Skip to main content
The EMBO Journal logoLink to The EMBO Journal
. 2021 Jun 14;40(13):e108130. doi: 10.15252/embj.2021108130

ATP and cancer immunosurveillance

Oliver Kepp 1,2, Lucillia Bezu 1,2, Takahiro Yamazaki 3, Francesco Di Virgilio 4, Mark J Smyth 5, Guido Kroemer 1,2,6,7,8,†,, Lorenzo Galluzzi 3,9,10,11,12,†,
PMCID: PMC8246257  PMID: 34121201

Abstract

While intracellular adenosine triphosphate (ATP) occupies a key position in the bioenergetic metabolism of all the cellular compartments that form the tumor microenvironment (TME), extracellular ATP operates as a potent signal transducer. The net effects of purinergic signaling on the biology of the TME depend not only on the specific receptors and cell types involved, but also on the activation status of cis‐ and trans‐regulatory circuitries. As an additional layer of complexity, extracellular ATP is rapidly catabolized by ectonucleotidases, culminating in the accumulation of metabolites that mediate distinct biological effects. Here, we discuss the molecular and cellular mechanisms through which ATP and its degradation products influence cancer immunosurveillance, with a focus on therapeutically targetable circuitries.

Keywords: ADORA2A, autophagy, CD39, CD73, immune checkpoint inhibitors, immunogenic cell death

Subject Categories: Cancer, Immunology, Metabolism


This review summarizes our current view on the dual roles of signaling via adenosine triphosphate and its degradation products in the tumor microenvironment, affecting intracellular metabolism as well as crosstalk between malignant and immune cells.

graphic file with name EMBJ-40-e108130-g004.jpg

Introduction

According to a widely accepted model, malignant transformation is initiated by relatively common genetic or epigenetic alterations that incapacitate tumor‐suppressing mechanisms (for the most part, mechanisms that ensure the preservation of cellular homeostasis) as they activate oncogenic drivers (generally, processes that enable accrued anabolism in support of hyperproliferation) (Hanahan & Weinberg, 2011; Timp & Feinberg, 2013). The vast majority of newly formed malignant cells, however, appears to be controlled by the host immune system prior to forming symptomatic tumors (Vesely & Schreiber, 2013; Lopez‐Otin & Kroemer, 2021). While in most cases such control involves the definitive “eradication” of malignant cell precursors, in some instances newly formed cancer cells can resist immune attacks and generate a rudimentary tumor microenvironment that enables some degree of proliferation, a dynamic battle between emerging tumors and their host commonly referred to as “equilibrium” (Vesely & Schreiber, 2013). In this context, malignant cells can acquire additional genetic and epigenetic alterations that either (i) impair their ability to initiate anticancer immune responses, such as the loss or downregulation of genes coding for endogenous danger signals, (ii) limit their visibility to immune effector cells, such as the loss of MHC class I‐coding genes or beta‐2‐microglobulin (B2M), (iii) increase their resistance to immune effector molecules, such as the loss of caspase 8 (CASP8), or (iv) establish a state of local immunosuppression, such as the upregulation of CD274 (best known as PD‐L1) (Galluzzi et al, 2018). In this context, the equilibrium between cancer cells and the host immune system ceases to exist in favor of an “escape” phase culminating in uncontrolled tumor growth and metastatic dissemination (Dunn et al, 2002; Dersh et al, 2021).

