system, in conjunction with the adaptability of tumor cells, create a challenging landscape for the successful deployment of cancer immunotherapy.

The complexity of the TME in solid tumors

The TME in solid tumors is complex, consisting of various immune cells, cytokines, chemokines, and metabolites. Specific features of the TME depend on the tumor type and the location within the patient. Some tumors develop an extracellular matrix (ECM) of fibrous proteins and stromal cells that define and isolate the TME from the surrounding tissue[5,6]. Within “cold” solid tumors, very few antitumor CD8+ T cells, NK cells, and DCs are present, due to failure by immune cells to enter through the ECM[7]. Immune cells that contribute to the immunosuppressive state are tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs). Monocytes that enter tumors can polarize into M1 (proinflammatory) or M2 (immuno-suppressive) cells. In cancer, most become M2 and MDSC[8] and function to secrete vascular endothelial growth factor (VEGF) and transforming growth factor β (TGF-β), which stimulates angiogenesis within the tumor[9]. M2-TAMs and MDSCs also suppress CD8+ T cell infiltration and increase regulatory T cell (Treg) differentiation from precursor CD4+ T cells[10]. Tregs release suppressive cytokines such as interleukin-10 (IL-10) and TGF-β that inhibit CD8+ T cell function and enhance cancer cell escape from the immune attack[11]. In some solid tumors, such as pancreatic ductal adenocarcinoma (PDAC), increased IL-10 is correlated with reduced survival[12-14].

MDSCs are considered immature myeloid cells and secrete reactive oxygen species (ROS), IL-10, IL-13, TGF-β, arginase-I, inducible nitric oxide synthase (iNOS), and other immunosuppressive factors[15-17].

The TME is usually hypoxic due to a poorly developed tumor vasculature. Most cytotoxic immune cells (CD8+ T cells and NK cells) function poorly in hypoxic states, while suppressive cells (M2 TAMs and Tregs) thrive[18]. The hypoxic TME favors the production of ADO and the induction of immunosuppressive A2A receptors (A2AR) and A2B receptors (A2BR) in immune cells. A shift in tumor metabolism from oxidative phosphorylation to primarily glycolysis also suppresses immune cell infiltration due to increased lactate in the TME. The increase in lactate lowers tumor pH[19]. This decrease in pH drives M2 polarization while inhibiting the nuclear factor of activated T cells (NFAT) within T cells[20,21]. This suppression of NFAT inhibits chemotaxis into tumors and reduces T cell activation.

The role of immune checkpoint inhibitors (ICIs) in cancer immunotherapy

Immune checkpoint inhibitors (ICIs) have greatly advanced immunotherapy, especially in some hard-to-treat tumors. The two most studied checkpoints are CTLA4 and PD-1[1,22,23]. Although antibodies that block these inhibitory signals have improved survival, most patients with solid tumors eventually develop either primary or secondary resistance to ICI. In primary resistance, tumors display early resistance to ICI and progress soon (within six months) after ICI treatment. In secondary resistance, patients respond to treatment initially but develop resistance later[1]. Studies have demonstrated that the tumor mutational burden (TMB) influences the response to ICI[24-26]. Reduced TMB within tumors treated with ICI can result in acquired resistance to ICI immunotherapy.

Contributions to therapy resistance by suppressive state and hypoxia

Suppressive immune cells within the TME contribute to checkpoint inhibitor resistance. Tregs, MDSCs, and M2-TAMs secrete immunosuppressive cytokines (TGF-β, CXCL8, CCL5, and IL-10) that prevent cytotoxic infiltrating immune cells from entering the tumor[27-31]. An increase in VEGF due to the activation of the mitogen-activated protein kinase (MAPK) pathway can also stimulate tumor angiogenesis[27-31] and inhibit immune cell infiltration[32].

Hypoxia results from various physiological and pathological conditions, including solid tumors, ischemia-reperfusion injury, stroke, and chronic obstructive pulmonary disease (COPD)[33]. A hypoxic TME contributes to an increase in extracellular ADO. The molecular mechanisms underlying hypoxia-driven responses include Adora2a and Adora2b via HIF-1α and HIF-2α[34,35]. HIFs also stimulate angiogenesis, vasodilation, and attenuation of inflammation[34,36]. HIF-1α induces CD73 and CD39 and increases the conversion of ATP into ADO, leading to T cell inhibition, metastasis, and increased angiogenesis[37-40]. The accumulation of ADO within tumor suppresses cytotoxic immune cells and APCs, such as CD8+ T cells and DCs, while enhancing the accumulation of immunosuppressive cells[41,42]. When ADO encounters its receptors, it can affect the activity of neutrophils and macrophages, reducing the release of IL-12, tumor necrosis factor-alpha (TNFα), and ROS[43-45].

The role of cancer-associated fibroblasts (CAFs) in tumor development

Cancer-associated fibroblasts (CAFs) have a different morphology than other cells within the tumor. They lack epithelial, endothelial, and leukocyte markers, and do not have the same mutations as tumor cells[46]. CAF development within tumors occurs when there is an increase in inflammatory markers such as IL-6 and TGF-β due to cancer cell DNA damage[47,48]. When IL-6 and TGF-β are increased in the TME, they tend to reduce the number of T cells, limiting the extent of the antitumor response[49]. These cytokines increase JAK-STAT signaling and ECM transition, promoting CAF formation[50-52]. Breast cancers increase Notch signaling within the TME to increase CAFs[53]. Patients who receive chemotherapy and radiation to treat solid tumors experience DNA breaks, and this stress can promote fibrosis or CAF accumulation and function. This change in CAF function causes resistance to therapy in various solid tumors[54-56].

Solid tumor CAFs may exhibit different phenotypes depending on the TME. The different phenotypes exhibit different cell surface markers, but identifying these can be challenging[46]. Breast cancers increase expression of fibroblast activation protein (FAP) to cause high immunosuppression through Treg activation[57]. Pancreatic cancers express both myofibroblastic CAFs (myCAFs) and inflammatory CAFs (iCAFs) at different locations in the tumor. MyCAFs have high expression of TGF-β and αSMA and are located close to tumor cells, while iCAFs have high IL-6 secretion and are more distal in the TME[58,59]. iCAFs can recruit TAMs and MDSCs to the TME to increase the immunosuppressive state[13,60]. Targeting the MAPK/STAT pathways in iCAFs through inhibitors in combination with checkpoint inhibitor therapy (e.g., anti-PD-1) can lead to increased survival of patients with solid tumors like PDAC[60]. McAndrews et al. discovered that in early-stage PDAC, iCAFs tend to be more abundant, while myCAFs have a higher abundance in late-stage cancer. When FAP+ CAFs were depleted, there was an increase in mouse survival. Conversely, when αSMA+ CAFs were depleted, there was a decrease in survival. In the TME, when FAP+ CAFs were depleted, there was a decrease in macrophages and B cells. However, αSMA+ CAF loss showed a decrease in effector T cells (Teff) and increased Tregs. A loss in IL-6 production in FAP+ CAFs increased responses in mice to gemcitabine therapy and combination therapy of gemcitabine + checkpoint inhibitors[57].

THE ADENOSINE PATHWAY: A NEW APPROACH TO OVERCOMING THERAPEUTIC RESISTANCE TO CHECKPOINT INHIBITORS

Understanding the interaction between cells in the TME is crucial to overcoming therapeutic resistance. The concept of targeting ADO biosynthesis or inhibiting its receptors has garnered increased interest from the scientific community[58]. Targeting extracellular ADO and its receptors opens opportunities for increasing the antitumor immune response in innate and adaptive immune cell populations normally suppressed within the TME[59]. Since high ADO levels in the TME are predictive of immunosuppressive responses[60], combining current immunotherapies with ADO blockade may help to overcome ICI resistance in solid tumors.

Chemotherapies and various cancer treatments result in elevated cell death and heightened ATP release[61]. ATP is rapidly converted to ADO within solid tumors. This process is mediated by ectonucleotidases CD39 and CD73 and contributes to the formation of an immunosuppressive TME[62]. CD39 acts on ATP to produce adenosine monophosphate (AMP), which is subsequently converted into ADO by CD73. The resulting extracellular ADO interacts with one of four G-protein-coupled receptors (A1R, A2AR, A2BR, and A3R) found in tumor cells, immune cells, and endothelial cells. This increase in ADO levels within the TME hinders the activity of effector immune cells and promotes the expansion of immunosuppressive regulatory T cells[63]. Exosomes released into the TME during cell death also express CD73 and CD39[62].