Importantly, the TME of malignancies that escaped immunosurveillance (Rao et al, 2019) undergoes a considerable reconfiguration, generally involving the accumulation of immunosuppressive myeloid and lymphoid cells including M2‐like tumor‐associated macrophages (TAMs) and CD4+CD25+FOXP3+ regulatory T (TREG) cells at the expense of M1‐like TAMs, type I conventional dendritic cells (cDC1s), TH1 CD4+ T cells, CD8+ cytotoxic T lymphocytes (CTLs), and natural killer (NK) cells, all of which promote tumor‐targeting immunity (Talmadge & Gabrilovich, 2013; Mantovani et al, 2017; Lee & Radford, 2019; Sprooten et al, 2019; Togashi et al, 2019). Beyond such a general trend, however, the precise immune contexture of each neoplasm exhibits considerable heterogeneity (De Sousa et al, 2013; Vitale et al, 2021) and has a major impact on disease course and response to therapy (Fridman et al, 2017). Indeed, it has now become clear that the efficacy of most anticancer agents commonly employed in the clinic, encompassing cytotoxic chemotherapeutics, radiation therapy (RT), and targeted anticancer agents, relies at least partially on the (re)activation of immunosurveillance (Galluzzi et al, 2020; Rodriguez‐Ruiz et al, 2020; Petroni et al, 2021). In line with this notion, considerable efforts are being dedicated to the identification of clinically relevant approaches to alter the TME in favor of treatment efficacy, especially for tumors that exhibit rather scarce infiltration by immune effector cells, such as luminal breast cancer and pancreatic carcinoma (Kroemer et al, 2015; Ho et al, 2020).

All cellular components of the TME including malignant and immune cells engage in a dynamic competition for nutrients, oxygen, and trophic signals (all of which are generally scarce as a consequence of relatively poor vascularization) (Martinez‐Outschoorn et al, 2017; O'Sullivan et al, 2019; Garner & de Visser, 2020). Moreover, the availability of nutrients, oxygen, and trophic signals is not equal across all tumor regions and is not constant over time (e.g., before and after therapy), hence constituting a major driver of intratumoral heterogeneity (ITH) (De Sousa et al, 2013; Vitale et al, 2021). Indeed, such restrictions de facto operate as Darwinian pressures, fostering the selection of cells with an accrued capacity to harness alternative carbon sources (e.g., lactate, extracellular amino acids) for catabolic and anabolic reactions in support of proliferation and tolerance to hypoxia (Chang et al, 2015; Xiao et al, 2019).

Adenosine triphosphate (ATP) occupies a key position in the overall configuration of the TME. On one hand, intracellular ATP is crucial for each cellular TME component to survive and mediate its functions (including proliferation, for malignant and non‐terminally differentiated immune cells) (Leone & Powell, 2020; Bergers & Fendt, 2021). On the other hand, the pool of ATP that accesses the TME upon active secretion by living or dying cells into the extracellular space constitutes a major signal transducer (Di Virgilio et al, 2018). The net effect of ATP signaling on the TME, however, depends on multiple factors, including the presence of extracellular ATP‐degrading enzymes as well as the expression pattern of purinergic receptors. Here, we will critically discuss the molecular and cellular mechanisms through which extracellular ATP and its degradation products influence the crosstalk between malignant and immune cells and present recent advances on the purinergic system as a potential target for the development of novel anticancer interventions.

Extracellular ATP homeostasis in the TME

Since ATP cannot be synthesized in the extracellular milieu, the microenvironmental levels of ATP are entirely controlled by the balance between its secretion/release and degradation (Fig 1).

Figure 1. Extracellular ATP homeostasis in the tumor microenvironment.

Figure 1

The concentration of extracellular ATP in the tumor microenvironment is determined by the balance between ATP release and degradation. A variety of cells release ATP either as part of their physiological state or as they respond to stress and potentially die, including cancer cells, dendritic cells (DCs), tumor‐infiltrating neutrophils (TINs), tumor‐associated macrophages (TAMs), and platelets. Extracellular ATP is catabolized by the sequential activity of ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1, best known as CD39), which converts ATP into ADP and AMP, and 5'‐nucleotidase ecto (NT5E, best known as CD73), which converts AMP into adenosine (ADO). CD39 and CD73 are expressed by multiple cell type that populate the tumor microenvironment, including some malignant cells, cancer‐associated fibroblasts (CAFs), exhausted cytotoxic T lymphocytes (CTLs), regulatory T (TREG) cells, an immunosuppressive subset of natural killer (NK) cells, M2‐like TAMs, and myeloid‐derived suppressor cells (MDSCs). Hypoxia is a major driver of ATP degradation in the tumor microenvironment.