The A2AR is associated with elevated levels of checkpoint molecules like PD-1, CTLA4, and LAG-3 on T cells[64]. Activation of this receptor tends to inhibit the antitumor functions of macrophages and the proliferation and cytokine production of cytotoxic T cells[65]. However, increased expression of CD39 and CD73 within the TME leads to an upsurge in MDSCs and Tregs[59].

The influence of adenosine receptor expression in key immune cells

Innate immunity: the role of adenosine receptors in macrophages, dendritic cells, and natural killer cells

Innate immunity plays a critical role in the body’s defense against cancer by providing the first line of protection against malignant cells [Figure 1]. This system, comprising various immune cells such as NKs, TAMs, DCs, and soluble factors like cytokines and chemokines, acts to identify and eliminate transformed cells. Recent studies have highlighted the dynamic interplay between innate immunity and cancer progression, shedding light on the delicate balance between tumor-promoting and tumor-suppressing functions of the innate immune system. An essential aspect of the innate immune system’s interaction with cancer cells involves the A2BR, which modulates immune cell functions and significantly impacts the balance between tumor-promoting and tumor-suppressing activities.

Unlocking antitumor immunity with adenosine receptor blockers

Figure 1. Adenosine’s pleiotropic effects on immune cells. ADO facilitates the evasion of tumor cells from immune detection by restricting the activity of T cells, DCs, NK cells, macrophages, and neutrophils. Concurrently, adenosine amplifies the functionality of immunosuppressive cell types like MDSCs and Tregs. ADO: Adenosine; A2AR: A2A receptors; A2BR: A2B receptors; DCs: dendritic cells; MDSCs: myeloid-derived suppressor cells; NK: natural killer; TNFα: tumor necrosis factor-alpha; Tregs: regulatory T cells; VEGF: vascular endothelial growth factor.

Tumor-associated macrophages

TAMs exhibit substantial plasticity that can play a part in tumor progression and drug resistance[66]. The two main classes of TAMs within the TME are activated M1 and alternatively activated M2[67]. M1 is known to be the proinflammatory subset within the tumor, while M2 is considered to be suppressive. However, the classes are not static; the cells can change their state based on the cytokines present. There are also subsets of M2 within the TME, and each class plays a role in tumor formation and progression[67,68].

The TME regulates M1 and M2 macrophages to regulate the immune response to tumors. TAM precursors are derived from embryogenic or bone marrow-derived monocytes[66]. TAMs tend to differentiate primarily into M2-like phenotypes. These cells express high levels of VEGF (pro-angiogenic), mannose receptor (CD206), and scavenger receptor (CD163). They release suppressive cytokines such as IL-10 and promote immunosuppression within the tumor. M1 plays a key role in vaso-proliferation through the secretion of inflammatory cytokines such as IL-6, IL-8, TNF-α, and IL-1β[67,69,70]. The presence of TAMs with high levels of IL-1β within the TME contributes to neovascularization and is a predictor[69].

Apoptotic cells phagocytosed by TAMs (aka efferocytosis) release ATPs into the extracellular space in tissues[71]. ATP derivatives, specifically ADO, affect the immune activation of TAM through ADO receptors[71-73]. The A2BR is upregulated on TAMs in response to interferon-gamma (IFN-γ). When activated, A2BR suppresses the production of TNFα in infiltrating TAMs, inhibiting their capacity to secrete cytokines that are crucial for antitumor immunity. This process ultimately promotes tumor growth[71].

Dendritic cells

DCs are important APCs that present antigens to T cells on MHC proteins. T cells that recognize self-proteins abundant in tumors do not survive selection in the thymus. Intra-tumoral injections of DCs that initiate CD8+ T cell activation have been used to increase responses to immune checkpoint blockade immunotherapies[74-76]. Damage-associated molecular patterns (DAMPs) are used to determine if an immune response needs to be stimulated or if there is immune tolerance to that antigen[74,77]. DAMPs initiate a CD8+ T cell response within tumors but can also help re-prime effector CD8+ T cells to continue the adaptive immune response. However, when tumor cells overcome immune surveillance, DCs may have altered antigen processing and defective T cell activation[78].

Within the TME, there are numerous subsets of DCs along with migratory/tissue-resident DCs[79]. Classical DCs are derived from common myeloid progenitors that differentiate into common DC progenitors. Plasmacytoid DCs (pDC) are believed to be derived from lymphoid cells but can also be derived from myeloid precursors. Common monocyte precursors differentiate into a third major subtype of DCs[80]. Classical DCs have two major states: type 1 and type 2 (cDC1 and cDC2). cDC1 acts to recognize apoptotic and necrotic cell debris presented on its MHC-I receptors to activate CD8+ effector T cells. Their function helps to drive an antitumor response within the TME[75]. cDC2 are more heterogenous than cDC1 cells in tumors but are believed to play a role in recognizing exogenous tumor antigens and presenting them to CD4+ T cells on MHC-II[81]. Within cDC2 populations, there are two further subtypes: anti-inflammatory (cDC2A) and proinflammatory (cDC2B). Classical DCs are now emerging as a potential target for PD-1/PD-L1 immune checkpoint blockade. It has been demonstrated that the proper functioning of checkpoint blockade requires cis interactions with CD80 and PD-L1, as well as PD-1 and PD-L1, between T cells and the DCs[82,83].

DCs that have differentiated during exposure to ADO display diminished activity. Moreover, these DCs express high levels of angiogenic, immunosuppressive, proinflammatory, and tolerogenic factors, such as cyclooxygenase-2 (COX-2), indoleamine 2,3-dioxygenase (IDO), interleukin-6 (IL-6), interleukin-8 (IL-8), IL-10, TGF-β, and VEGF[84,85]. These DCs depend on the upregulation of A2BR when producing these factors to promote ongoing tumor growth and increased angiogenesis for metastasis[85]. As a result, blocking A2BR can preserve the activity of DCs to present neoantigens to T cells, thereby facilitating the process of tumor cell destruction.

NK cells

NKs are derived from CD34+ hematopoietic stem cell progenitors in bone marrow. These cells kill targets that express either no or extremely low MHC-I on their surface[86,87]. NK cells have a specific killer immunoglobulin-like receptor (KIR) on their surface that recognizes MHC-I molecules. When the KIRs recognize MHC-I in self-cells, NK cells are downregulated to prevent an immune response[86,88,89]. Tumor cells tend to have low MHC-I, which can stimulate suppressed NK cells to become activated[90]. NK cells often recognize specific cancer ligands upregulated within tumors, such as MHC-I polypeptide-related sequence A (MICA), MICB, UL-16 binding proteins, complement factor P, and platelet-derived growth factor DD[91-93].

NK cells may have an important role in tumor immunosurveillance. In some cancers, such as colon cancer and gastrointestinal stromal tumors, low NK cell expression is associated with poor outcomes[94,95]. NK cells can also kill circulating tumor cells that are implicated in metastasis[96]. Tumors may become resistant to NK cell effects by suppressing immune cell activation. Like other immune cells, the hypoxic and low-nutrient environment in tumors can decrease NK cell activation[97]. Tumors increase the interactions of activating ligands with their receptors on NK cells to stimulate later resistance to the cells. This increased stimulation can suppress the NK cell function[98].

When NK cells are activated and encounter adenosine through the A2BR, it triggers the cyclic adenosine monophosphate (cAMP) pathway. This activation subsequently blocks cytotoxic activity and cytokine production, diminishing antitumor activity[99,100]. NK cells can also increase CD73 on their surface after encountering mesenchymal stromal cells, thereby contributing to an increase in ADO and tumor growth[101]. This increase in ADO associated with high levels of A2AR contributes to suppressing NK antitumor function[102,103].

Adaptative immunity - the influence of adenosine receptors on t cell function

Adaptive immunity is the second line of defense in the immune system during infection or cancer. These immune responses are cytotoxic to tumor cells. The main two tumor-infiltrating lymphocyte (TIL) cell populations that mediate adaptive immunity are T cells (CD4+ and CD8+) and B cells. T cell infiltration within tumors depends on the tumor chemokine profile and how easily immune cells can enter through the tumor extracellular matrix. Over the last decade, improved methods have been developed to engineer T cells to be better at avoiding cancer immunosuppression. These techniques have resulted in clinical trials of blood-derived tumors and some sarcomas. Very little progress has been made for solid tumors. However, targeting the A2AR on T cells may help overcome the difficulties of T cell immunotherapies in solid tumors. Since ADO causes an increase in Tregs among infiltrating T cells, blocking A2AR can help maintain a high amount of CD4+ T cells in solid tumors. Relative to T cells, B cells have very low levels of A2BR, and limited studies have investigated its role in A2BR activation from ADO[104]. A2AR are more abundant in human than mouse B cells, but their role in immunotherapy is unknown.

T cells

T cells have been the main target of immunotherapy. CAR T cells and immune checkpoint inhibitors have been used to enhance T cell-mediated tumor killing. Dangaj et al. demonstrated that CCL5 must be present within the TME for TILs to enter solid tumors. The macrophages and DCs within the tumor also need to produce CXCL9 to aid in T cell infiltration[105]. Anti-PD-1 therapies have only shown limited success in solid tumors. In metastatic head and neck squamous cell carcinoma, only 15% of patients responded to anti-PD-1 treatment, and very few responses have been seen in microsatellite-stable colorectal cancer[106,107]. Duhen et al. discovered a subset of CD4+ T cells in tumors that are double positive for PD-1 and inducible costimulator (ICOS)[106]. These cells can have a tumor tissue-resident phenotype that allows them to recognize both tumor antigens and neoantigens on MHC-II. CD8+ TILs, on the other hand, are more heterogeneous in their response to tumor antigens[108]. The presence of PD-1 and ICOS on CD4+ T cells may work in conjunction with CD8+ T cells to stimulate a robust antitumor response.

Co-expression of CD39 and CD103 on CD8+ TIL within solid tumors has shown promise in targeting tumor cells. CD8+ T cells that have high expression of CD39 and CD103 can be identified in both primary tumors and metastatic sites but not in the periphery[109]. The level of CD39 and CD103 double positive (DP) cells determines how well patients will respond to immunotherapy[110,111]. However, tumors can still escape these DP TILs through exhaustion mechanisms. All DP TILs express high levels of PD-1 and other exhaustion markers[112]. Checkpoint inhibitors may be useful, but there are also additional ADO pathways linked to CD39+ cells. Blocking the ADO pathway and using immune checkpoint inhibitors may help keep the DP CD8+ T cells active and prevent tumor growth.

Tumor cells increase their expression of CD39 to suppress both CD4+ and CD8+ T cell proliferation and cytotoxicity in the TME[100]. Activation of A2AR in T cells causes increased CD4+ differentiation into Treg cells. There is also an increase in additional suppressive receptors such as PD-1, LAG-3, CTLA4, and T cell immunoglobulin and mucin domain-containing protein 3 (TIM3) on the T cells. Increases in ADO may have a negative effect on immunotherapies when checkpoint inhibitors are given to patients alone[113].

The importance of ADO biosynthesis in the TME

Extracellular ADO is found at low levels in unstressed tissues[114]. It is produced in response to the breakdown of adenine nucleotides and AMP outside injured cells [Figure 2][115]. In response to cancer initiation, ADO levels rapidly increase within tissues due to hypoxic, inflammatory, and/or ischemic conditions. Stressed cells release ATP into the extracellular space as a distress signal that transiently signals via P2 purinergic receptors. Ectoenzymes CD39 and CD73 can rapidly break down extracellular ATP on cell surfaces to produce extracellular ADO[116,117]. Initially, CD39 converts ATP into adenosine diphosphate (ADP) and AMP, followed by CD73-mediated conversion of AMP into ADO[118]. The accumulation of ADO in the TME helps create the suppressive niche.

Unlocking antitumor immunity with adenosine receptor blockers

Figure 2. Adenosine Biosynthesis in Inflamed Tissues. The accumulation of extracellular ATP, driven by stress-induced conditions, stimulates extracellular ADO production by the enzymes CD39 and CD73. ADO binds four G protein-coupled receptors, A1, A2A, A2B, and A3. ADO: Adenosine; ADP: adenosine diphosphate; AMP: adenosine monophosphate; ATP: adenosine triphosphate.

Inhibition of critical immune mechanisms stimulates the formation of the pro-adenosine niche and fibrotic remodeling

The formation of solid tumors in tissues begins with an increase in cell death, inflammation, and hypoxia. This leads to an increase in extracellular ATP and ADO within the TME. When the proinflammatory metabolite extracellular ATP is cleaved into extracellular ADO and is recognized by the A2AR and A2BR within tumors, there is suppression of immune functions[59]. Endothelial cells within the forming tumor and infiltrating immune cells express CD39 and CD73 on their surface. This allows for an increase in ADO within the TME. Endothelial and immune cells also express A2BR on their surface, and when activated by ADO, the tumor can suppress immune cell infiltration. Solid tumors become hypoxic, which feeds back to increase ATP, CD73, and CD39 in the TME to further suppress the immune infiltration[115]. Tumor cells interact with suppressive immune cells to increase A2BR expression, leading to metastasis, proliferation, and VEGF production[119].

CAF increases within solid tumors, forming a dense tumor stroma. These CAFs express high levels of CD39 and CD73 on their surface in various solid tumors such as ovarian, pancreatic, colorectal, and breast cancer, which contribute to ADO production[120-122]. A dense fibrotic stroma allows ADO to remain in high concentration to drive immunosuppressive signaling throughout the tumor. An increase in A2BR on CAFs increases the secretion of IL-6 into the TME, which can convert epithelial cells to a more mesenchymal phenotype[63]. This remodeling of the TME leads to increased metastasis and therapy resistance.

Increases in the ADO pathway cause resistance to immune checkpoint inhibitor therapies

Immune checkpoint inhibitors such as anti-PD-1 and anti-CTLA4 have shown great promise for improving the survival of patients with solid tumors. This form of therapy targets PD-1 and CTLA4 on CD8+ T cells. Tumor cells inhibit CD8+ T cell function by targeting these checkpoint molecules. By blocking PD-1 and CTLA4 from being recognized, the cytotoxic function of the CD8+ T cells is increased to clear the tumor cells[113]. However, patients with solid tumors tend to relapse and become resistant to checkpoint therapy. Maj et al. discovered that when checkpoint therapy is given, there is an increase in the death of cancer cells and Tregs. The sudden death of these cells releases a high amount of ATP in the TME, which is then converted to adenosine by CD39 and CD73. This increase in ADO in the TME counteracts checkpoint therapy and suppresses the antitumor immune response[123].

While an increase in CD73 and CD39 is associated with poor prognosis in patients, increasing A2AR and A2BR expression also contributes to an increased risk of resistance to checkpoint therapy. It has been found that having an increase in A2AR in non-small cell lung cancer (NSCLC) or A2BR in triple-negative breast cancer (TNBC) contributes to poor survival[119,124]. Chalmin et al. showed that the adenosine pathway is involved in resistance to anti-PD-1 therapies. They demonstrated that when patients were given checkpoint therapy, there was an increase in CD73, leading to resistance[125]. Combination therapies targeting CD73, and checkpoint inhibitors may help overcome early resistance in solid tumors. Studies have shown that targeting CD73 and PD-1 in murine colon tumors can inhibit tumor growth[126]. The idea that targeting both the adenosine pathway and checkpoint inhibitors may overcome resistance in solid tumors gives promise to advancing immunotherapy.

Endothelial cells increase CD39 and CD73 levels during hypoxia in tumors, leading to angiogenesis

Hypoxia occurs in solid tumors and suppresses immune cell infiltration by activating hypoxia-inducible factor 1/2 (HIF1/2), IL-6, TGFβ, and TNF. Under these conditions, endothelial, tumor, and various suppressive immune cells increase CD73 and CD39 to increase ATP conversion to AMP and ADO[39,127]. Tumor cells that increase CD73 expression can generate ADO to interact with A2ARs on the tumor cells to stimulate an increase in VEGF secretion[128]. VEGF works to increase angiogenesis within the tumor and provides nutrients and oxygen for growth and metastasis [Figure 3].