Adenosine triphosphate secretion is an active, regulated process that can occur via multiple mechanisms and involve different cellular sources. Molecular mechanisms for ATP secretion encompass: (i) exocytosis of ATP‐containing vesicles, a process that may (but does not necessarily) involve cell death (Imura et al, 2013; Martins et al, 2014) and mechanistically relies on vesicular loading by solute carrier family 17 member 9 (SLC17A9) (Imura et al, 2013; Cao et al, 2014) and the SNAP receptor (SNARE)‐ and Rho‐associated, coiled‐coil containing protein kinase 1 (ROCK1)‐dependent fusion of exocytosis‐competent ATP‐rich vesicles with the plasma membrane (Imura et al, 2013; Martins et al, 2014); (ii) liberation of cytosolic ATP molecules via gap junction protein alpha 1 (GJA1, best known as CX43) hemichannels at gap junctions (Stout et al, 2002; Eltzschig et al, 2006; Kang et al, 2008); and (iii) gradient‐driven efflux via oligomeric pannexin 1 (PANX1) channels (also known as “pannexons”) (Dahl, 2015), which are sensitive to activation by mechanical forces (Bao et al, 2004), by the pro‐inflammatory CASP1 (Narahari et al, 2021), and by apoptotic caspases such as CASP3 and CASP7 (Chekeni et al, 2010; Medina et al, 2020). That said, while both vesicular ATP secretion and PANX1‐dependent release have been documented in living and dying malignant cells (Martins et al, 2014; Martin et al, 2017), CX43 hemichannels appear to be mostly operational in non‐malignant cells of the TME, including astrocytes (Stout et al, 2002), as well as (at least potentially) neutrophils (Dosch et al, 2018) and macrophages (Dosch et al, 2019). Indeed, while both neutrophils and macrophages have been shown to release ATP via CX43 hemichannels in non‐oncological settings, whether such function is preserved in TAMs and tumor‐infiltrating neutrophils (TINs) remains to be elucidated. Along similar lines, platelets are known as major sources of extracellular ATP upon degranulation (Yeaman, 2014), but their contribution to extracellular ATP availability in the TME has just begun to emerge (Schumacher et al, 2013; Gaertner & Massberg, 2019). Additional cellular compartments that may secrete ATP in the TME encompass (at least in some settings) endothelial cells (Sáez et al, 2018; Yang et al, 2020d), fibroblasts (Pinheiro et al, 2013; Murata et al, 2014), dendritic cells (DCs) (Tappe et al, 2018; Martinek et al, 2019), and activated CTLs (Tokunaga et al, 2010). Importantly, while some cells spontaneously secrete at least some ATP in their physiological status, for the most part, ATP is actively released in the context of adaptive responses to microenvironmental perturbations, including mechanical cues (Bao et al, 2004), inflammatory signals (Beckel et al, 2018), hypoxia (Lim To et al, 2015), and exposure to a variety of cancer therapeutics (Michaud et al, 2011; Tatsuno et al, 2019; Rodriguez‐Ruiz et al, 2020). In most such instances, abundant ATP secretion by stressed cells (which is key for extracellular ATP to mediate immunostimulatory effects, see below) involves functional autophagic responses (Michaud et al, 2011), potentially linked to the ability of autophagy to preserve intracellular ATP pools during stress (Rybstein et al, 2018; Anderson & Macleod, 2019). Consistent with this notion, genetic and pharmacological interventions aimed at blocking or boosting autophagic responses in cancer cells have been consistently associated with reduced and increased ATP secretion, respectively, in response to immunogenic chemotherapy (Michaud et al, 2011; Pietrocola et al, 2016; Chen et al, 2019; Kepp & Kroemer, 2020; Wang et al, 2020). Obviously, all dying cells abruptly release their cytosolic ATP pool when they undergo plasma membrane permeabilization (PMP) as the final step of cellular demise. However, while PMP itself has now been shown to be an active (rather than an osmosis‐driven) process even in the context of post‐apoptotic, secondary necrosis (Kayagaki et al, 2021), the consequent spillage of cytosolic content into the extracellular milieu remains a largely unregulated phenomenon.