Unlocking antitumor immunity with adenosine receptor blockers

Figure 3. Hypoxia in the TME. This figure illustrates a region of a tumor devoid of vasculature, leading to an oxygen-starved environment. This hypoxic zone is characterized by low pH and a predominance of lactate. Within this environment, TAMs tend to adopt an M2 phenotype, and Tregs become the predominant T cell population. Hypoxic conditions also encourage the expression of the ectoenzymes CD39 and CD73, leading to a surge in extracellular ADO. This increase in adenosine, in turn, helps to sustain an immunosuppressive environment within the tumor. Note: this illustration does not depict the anatomical structure of the tumor, but rather represents the phenomena occurring at different levels of tumor oxygenation. ADO: Adenosine; A2BR: A2B receptors; NK: natural killer; TAM: tumor-associated macrophage; TME: tumor microenvironment; Tregs: regulatory T cells.

Endothelial cells express CD39 within tumors to degrade ATP and promote increased neovascularization and tumor growth[129]. To have the ADO concentration needed to sustain the tumor-protective endothelial barrier, CD73 and A2BR are needed within the TME[130]. Feng et al. and Sun et al. demonstrated that inhibiting CD39 on solid tumor endothelial cells decreased angiogenesis and tumor growth[129,131]. CD39 on endothelial cells and the vascular is highly expressed within pancreatic and rectal carcinoma. High expression of these cells in these tumors is correlated with early TNM and better survival after tumor resection[132,133]. Studies have shown that having high levels of CD73 in many solid tumors is associated with worse outcomes. High CD73 levels correlate with higher adenosine concentrations in the tumor, leading to a sustained immunosuppressive TME. With a high expression of both CD39 and CD73 within solid tumors, future combination therapies targeting CD39/CD73, PD-1/CTLA4, and A2BR may allow for better survival in patients.

ADO receptors play various roles in cancer growth

An increase in adenosine within the TME allows for immunosuppression that promotes tumor growth. Receptors for ADO on tumor cells, endothelial cells, and immune cells are drivers of tumor growth and metastasis. The ADO G-protein coupled receptors have four subtypes: A1, A2A, A2B, and A3 [Figure 4]. The A1R, A2AR, and A2BR proteins are highly conserved, while the A3R varies among species. These receptors interact with MAPK pathways to promote proliferation. A2AR and A2BR also increase activation of the mTOR and ERK pathways[134]. However, receptor signaling is dependent on the concentration of extracellular ADO. This level of ADO is primarily dictated by ATP and ADP metabolism by CD39 and CD73 on cells to make AMP and then ADO. In the following, we will focus only on A2AR and A2BR, which appear to play major roles in tumor immunosuppression.

Unlocking antitumor immunity with adenosine receptor blockers

Figure 4. Adenosine receptors. Adenosine interacts with four distinct receptors - A1R, A2AR, A2BR, and A3R. Each of these receptors is linked to a G protein-coupled receptor. The A1R and A3R couple with the Gi protein to inhibit Adenylyl Cyclase. In contrast, the G protein coupled to A2AR and A2BR activates Adenylyl Cyclase, leading to an increased formation of intracellular cyclic AMP. A2BRs also couple to Gq which mobilizes Ca2+ upon activation. AMP: Adenosine monophosphate; A2AR: A2A receptors; A2BR: A2B receptors; cAMP: cyclic adenosine monophospha.

A2AR and A2BR increase immunosuppression in solid tumors

Both A2AR and A2BR are involved in ADO’s immunosuppressive functions. A2AR and A2BR are expressed in most immune and tumor cells[59,135]. A2BR on myeloid cells normally has lower expression than A2AR on other immune cells. However, this receptor increases in the presence of pathological responses such as infections or cancer[136]. A2BR upregulation and activation signal macrophages towards the suppressive M2-like phenotype and secrete IL-10 and VEGF to promote angiogenesis and tumor growth[43]. A2ARs on infiltrating T cells within the TME are activated when ADO increases in the surroundings. This activation suppresses the effector CD8+ T cells in the TME while signaling CD4+ T cell differentiation into Tregs[137-139]. IFN-γ production decreases in NK cells when the A2AR on these cells is activated by ADO[99]. Decreases in antitumor cytotoxic cells and cytokines allow for increased suppressive factors that drive tumor growth and resistance to antitumor therapies.

A2BR activation due to TME hypoxia works to maintain the epithelial barrier of the tumor[39]. Maintaining this barrier prevents antitumor immune cells from penetrating the tumor. A2AR and A2BR-mediated immunosuppression also allow for an increase in metastasis amongst solid tumors[58].

A2BR is key for immunosuppression in solid tumors caused by ADO

Since A2BRs are expressed at low levels under normal conditions, this receptor may be key for triggering immunosuppression within solid tumors. The A2BR has the lowest potency for ADO under normal physiological conditions. However, during inflammation and sudden increases in apoptosis, the A2BR is activated to create an immunosuppressive niche. ADO drives immunosuppression in solid tumors by binding to the A2BR on immune and tumor cells. Once activated, the A2BR increases the secretion of VEGF and IL-8 into the TME[140,141]. This secretion from immune cells comes mainly from monocyte-derived immune cells. When activated by ADO and A2BR interactions, these cells contribute major driving factors in tumor immunosuppression. Ben Addi et al. discovered that the A2BR, not the A2AR, on bone marrow-derived DCs decreased the production of IL-12p70 in mice[84]. A2BR knock-out lung carcinoma cells produced lower VEGF levels in this model when stimulated with adenosine than wildtype controls[140].

Hypoxia increases A2BR expression

As described earlier, hypoxia is a hallmark of solid tumors. Recent studies have expanded our understanding of the role of hypoxia in solid tumors, emphasizing its impact on angiogenesis and tumor growth and its influence on other aspects of tumor biology, such as immune evasion and therapy resistance[18,142]. Hypoxia-driven upregulation of A2BRs and ADO signaling contributes to tumor progression, angiogenesis, and immune escape[44,113].

The hypoxic response also modulates the immune system, affecting innate and adaptive immunity. Hypoxia can alter the functions of immune cells such as TAMs, neutrophils, DCs, T cells, and NK cells[18,143]. Hypoxia-driven changes in the TME, for example, can create an immunosuppressive milieu, impairing the ability of immune cells to target and eliminate cancer cells[144]. Novel therapeutic strategies targeting hypoxia and adenosine signaling pathways, including A2BRs, are currently being investigated to improve the efficacy of existing cancer treatments and overcome treatment resistance[14].

The role of ADO and its receptors blockade to overcome resistance

ADO and its receptors are critical in maintaining immunosuppression in the TME, contributing significantly to immunotherapy resistance. Several clinical trials are exploring the potential of ADO receptor blockade as a novel strategy to counteract this resistance.

Several A2AR antagonists are currently being explored in clinical trials [Table 1], and two of the more advanced therapies are discussed further as part of this review: Corvus Pharmaceutical’s ciforadenant and AstraZeneca’s AZD4635[145].

Table 1

List of current clinical trials evaluating A2AR and A2BR antagonists alone or in combination with cancer immunotherapies