Extracellular ATP is rapidly catalyzed by the sequential activity of two ectonucleotidases, that is, ectonucleoside triphosphate diphosphohydrolase 1 (ENTPD1, best known as CD39), which converts ATP into ADP and AMP, and 5'‐nucleotidase ecto (NT5E, best known as CD73), which converts AMP into adenosine as the rate‐limiting step of this enzymatic cascade (Allard et al, 2020; Moesta et al, 2020). CD39 is mostly expressed by TREG cells (Borsellino et al, 2007), M2‐like TAMs (d'Almeida et al, 2016), and myeloid‐derived suppressor cells (MDSCs, an immature population of myeloid cells with potent immunosuppressive activity) (Li et al, 2017), as well as by specific cancer cell types, such as adult T‐cell leukemia/lymphoma cells (Nagate et al, 2021), type 16 human papillomavirus (HPV‐16)‐associated cervical carcinoma cells (de Lourdes Mora‐García et al, 2019), and ovarian carcinoma cells (Häusler et al, 2011). Moreover, CD8+ CTLs undergoing terminal exhaustion as a consequence of chronic antigen stimulation generally exhibit a CD39+ phenotype (Canale et al, 2018). Conversely, CD73 is expressed by a wide variety of malignant cells as well as by cancer‐associated fibroblasts (CAFs) (Yu et al, 2020), TREG cells (Stagg et al, 2011), and a regulatory subset of NK cells (Neo et al, 2020). Interestingly, the whole‐body deletion of purinergic receptor P2X 7 (P2RX7), which codes for one of the main receptors of extracellular ATP, has a major impact on extracellular ATP levels in the TME of experimental P2RX7‐competent melanomas (De Marchi et al, 2019), at least in part as a consequence of altered tumor infiltration by CD39+ and CD73+ TREG cells and decreased ATP release by TAMs (De Marchi et al, 2019). Such an effect, however, cannot be recapitulated by the pharmacological P2RX7 antagonist A740003 as a result of its mixed activity on immune cells (it fails to alter tumor infiltration by TREG cells, decreases the abundance of intratumoral CD39+ and CD73+ effector T (TEFF) cells, and inhibits ATP secretion by TAMs) and malignant cells (it favors ATP secretion by malignant cells) (De Marchi et al, 2019).

Of note, extracellular ATP degradation does not necessarily require the expression of CD39 and CD73 on the same cell (in cis), but can also occur efficiently when these ectonucleotidases are expressed by different cellular compartments that are in proximity to each other within the TME (Schuler et al, 2014). CD73 is abundant in TREG cell‐derived exosomes (Smyth et al, 2013), which are highly mobile and hence further promote the overall catalytic efficiency of ATP degradation within the TME. Moreover, CD38 (also known as cyclic ADP‐ribose hydrolase) and ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1), which are expressed by some cancer cells and exhausted T cells, can compensate for limited CD39 activity as they catalyze the conversion of extracellular NAD+ into ADP ribose and AMP (Morandi et al, 2015). Finally, the expression of both CD39 and CD73 can be upregulated by hypoxia, which is relatively common in the TME of solid neoplasms, via a transcriptional mechanism that involves hypoxia‐inducible factor 1 subunit alpha (HIF1A, best known as HIF‐1α) (Giatromanolaki et al, 2020; Synnestvedt et al, 2002).

In summary, the levels of extracellular ATP in the TME are dynamically determined by the mutually opposed inputs of secretion/release vs. degradation. As the factors governing these aspects of the ATP biology exhibit a considerable degree of ITH, regional and temporal fluctuations in extracellular ATP levels are likely to play a major role in the outcome of purinergic signaling in the TME, as discussed further below.

Immunostimulation by extracellular ATP

Extracellular ATP mediates two main functions: (i) It operates as a chemotactic cue for myeloid cells, upon binding to the purinergic receptor P2Y2 (P2RY2), a metabotropic receptor (Elliott et al, 2009; Chekeni et al, 2010), and (ii) it promotes activation of the inflammasome and hence CASP1‐dependent secretion of interleukin 1 beta (IL1B) and IL18 upon binding to P2RX7, an ionotropic receptor (Perregaux et al, 2000). Importantly, both these effects are required for the optimal activation of tumor‐specific immune responses by (and hence the complete efficacy of) immunogenic chemotherapeutics such as anthracyclines and oxaliplatin, as demonstrated in P2ry2 −/−, P2rx7 −/−, Casp1 −/− and Il18 −/− mice, as well as mice lacking a core component of the inflammasome (Nlrp3 −/− mice), the main IL1B receptor (Il1r1 −/− mice) or treated with a purinergic receptor antagonist (suramin) or an IL1B‐blocking antibody (Ghiringhelli et al, 2009; Aymeric et al, 2010; Ma et al, 2013).

In this setting, DC precursors newly recruited to the TME via ATP released by cancer cells succumbing to immunogenic cell death (ICD) not only mature upon ATP‐driven inflammasome activation and migrate to tumor‐draining lymph nodes or tertiary lymphoid structures to prime adaptive anticancer immunity, but also recruit a population of IL17‐producing γδ T cells that is critical for tumor infiltration by primed CTLs (Ma et al, 2011). In accordance with this notion, optimal anticancer immune responses (and consequent superior therapeutic efficacy) driven by immunogenic chemotherapeutics are compromised in Il17a −/− and Il17ra −/− mice (Ma et al, 2011). Intriguingly, it has recently shown that the chemotactic activity of ATP on DCs also involves P2RX7 and PANX1 (Saez et al, 2017), suggesting the existence of a feed‐forward loop whereby intracellular ATP stores may contribute to DC migratory capacity (Saez et al, 2017). Moreover, elevated levels of extracellular ATP appear to induce pyroptosis in P2RX7+ M2‐like TAMs, hence supporting T cell‐mediated antitumor immunity upon the depletion of immunosuppressive cells from the TME (Bidula et al, 2019).

Importantly, the net immunomodulatory effect of extracellular ATP depends on the activation of additional signaling pathways. Indeed, the PANX1‐dependent co‐release of ATP and a wide panel of metabolites including ADP, AMP, GMP, creatine, spermidine, and glycerol‐3‐phosphate (G3P) by dying cells reportedly promote the removal of cell corpses while preventing the initiation of inflammatory reactions (Medina et al, 2020; Narahari et al, 2021). Moreover, extracellular ATP can have direct tumorigenic functions. Specifically, the cancer cell‐driven release of ATP from platelets initiates a P2RY2‐dependent signaling cascade that promotes tumor extravasation and metastatic dissemination upon the opening of endothelial barriers (Schumacher et al, 2013; Chen et al, 2019; Wang et al, 2020). The autophagy‐dependent secretion of ATP by melanoma cells has been shown to promote invasiveness and resistance to the BRAF inhibitor vemurafenib, a process that requires P2RX7 expression in the malignant cell compartment (Martin et al, 2017). Similar findings have been obtained with human triple‐negative breast cancer (TNBC) MDA‐MB‐231 cells upon the ATP‐dependent activation of the transcription factor SRY‐box transcription factor 9 (SOX9) (Yang et al, 2020a). Finally, NME/NM23 nucleoside diphosphate kinase 1 (NME1, best known as NDPK‐A) and NME2 (best known as NDPK‐B) expression on extracellular vesicles from MDA‐MB‐231 cells reportedly support the formation of pulmonary metastatic niches as a consequence of extracellular ATP generation in situ and consequent activation of purinergic receptor P2Y1 (P2RY1) (Duan et al, 2021).