DrugsCombinationsClinical trial information
PhaseIndicationsEnrollmentNCT numberCompletion
date
EOS448
(A2AR antagonist)
· EOS-448, a small molecule, combined with pembrolizumab, an anti-PD-1 antibody
· EOS-448 combined with inupadenant, an investigational adenosine A2AR antagonist
· EOS-448 combined with dostarlimab, an anti-PD-1 antibody
· Inupadenant combined with dostarlimab
· EOS-448 combined with inupadenant and dostarlimab
· EOS-448 combined with dostarlimab and standard-of-care chemotherapies in participants with NSCLC
1/2Lung/H&N cancers/Melanoma376NCT050604322024-09
NIR178
(Taminadenant)
· DFF332, a small molecule targeted to HIF-2α
· DFF332 in combination with everolimus, an mTOR inhibitor
· DFF332 in combination with spartalizumab (an anti-PD-1 antibody) plus taminadenant (A2AR antagonist)
1RCC180NCT048957482025-04
TT-10
(A2AR antagonist)
· TT-10, a small molecule, as a single agent1/2Prostate/NSCLC/RCC90NCT049693152025-08
ILB2109
(A2AR antagonist)
· ILB2109, a small molecule, as a single agent1Advanced solid tumor48NCT052785462024-01
AZD4635
(A2AR antagonist)
· AZD4635 as monotherapy
· Combination with durvalumab
· Combination with durvalumab plus oleclumab
· Combination with docetaxel
· Combination with either abiraterone acetate or enzalutamide
1Solid tumor313NCT027409852021-04
AZD4635
(A2AR antagonist)
· AZD4635 with durvalumab, an anti-PDL-1 antibody
· AZD4635 with oleclumab, an anti-CD73 antibody
2Prostate tumor59NCT040895532023-04
CPI-444
(A2AR antagonist)
· CPI-444, a small molecule, in combination with ipilimumab, an anti-CTLA4 antibody
· CPI-444 in combination with nivolumab, an anti-PD-1 antibody
1/2RCC15NCT055010542026-11
CPI-444
(A2AR antagonist)
· CPI-444 (ciforadenant) as a single
· Combination with atezolizumab, a PD-L1 inhibitor
1RCC502NCT026558222021-07
PBF-1129
(A2BR antagonist)
· Combination of adenosine A2BR antagonist PBF-1129 (mAb) and nivolumab, an anti-PD-1 antibody1Metastatic NSCLC30NCT052343072025-12
· PBF-1129 as a single agent1Metastatic NSCLC18NCT032744792023-12
M1069
(Dual A2AR/A2BR antagonist)
· M1069, a small molecule, as a single agent1Unresectable solid tumors30NCT051983492023-12
AB928
(Dual A2AR/A2BR antagonist)
· Combination of SRF617, an anti-CD39 antibody, etrumadenant (AB928), and zimberelimab (AB122), an anti-PD-1 antibody2Prostate cancer15NCT051777702023-04
TT-4
(A2BR antagonist)
· TT-4, a small molecule, as a single agent1/2GI cancers69NCT049766602023-09

In a first-in-human Phase 1 dose-escalation study in patients with advanced refractory cancers (NCT02655822), ciforadenant (either monotherapy or in combination with atezolizumab) was administered to 502 patients. Of those, a cohort of 68 renal cell carcinoma (RCC) patients yielded clinical responses, including partial responses (PR) in 11% of patients treated with a combination of A2AR antagonists and anti-PD-L1 antibodies, and in 3% of patients treated with A2AR antagonists alone[146]. Further, tumor regression was observed in an additional 24% of patients, although the regression was not significant enough to be classified as PR by RECIST criteria. These findings are noteworthy, especially considering that the patients involved in the study were not only resistant to PD-1 blockade but were also deemed untreatable prior to the trial.

According to Michail Sitkovsky[147], the observed tumor regressions in patients with RCC, who were previously untreatable and refractory to PD-1 blockade, likely occurred in patients meeting specific criteria: their tumors were immunogenic, developed tumor-reactive effector T cells, retained a significant number of effector cells post-toxic cancer chemotherapies, and were protected by immunosuppressive extracellular ADO to A2AR signaling. Ciforadenant appears to have facilitated the invasion and tumor-rejecting functions of T and NK cells in these patients; however, the levels of antitumor immunity in responsive patients were not high enough to achieve a complete response. The major limitation appears to be the lack or low numbers of tumor-reactive T and NK cells in refractory patients, either due to the tumor’s poor immunogenicity or past toxic chemotherapies.

A2AR antagonists are anticipated to be most efficacious in patients with sufficient aggressive, multifunctional tumor-reactive T cells. Without these cells, it could be expected that A2AR antagonists would only have antitumor effects when combined with cancer vaccines or T-cell transfers that increase the number of tumor-reactive T cells. Future treatments combining A2AR antagonism with adoptive cell transfer (ACT) are promising, especially for refractory patients, as ACT ensures the presence of sufficient T-cells and NK-cells in patients, enhancing the potential for A2AR antagonism as an immunotherapy.

These antagonists have been able to show, both in vitro and in vivo, that blocking the adenosine pathway at the A2AR increases cytotoxic T cells within the TME, increases cytokine production, and reverses T cell inhibition.

Another A2AR antagonist in development is AstraZeneca’s AZD4635. In a monotherapy phase 1 trial (NCT02740985), observed adverse events included nausea, fatigue, and vomiting. In addition, one patient with colorectal cancer had sudden death 15 days after the last dose of AZD4635, which was considered treatment-related by the investigator. However, AZD4635 was well tolerated both as a monotherapy and in combination with durvalumab in all patients. There were patients with responses such as stable disease, partial response, and complete response and RNA analysis confirmed that in 5 of 7 patients, intertumoral adenosine signaling decreased. Four of these 7 patients also had increases in gene-expression signatures of cytolytic activity and IFN-γ signaling. These findings suggest that there were observable positive responses to the treatment in some patients.

A2BR antagonists are also under investigation in several clinical trials. Arcus’ etrumadenant, a dual A2AR and A2BR antagonist, is being evaluated in several cancers and was recently discontinued in mCRPC due to a lack of efficacy (NCT05177770). Palofarma’s PBF-1129, an A2BR antagonist, is being evaluated in metastatic NSCLC (NCT03274479) and EMD Serono’s M1069 (NCT05198349), another A2AR and A2BR antagonist, is currently undergoing a first-in-human trial in patients with advanced malignancies. In addition, Portage is evaluating TT-4, an A2BR antagonist, as a single agent in gastrointestinal cancer (NCT04976660). Further investigation will likely provide more insights into the clinical potential of these promising strategies. Overall, compounds exhibiting the highest water solubility tend to possess increased bioavailability, making them more effective[148]. With the advancement in the development of ADO receptor antagonists, enhancing the solubility of these promising compounds while preserving their selectivity emerges as an avenue for improvement.

CONCLUSION

In the complex battleground of cancer, it is necessary to understand the adaptations tumors employ to resist therapies. This review has emphasized the role of ADO, a significant player in the TME, in driving immunosuppression and fostering cancer drug resistance. The importance of ADO and its receptors, particularly the A2AR and A2BR subtypes, in promoting an immune-escaping environment was thoroughly explored.

Current research endeavors focus on various approaches to counteract immunosuppression, including monoclonal antibodies against CD73 and the blockade of ADO receptors. Ongoing clinical trials investigate combinations of these approaches with existing therapies, aiming to stimulate immune responses and improve patient outcomes.

The results of ongoing clinical trials will inform new ways of overcoming cancer drug resistance. However, further research is required to understand and fully exploit ADO’s pathway. Targeting ADO could improve cancer treatments, providing hope for patients who previously had limited treatment options. This underscores the importance of ongoing research in this area, aiming to improve the prognosis for all cancer patients.

DECLARATIONS

Acknowledgments

The authors thank Schuyler Vinzant and Sara Adair for their critical feedback on the manuscript. All figures were created using Biorender.

Authors’ contributions

Made substantial contributions to the conception, outline, and writing: Remley VA, Dimastromatteo J

Performed critical review/editing for scientific accuracy: Linden J, Bauer TW

Availability of data and materials

Not applicable.

Financial support and sponsorship

None.

Conflicts of interest

Drs. Dimastromatteo and Linden are members of Adovate, a for-profit company dedicated to improving patient health using a new generation of adenosine analogs.

Ethical approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Copyright

© The Author(s) 2023.

REFERENCES

1. Fares CM, Van Allen EM, Drake CG, Allison JP, Hu-Lieskovan S. Mechanisms of resistance to immune checkpoint blockade: why does checkpoint inhibitor immunotherapy not work for all patients? Am Soc Clin Oncol Educ Book 2019;39:147-64.

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

3. Galon J, Bruni D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat Rev Drug Discov 2019;18:197-218.

4. Rubin SJS, Sojwal RS, Gubatan J, Rogalla S. The tumor immune microenvironment in pancreatic ductal adenocarcinoma: neither hot nor cold. Cancers 2022;14:4236.

5. Johnson A, Townsend M, O’Neill K. Tumor microenvironment immunosuppression: a roadblock to CAR T-cell advancement in solid tumors. Cells 2022;11:3626.

6. Nallanthighal S, Heiserman JP, Cheon DJ. The role of the extracellular matrix in cancer stemness. Front Cell Dev Biol 2019;7:86.

7. Croci DO, Zacarías Fluck MF, Rico MJ, Matar P, Rabinovich GA, Scharovsky OG. Dynamic cross-talk between tumor and immune cells in orchestrating the immunosuppressive network at the tumor microenvironment. Cancer Immunol Immunother 2007;56:1687-700.

8. Huang X, Li Y, Fu M, Xin HB. Polarizing macrophages in vitro. In: Rousselet G, editor. Macrophages. New York, NY: Springer New York; 2018. p. 119-26. Available from: http://link.springer.com/10.1007/978-1-4939-7837-3_12. [Last accessed on 18 Oct 2023].