Consistent with the multipronged effects of extracellular ATP on the TME, a large body of clinical literature suggests that genetic or epigenetic defects affecting ATP signaling influence disease outcome in cancer patients in a context‐dependent manner (Table 1). For instance, while loss‐of‐function polymorphisms in P2RX7 (rs3751143; rs208294) have been associated with advanced stage or poor disease outcome in cohorts of patients with breast carcinoma (Ghiringhelli et al, 2009), chronic lymphocytic leukemia (CLL) (Thunberg et al, 2002; Wiley et al, 2002; Zhang et al, 2003), and papillary thyroid carcinoma (PTC) (Dardano et al, 2009), no impact on clinicopathological variables could be attributed to rs3751143 in other cohorts of subjects with CLL (Starczynski et al, 2003; Nückel et al, 2004), multiple myeloma (Paneesha et al, 2006), and PTC (Dardano et al, 2009), while increased expression levels of P2RX7 have been linked to disease progression in an independent cohort of CLL patients (Adinolfi et al, 2002). Likewise, elevated P2RY2 levels have been associated with gastric malignant transformation (Aquea et al, 2014). A variety of immunohistochemical and transcriptional signatures of proficient autophagic responses have been linked to worsened disease outcome in cohorts of breast (Yamazaki et al, 2020), gastric (Kim et al, 2019; Wang et al, 2021), pancreatic (Ko et al, 2013; Cui et al, 2019), and head and neck (Jiang et al, 2012) cancer patients, while the contrary held true (or there was no impact on clinicopathological variables) in independent series of patients with breast (Ladoire et al, 2015; Tang et al, 2015; Ladoire et al, 2016), ovarian (Chen et al, 2020c), hepatocellular (Lee et al, 2013; Qin et al, 2018), gastric (Wang et al, 2021), colorectal (Li et al, 2020), and salivary gland (Li et al, 2019a) carcinoma.

Table 1.

Pathophysiological relevance of extracellular ATP signaling in human cancer.

Cancer No. patients Variable Technology Impact References
Breast cancer 225 P2RX7 rs3751143 SNP analysis

Metastatic dissemination

Decreased OS

Ghiringhelli et al (2009)

1,067

1,992

BECN1 Gene expression profiling Improved disease outcome Tang et al (2015)
152 MAP1LC3B IHC

Improved MFS

Improved OS

Ladoire et al (2016)

152

1,646

MAP1LC3B IHC Improved PFS Ladoire et al (2015)
CLL 36 P2RX7 rs3751143 SNP analysis Disease stage Wiley et al (2002)
144 P2RX7 rs3751143 SNP analysis Marginally decreased OS Zhang et al (2003)
170 P2RX7 rs3751143 SNP analysis Decreased OS Thunberg et al (2002)
111 P2RX7 rs3751143 SNP analysis No correlation with clinical outcome Nückel et al (2004)
121 P2RX7 rs3751143 SNP analysis No correlation with clinical outcome Starczynski et al (2003)
21 P2RX7 Immunoblotting Disease progression Adinolfi et al (2002)
Colorectal cancer 2,297 MAP1LC3B Gene expression profiling Increased OS Li et al (2020)
Gastric cancer 14 P2RY2 Gene expression profiling Disease Aquea et al (2014)
354 MAP1LC3C Gene expression profiling Improved OS Wang et al (2021)
354 ATG4D Gene expression profiling Decreased OS Wang et al (2021)
402 MAP1LC3B and SQSTM1 IHC, immunoblotting a,nd RT–PCR Decreased OS Kim et al (2019)
Head and neck cancer 79 MAP1LC3B IHC Disease stage Jiang et al (2012)
Hepatocellular carcinoma 190 MAP1LC3B IHC Improved OS Lee et al (2013)
1,086 BECN1 Gene expression profiling Improved OS Qin et al (2018)
Multiple myeloma 136 P2RX7 rs3751143 SNP analysis No correlation with clinical outcome Paneesha et al (2006)
Ovarian cancer 1,497 BECN1 Gene expression profiling

Improved OS and PFS

Chen et al (2020c)
1,497 MAP1LC3B Gene expression profiling No correlation with clinical outcome Chen et al (2020c)
Pancreatic cancer 73 BECN1 IHC Disease progression Ko et al (2013)
86 BECN1 and MAP1LC3B IHC and RT–PCR

Metastatic dissemination

Disease stage

Decreased OS

Cui et al (2019)
Papillary thyroid cancer 121 P2RX7 rs3751143 SNP analysis Disease type Dardano et al (2009)
121 P2RX7 rs208294 SNP analysis No effect on disease stage Dardano et al (2009)
Salivary gland carcinoma 48 BECN1 and MAP1LC3B IHC Low disease stage Li et al (2019a)

CLL, chronic lymphocytic leukemia; IHC, immunohistochemistry; MFS, metastasis‐free survival; N/A, not available; OS, overall survival; PFS, progression‐free survival; SNP, single nucleotide polymorphism.

In summary, while an abundant preclinical literature mechanistically implicates ATP secretion by stressed and dying cells in the initiation of anticancer immune responses (Fig 2), additional, hitherto poorly characterized factors and mechanisms appear to influence the net effect of ATP signaling in the TME.

Figure 2. Immunostimulation by extracellular ATP.

Figure 2

Extracellular ATP mediates chemotactic effects on myeloid cells, upon binding to purinergic receptor P2Y2 (P2RY2), and promotes the inflammasome‐dependent secretion of interleukin 1 beta (IL1B) and IL18 upon binding to purinergic receptor P2X 7 (P2RX7). These effects are critical for mature dendritic cells (DCs) to prime cytotoxic T lymphocytes (CTLs) against tumor‐derived antigens and hence initiate adaptive anticancer immunity. Alongside, extracellular ATP triggers pyroptosis in tumor‐associated macrophages (TAMs), hence depleting the tumor microenvironment of generally immunosuppressive cells along with the emission of pyroptosis‐dependent immunostimulatory signals.

Immunosuppression by extracellular ATP metabolites

Extracellular ATP degradation by the sequential activity of CD39 and CD73 mediates immunosuppressive effects not only as a consequence of limited ATP‐dependent immunostimulation (see above), but also due to the generation of adenosine, which per se promotes tumor progression via immunological and non‐immunological mechanisms (Allard et al, 2020). Consistent with this notion, transgene‐driven overexpression of CD39 by malignant cells has been associated with impaired anticancer immunity driven by immunogenic chemotherapy (and hence poor disease outcome) in syngeneic immunocompetent mouse models of fibrosarcoma (Michaud et al, 2011; Pietrocola et al, 2016). Similarly, experimental CD73 overexpression in human cervical carcinoma as well as human and mouse breast carcinoma cells has been shown to promote invasiveness and metastatic potential (Zhou et al, 2007; Gao et al, 2017), at least in part as a consequence of autocrine–paracrine adenosine signaling via adenosine A2b receptor (ADORA2B) and ADORA2B‐driven neovascularization (Stagg et al, 2010; Mittal et al, 2016; Ludwig et al, 2020). Together with ADORA2A, ADORA2B is indeed the main receptor for extracellular adenosine signaling in the TME (Allard et al, 2020). However, while ADORA2B is expressed by various malignant cell types including breast cancer (Stagg et al, 2010; Lan et al, 2018), cervical cancer (Torres‐Pineda et al, 2020), and melanoma (Mittal et al, 2016) cells, as well as by endothelial cells (Ludwig et al, 2020), DCs (Chen et al, 2020b), and M2‐like TAMs (Cohen et al, 2015), ADORA2A expression is mostly restricted to myeloid cells (Nakamura et al, 2020), NK cells (Young et al, 2018), CTLs (Kjaergaard et al, 2018; Shi et al, 2019), and lymphatic endothelial cell precursors (Allard et al, 2019).