9. Solinas G, Germano G, Mantovani A, Allavena P. Tumor-associated macrophages (TAM) as major players of the cancer-related inflammation. J Leukoc Biol 2009;86:1065-73.

10. Qian BZ, Pollard JW. Macrophage diversity enhances tumor progression and metastasis. Cell 2010;141:39-51.

11. Facciabene A, Motz GT, Coukos G. T-regulatory cells: key players in tumor immune escape and angiogenesis. Cancer Res 2012;72:2162-71.

12. Choueiry F, Torok M, Shakya R, et al. CD200 promotes immunosuppression in the pancreatic tumor microenvironment. J Immunother Cancer 2020;8:e000189.

13. Shi R, Tang YQ, Miao H. Metabolism in tumor microenvironment: implications for cancer immunotherapy. MedComm 2020;1:47-68.

14. Hatfield SM, Kjaergaard J, Lukashev D, et al. Immunological mechanisms of the antitumor effects of supplemental oxygenation. Sci Transl Med 2015;7:277ra30.

15. Gallina G, Dolcetti L, Serafini P, et al. Tumors induce a subset of inflammatory monocytes with immunosuppressive activity on CD8+ T cells. J Clin Invest 2006;116:2777-90.

16. Zea AH, Rodriguez PC, Atkins MB, et al. Arginase-producing myeloid suppressor cells in renal cell carcinoma patients: a mechanism of tumor evasion. Cancer Res 2005;65:3044-8.

17. Mundy-Bosse BL, Lesinski GB, Jaime-Ramirez AC, et al. Myeloid-derived suppressor cell inhibition of the IFN response in tumor-bearing mice. Cancer Res 2011;71:5101-10.

18. Chouaib S, Noman MZ, Kosmatopoulos K, Curran MA. Hypoxic stress: obstacles and opportunities for innovative immunotherapy of cancer. Oncogene 2017;36:439-45.

19. Corbet C, Feron O. Tumour acidosis: from the passenger to the driver’s seat. Nat Rev Cancer 2017;17:577-93.

20. Colegio OR, Chu NQ, Szabo AL, et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014;513:559-63.

21. Hayes C, Donohoe CL, Davern M, Donlon NE. The oncogenic and clinical implications of lactate induced immunosuppression in the tumour microenvironment. Cancer Lett 2021;500:75-86.

22. Leach DR, Krummel MF, Allison JP. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996;271:1734-6.

23. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11:3887-95.

24. Rizvi NA, Hellmann MD, Snyder A, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124-8.

25. Rizvi H, Sanchez-Vega F, La K, et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J Clin Oncol 2018;36:633-41.

26. Strickland LN, Faraoni EY, Ruan W, Yuan X, Eltzschig HK, Bailey-Lundberg JM. The resurgence of the Adora2b receptor as an immunotherapeutic target in pancreatic cancer. Front Immunol 2023;14:1163585.

27. Rudensky AY. Regulatory T cells and Foxp3. Immunol Rev 2011;241:260-8.

28. Yang L, Huang J, Ren X, et al. Abrogation of TGFβ signaling in mammary carcinomas recruits Gr-1+CD11b+ myeloid cells that promote metastasis. Cancer Cell 2008;13:23-35.

29. Noy R, Pollard JW. Tumor-associated macrophages: from mechanisms to therapy. Immunity 2014;41:49-61.

30. Highfill SL, Cui Y, Giles AJ, et al. Disruption of CXCR2-mediated MDSC tumor trafficking enhances anti-PD1 efficacy. Sci Transl Med 2014;6:237ra67.

31. Najafi M, Farhood B, Mortezaee K. Contribution of regulatory T cells to cancer: a review. J Cell Physiol 2019;234:7983-93.

32. Liu C, Peng W, Xu C, et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin Cancer Res 2013;19:393-403.

33. Semenza GL. Oxygen sensing, homeostasis, and disease. N Engl J Med 2011;365:537-47.

34. Eltzschig HK, Abdulla P, Hoffman E, et al. HIF-1-dependent repression of equilibrative nucleoside transporter (ENT) in hypoxia. J Exp Med 2005;202:1493-505.

35. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 2003;9:677-84.

36. Eckle T, Krahn T, Grenz A, et al. Cardioprotection by ecto-5'-nucleotidase (CD73) and A2B adenosine receptors. Circulation 2007;115:1581-90.

37. Zhang B. CD73 promotes tumor growth and metastasis. Oncoimmunology 2012;1:67-70.

38. Gao ZW, Dong K, Zhang HZ. The roles of CD73 in cancer. Biomed Res Int 2014;2014:460654.

39. Allard B, Longhi MS, Robson SC, Stagg J. The ectonucleotidases CD39 and CD73: novel checkpoint inhibitor targets. Immunol Rev 2017;276:121-44.

40. Beavis PA, Slaney CY, Milenkovski N, et al. CD73: a potential biomarker for anti-PD-1 therapy. Oncoimmunology 2015;4:e1046675.

41. Giannone G, Ghisoni E, Genta S, et al. Immuno-metabolism and microenvironment in cancer: key players for immunotherapy. Int J Mol Sci 2020;21:4414.

42. Linnemann C, Schildberg FA, Schurich A, et al. Adenosine regulates CD8 T-cell priming by inhibition of membrane-proximal T-cell receptor signalling. Immunology 2009;128:e728-37.

43. Csóka B, Selmeczy Z, Koscsó B, et al. Adenosine promotes alternative macrophage activation via A2A and A2B receptors. FASEB J 2012;26:376-86.

44. Antonioli L, Blandizzi C, Pacher P, Haskó G. Immunity, inflammation and cancer: a leading role for adenosine. Nat Rev Cancer 2013;13:842-57.

45. McColl SR, St-Onge M, Dussault AA, et al. Immunomodulatory impact of the A2A adenosine receptor on the profile of chemokines produced by neutrophils. FASEB J 2006;20:187-9.

46. Sahai E, Astsaturov I, Cukierman E, et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer 2020;20:174-86.

47. Fordyce C, Fessenden T, Pickering C, et al. DNA damage drives an activin a-dependent induction of cyclooxygenase-2 in premalignant cells and lesions. Cancer Prev Res 2010;3:190-201.

48. Fordyce CA, Patten KT, Fessenden TB, et al. Cell-extrinsic consequences of epithelial stress: activation of protumorigenic tissue phenotypes. Breast Cancer Res 2012;14:R155.

49. Fearon DT. The carcinoma-associated fibroblast expressing fibroblast activation protein and escape from immune surveillance. Cancer Immunol Res 2014;2:187-93.

50. Albrengues J, Bourget I, Pons C, et al. LIF mediates proinvasive activation of stromal fibroblasts in cancer. Cell Rep 2014;7:1664-78.

51. Albrengues J, Bertero T, Grasset E, et al. Epigenetic switch drives the conversion of fibroblasts into proinvasive cancer-associated fibroblasts. Nat Commun 2015;6:10204.

52. Calvo F, Ege N, Grande-Garcia A, et al. Mechanotransduction and YAP-dependent matrix remodelling is required for the generation and maintenance of cancer-associated fibroblasts. Nat Cell Biol 2013;15:637-46.

53. Strell C, Paulsson J, Jin SB, et al. Impact of epithelial-stromal interactions on peritumoral fibroblasts in ductal carcinoma in situ. J Natl Cancer Inst 2019;111:983-95.

54. Hirata E, Girotti MR, Viros A, et al. Intravital imaging reveals how BRAF inhibition generates drug-tolerant microenvironments with high integrin β1/FAK signaling. Cancer Cell 2015;27:574-88.

55. Hirata E, Sahai E. Tumor microenvironment and differential responses to therapy. Cold Spring Harb Perspect Med 2017;7:a026781.

56. Sun Y, Campisi J, Higano C, et al. Treatment-induced damage to the tumor microenvironment promotes prostate cancer therapy resistance through WNT16B. Nat Med 2012;18:1359-68.

57. McAndrews KM, Chen Y, Darpolor JK, et al. Identification of functional heterogeneity of carcinoma-associated fibroblasts with distinct IL6-mediated therapy resistance in pancreatic cancer. Cancer Discov 2022;12:1580-97.

58. Vigano S, Alatzoglou D, Irving M, et al. Targeting adenosine in cancer immunotherapy to enhance T-cell function. Front Immunol 2019;10:925.

59. Allard B, Allard D, Buisseret L, Stagg J. The adenosine pathway in immuno-oncology. Nat Rev Clin Oncol 2020;17:611-29.

60. Sidders B, Zhang P, Goodwin K, et al. Adenosine signaling is prognostic for cancer outcome and has predictive utility for immunotherapeutic response. Clin Cancer Res 2020;26:2176-87.

61. Ahmed A, Tait SWG. Targeting immunogenic cell death in cancer. Mol Oncol 2020;14:2994-3006.

62. Xia GQ, Cai JN, Wu X, Fang Q, Zhao N, Lv XW. The mechanism by which ATP regulates alcoholic steatohepatitis through P2X4 and CD39. Eur J Pharmacol 2022;916:174729.

63. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosuppressive adenosine in cancer. Nat Rev Cancer 2017;17:765.

64. Marin-Acevedo JA, Kimbrough EO, Lou Y. Next generation of immune checkpoint inhibitors and beyond. J Hematol Oncol 2021;14:45.

65. Sun C, Wang B, Hao S. Adenosine-A2A receptor pathway in cancer immunotherapy. Front Immunol 2022;13:837230.

66. Sylvestre M, Crane CA, Pun SH. Progress on modulating tumor-associated macrophages with biomaterials. Adv Mater 2020;32:e1902007.

67. Gao J, Liang Y, Wang L. Shaping polarization of tumor-associated macrophages in cancer immunotherapy. Front Immunol 2022;13:888713.

68. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004;25:677-86.

69. Nakao S, Kuwano T, Tsutsumi-Miyahara C, et al. Infiltration of COX-2-expressing macrophages is a prerequisite for IL-1 beta-induced neovascularization and tumor growth. J Clin Invest 2005;115:2979-91.

70. Fujimoto J, Aoki I, Khatun S, Toyoki H, Tamaya T. Clinical implications of expression of interleukin-8 related to myometrial invasion with angiogenesis in uterine endometrial cancers. Ann Oncol 2002;13:430-4.

71. Murphy PS, Wang J, Bhagwat SP, et al. CD73 regulates anti-inflammatory signaling between apoptotic cells and endotoxin-conditioned tissue macrophages. Cell Death Differ 2017;24:559-70.

72. Yamaguchi H, Maruyama T, Urade Y, Nagata S. Immunosuppression via adenosine receptor activation by adenosine monophosphate released from apoptotic cells. Elife 2014;3:e02172.

73. Köröskényi K, Duró E, Pallai A, et al. Involvement of adenosine A2A receptors in engulfment-dependent apoptotic cell suppression of inflammation. J Immunol 2011;186:7144-55.

74. Marciscano AE, Anandasabapathy N. The role of dendritic cells in cancer and anti-tumor immunity. Semin Immunol 2021;52:101481.

75. Albert ML, Sauter B, Bhardwaj N. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature 1998;392:86-9.

76. Nirschl CJ, Suárez-Fariñas M, Izar B, et al. IFNγ-dependent tissue-immune homeostasis is co-opted in the tumor microenvironment. Cell 2017;170:127-41.e15.

77. Joffre O, Nolte MA, Spörri R, Reis e Sousa C. Inflammatory signals in dendritic cell activation and the induction of adaptive immunity. Immunol Rev 2009;227:234-47.

78. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002;3:991-8.

79. Gerhard GM, Bill R, Messemaker M, Klein AM, Pittet MJ. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J Exp Med 2021;218:e20200264.

80. Dress RJ, Dutertre CA, Giladi A, et al. Plasmacytoid dendritic cells develop from Ly6D+ lymphoid progenitors distinct from the myeloid lineage. Nat Immunol 2019;20:852-64.

81. Binnewies M, Mujal AM, Pollack JL, et al. Unleashing type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity. Cell 2019;177:556-71.e16.

82. Oh SA, Wu DC, Cheung J, et al. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nat Cancer 2020;1:681-91.

83. Mayoux M, Roller A, Pulko V, et al. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci Transl Med 2020;12:eaav7431.

84. Ben Addi A, Lefort A, Hua X, et al. Modulation of murine dendritic cell function by adenine nucleotides and adenosine: involvement of the A2B receptor. Eur J Immunol 2008;38:1610-20.

85. Novitskiy SV, Ryzhov S, Zaynagetdinov R, et al. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 2008;112:1822-31.

86. Shimasaki N, Coustan-Smith E, Kamiya T, Campana D. Expanded and armed natural killer cells for cancer treatment. Cytotherapy 2016;18:1422-34.

87. Kärre K, Ljunggren HG, Piontek G, Kiessling R. Selective rejection of H-2-deficient lymphoma variants suggests alternative immune defence strategy. Nature 1986;319:675-8.

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

89. Guillerey C, Huntington ND, Smyth MJ. Targeting natural killer cells in cancer immunotherapy. Nat Immunol 2016;17:1025-36.

90. Raulet DH, Vance RE. Self-tolerance of natural killer cells. Nat Rev Immunol 2006;6:520-31.

91. Bauer S, Groh V, Wu J, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science 1999;285:727-9.

92. Gasser S, Orsulic S, Brown EJ, Raulet DH. The DNA damage pathway regulates innate immune system ligands of the NKG2D receptor. Nature 2005;436:1186-90.

93. Barrow AD, Edeling MA, Trifonov V, et al. Natural killer cells control tumor growth by sensing a growth factor. Cell 2018;172:534-48.e19.

94. Delahaye NF, Rusakiewicz S, Martins I, et al. Alternatively spliced NKp30 isoforms affect the prognosis of gastrointestinal stromal tumors. Nat Med 2011;17:700-7.

95. Mlecnik B, Bindea G, Angell HK, et al. Functional network pipeline reveals genetic determinants associated with in situ lymphocyte proliferation and survival of cancer patients. Sci Transl Med 2014;6:228ra37.

96. López-Soto A, Gonzalez S, Smyth MJ, Galluzzi L. Control of metastasis by NK cells. Cancer Cell 2017;32:135-54.

97. O’Brien KL, Finlay DK. Immunometabolism and natural killer cell responses. Nat Rev Immunol 2019;19:282-90.

98. Coudert JD, Zimmer J, Tomasello E, et al. Altered NKG2D function in NK cells induced by chronic exposure to NKG2D ligand-expressing tumor cells. Blood 2005;106:1711-7.

99. Raskovalova T, Huang X, Sitkovsky M, Zacharia LC, Jackson EK, Gorelik E. Gs protein-coupled adenosine receptor signaling and lytic function of activated NK cells. J Immunol 2005;175:4383-91.

100. Bastid J, Regairaz A, Bonnefoy N, et al. Inhibition of CD39 enzymatic function at the surface of tumor cells alleviates their immunosuppressive activity. Cancer Immunol Res 2015;3:254-65.

101. Chatterjee D, Tufa DM, Baehre H, Hass R, Schmidt RE, Jacobs R. Natural killer cells acquire CD73 expression upon exposure to mesenchymal stem cells. Blood 2014;123:594-5.

102. Wallace KL, Linden J. Adenosine A2A receptors induced on iNKT and NK cells reduce pulmonary inflammation and injury in mice with sickle cell disease. Blood 2010;116:5010-20.

103. Beavis PA, Divisekera U, Paget C, et al. Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci U S A 2013;110:14711-6.

104. Burnstock G, Boeynaems JM. Purinergic signalling and immune cells. Purinergic Signal 2014;10:529-64.

105. Dangaj D, Bruand M, Grimm AJ, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors. Cancer Cell 2019;35:885-900.e10.

106. Duhen R, Fesneau O, Samson KA, et al. PD-1 and ICOS coexpression identifies tumor-reactive CD4+ T cells in human solid tumors. J Clin Invest 2022;132:e156821.

107. Addeo R, Ghiani M, Merlino F, Ricciardiello F, Caraglia M. CheckMate 141 trial: all that glitters is not gold. Expert Opin Biol Ther 2019;19:169-71.

108. van den Bulk J, Verdegaal EME, Ruano D, et al. Neoantigen-specific immunity in low mutation burden colorectal cancers of the consensus molecular subtype 4. Genome Med 2019;11:87.

109. Duhen T, Duhen R, Montler R, et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat Commun 2018;9:2724.

110. Aparicio T. PD-1 blockade in tumors with mismatch-repair deficiency. Colon Rectum 2015;9:182-4.

111. Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409-13.

112. Pauken KE, Wherry EJ. Overcoming T cell exhaustion in infection and cancer. Trends Immunol 2015;36:265-76.

113. Young A, Mittal D, Stagg J, Smyth MJ. Targeting cancer-derived adenosine: new therapeutic approaches. Cancer Discov 2014;4:879-88.

114. Antonioli L, Fornai M, Blandizzi C, Pacher P, Haskó G. Adenosine signaling and the immune system: when a lot could be too much. Immunol Lett 2019;205:9-15.

115. Fredholm BB. Adenosine, an endogenous distress signal, modulates tissue damage and repair. Cell Death Differ 2007;14:1315-23.

116. Robson SC, Wu Y, Sun X, Knosalla C, Dwyer K, Enjyoji K. Ectonucleotidases of CD39 family modulate vascular inflammation and thrombosis in transplantation. Semin Thromb Hemost 2005;31:217-33.

117. Picher M, Burch LH, Hirsh AJ, Spychala J, Boucher RC. Ecto 5'-nucleotidase and nonspecific alkaline phosphatase. Two AMP-hydrolyzing ectoenzymes with distinct roles in human airways. J Biol Chem 2003;278:13468-79.

118. Yegutkin GG. Nucleotide- and nucleoside-converting ectoenzymes: important modulators of purinergic signalling cascade. Biochim Biophys Acta 2008;1783:673-94.

119. Mittal D, Sinha D, Barkauskas D, et al. Adenosine 2B receptor expression on cancer cells promotes metastasis. Cancer Res 2016;76:4372-82.

120. Costa A, Kieffer Y, Scholer-Dahirel A, et al. Fibroblast heterogeneity and immunosuppressive environment in human breast cancer. Cancer Cell 2018;33:463-79.e10.

121. Turcotte M, Spring K, Pommey S, et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res 2015;75:4494-503.

122. Yu M, Guo G, Huang L, et al. CD73 on cancer-associated fibroblasts enhanced by the A2B-mediated feedforward circuit enforces an immune checkpoint. Nat Commun 2020;11:515.

123. Maj T, Wang W, Crespo J, et al. Oxidative stress controls regulatory T cell apoptosis and suppressor activity and PD-L1-blockade resistance in tumor. Nat Immunol 2017;18:1332-41.

124. Limagne E, Euvrard R, Thibaudin M, et al. Accumulation of MDSC and Th17 cells in patients with metastatic colorectal cancer predicts the efficacy of a FOLFOX-bevacizumab drug treatment regimen. Cancer Res 2016;76:5241-52.

125. Chalmin F, Mignot G, Bruchard M, et al. Stat3 and Gfi-1 transcription factors control Th17 cell immunosuppressive activity via the regulation of ectonucleotidase expression. Immunity 2012;36:362-73.

126. Hay CM, Sult E, Huang Q, et al. Targeting CD73 in the tumor microenvironment with MEDI9447. Oncoimmunology 2016;5:e1208875.

127. Allard B, Beavis PA, Darcy PK, Stagg J. Immunosuppressive activities of adenosine in cancer. Curr Opin Pharmacol 2016;29:7-16.

128. Allard B, Turcotte M, Spring K, Pommey S, Royal I, Stagg J. Anti-CD73 therapy impairs tumor angiogenesis. Int J Cancer 2014;134:1466-73.

129. Feng L, Sun X, Csizmadia E, et al. Vascular CD39/ENTPD1 directly promotes tumor cell growth by scavenging extracellular adenosine triphosphate. Neoplasia 2011;13:206-16.

130. Jackson SW, Hoshi T, Wu Y, et al. Disordered purinergic signaling inhibits pathological angiogenesis in cd39/Entpd1-null mice. Am J Pathol 2007;171:1395-404.

131. Sun X, Wu Y, Gao W, et al. CD39/ENTPD1 expression by CD4+Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology 2010;139:1030-40.

132. Künzli BM, Berberat PO, Giese T, et al. Upregulation of CD39/NTPDases and P2 receptors in human pancreatic disease. Am J Physiol Gastrointest Liver Physiol 2007;292:G223-30.

133. Zhang B, Cheng B, Li FS, et al. High expression of CD39/ENTPD1 in malignant epithelial cells of human rectal adenocarcinoma. Tumour Biol 2015;36:9411-9.

134. Borea PA, Gessi 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;37:419-34.

135. Huang Y, Gu Z, Fan Y, et al. Inhibition of the adenosinergic pathway: the indispensable part of oncological therapy in the future. Purinergic Signal 2019;15:53-67.

136. Vecchio EA, White PJ, May LT. The adenosine A2B G protein-coupled receptor: recent advances and therapeutic implications. Pharmacol Ther 2019;198:20-33.

137. 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.

138. Leone RD, Sun IM, Oh MH, et al. Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother 2018;67:1271-84.

139. Zarek PE, Huang CT, Lutz ER, et al. A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood 2008;111:251-9.

140. Ryzhov S, Novitskiy SV, Zaynagetdinov R, et al. Host A2B adenosine receptors promote carcinoma growth. Neoplasia 2008;10:987-95.

141. Merighi S, Benini A, Mirandola P, et al. Caffeine inhibits adenosine-induced accumulation of hypoxia-inducible factor-1α, vascular endothelial growth factor, and interleukin-8 expression in hypoxic human colon cancer cells. Mol Pharmacol 2007;72:395-406.

142. Wilson JM, Kurtz CC, Black SG, et al. The A2B adenosine receptor promotes Th17 differentiation via stimulation of dendritic cell IL-6. J Immunol 2011;186:6746-52.

143. Taylor CT, Colgan SP. Regulation of immunity and inflammation by hypoxia in immunological niches. Nat Rev Immunol 2017;17:774-85.

144. Palazon A, Goldrath AW, Nizet V, Johnson RS. HIF transcription factors, inflammation, and immunity. Immunity 2014;41:518-28.

145. Xia C, Yin S, To KKW, Fu L. CD39/CD73/A2AR pathway and cancer immunotherapy. Mol Cancer 2023;22:44.

146. Fong L, Hotson A, Powderly JD, et al. Adenosine 2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov 2020;10:40-53.

147. Sitkovsky MV. Lessons from the A2A adenosine receptor antagonist-enabled tumor regression and survival in patients with treatment-refractory renal cell cancer. Cancer Discov 2020;10:16-9.

148. Bhalani DV, Nutan B, Kumar A, Singh Chandel AK. Bioavailability enhancement techniques for poorly aqueous soluble drugs and therapeutics. Biomedicines 2022;10:2055.

Cite This Article

Export citation file: BibTeX | RIS

OAE Style

Remley VA, Linden J, Bauer TW, Dimastromatteo J. Unlocking antitumor immunity with adenosine receptor blockers. Cancer Drug Resist 2023;6:748-67. http://dx.doi.org/10.20517/cdr.2023.63

AMA Style

Remley VA, Linden J, Bauer TW, Dimastromatteo J. Unlocking antitumor immunity with adenosine receptor blockers. Cancer Drug Resistance. 2023; 6(4): 748-67. http://dx.doi.org/10.20517/cdr.2023.63

Chicago/Turabian Style

Remley, Victoria A., Joel Linden, Todd W. Bauer, Julien Dimastromatteo. 2023. "Unlocking antitumor immunity with adenosine receptor blockers" Cancer Drug Resistance. 6, no.4: 748-67. http://dx.doi.org/10.20517/cdr.2023.63

ACS Style

Remley, VA.; Linden J.; Bauer TW.; Dimastromatteo J. Unlocking antitumor immunity with adenosine receptor blockers. Cancer Drug Resist. 2023, 6, 748-67. http://dx.doi.org/10.20517/cdr.2023.63

About This Article

Special Issue

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

Data & Comments

Data

Views
971
Downloads
241
Citations
0
Comments
0
12

Comments

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

0
Download PDF
Cite This Article 10 clicks
Like This Article 12 likes
Share This Article
Scan the QR code for reading!
See Updates
Contents
Figures
Related
Cancer Drug Resistance
ISSN 2578-532X (Online)

Portico

All published articles will preserved here permanently:

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

Portico

All published articles will preserved here permanently:

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