From a mechanistic standpoint, ADORA2A‐ and ADORAB‐driven immunosuppression results from the activation of intracellular cyclic AMP (cAMP) signaling, the ultimate functional outcome of which depends on the specific immune cell type expressing these receptors. Thus, cAMP signaling directly inhibits TCR activation and cytokine production in CTLs (Lappas et al, 2005), promotes forkhead box P3 (FOXP3) synthesis and the upregulation of the co‐inhibitory receptor cytotoxic T lymphocyte‐associated protein 4 (CTLA4) in TREG cells (Klein & Bopp, 2016), impairs NK cell effector and secretory functions (Raskovalova et al, 2005), inhibits NF‐κB‐dependent inflammatory responses (Minguet et al, 2005) and drives the secretion of immunosuppressive cytokines such as IL10 from TAMs and MDSCs (Németh et al, 2005; Cekic et al, 2014), and perturbs the maturation of DCs (Kayhan et al, 2019). Of note, both ADORA2A and ADORA2B can be upregulated by hypoxia (Sitkovsky et al, 2014; Lan et al, 2018), in thus far resembling CD39 and CD73. Thus, particularly hypoxic regions of the TME are expected to exhibit robust adenosinergic signaling and hence (i) potent immunosuppression, (ii) accrued neovascularization, (iii) increased vascular permeability, and (iv) enhanced cancer cell motility, de facto representing ideal niches for metastatic dissemination. This is further aggravated by immunometabolic circuitries driven by hypoxia. Specifically, hypoxic tumor regions are rich in glycolytic lactate, which mediates multipronged immunosuppressive effects involving CTLs (Brand et al, 2016), NK cells (Husain et al, 2013), TREG cells (Watson et al, 2021), and MDSCs (Yang et al, 2020c).

Consistent with the potent immunosuppressive effects of adenosine signaling in the TME, an abundant clinical literature links elevated expression levels of adenosine‐generating enzymes or adenosine receptors to poor disease progression in cohorts of patients with various tumors (Table 2). Thus, high levels of CD39 have been associated with advanced grade or poor disease outcome in multiple cohorts of patients with CLL (Pulte et al, 2011), renal cell carcinoma (Wu et al, 2020a), endometrial tumors (Aliagas et al, 2014), pancreatic carcinoma (Künzli et al, 2007), and non‐small cell lung cancer (NSCLC; Li et al, 2017). Similar clinical findings have been correlated with various single nucleotide polymorphisms affecting Entpd1 (i.e., rs10748643, rs11188513, rs2226163) in patients with colorectal carcinoma (Tokunaga et al, 2019; Gallerano et al, 2020), but the functional impact of these variants on CD39 functions remains unclear. Moreover, abundant tumor‐infiltrating or circulating levels CD4+ or CD8+ cells expressing CD39 have been linked to disease progression or resistance to therapy in various cohorts of individuals with CLL (Perry et al, 2012), colorectal carcinoma (Gallerano et al, 2020), head and neck squamous cell carcinoma (Gallerano et al, 2020), pancreatic cancer (Gallerano et al, 2020), NSCLC (Koh et al, 2020), and renal cell carcinoma (Qi et al, 2020). Along these lines, intratumoral or circulating biomarkers of CD73 proficiency (including expression levels and enzymatic activity) have been correlated with poor disease outcome in patients with diffuse large B‐cell lymphoma (Wang et al, 2019), ovarian cancer (Turcotte et al, 2015), pancreatic carcinoma (Chen et al, 2020a; Tahkola et al, 2021), NSCLC (Li et al, 2017), renal cell carcinoma (Tripathi et al, 2020), breast cancer (Loi et al, 2013; Buisseret et al, 2018), glioma (Xu et al, 2013), colorectal carcinoma (Messaoudi et al, 2020), and metastatic melanoma (Turiello et al, 2020). The Nt5e polymorphism rs2229523 (of hitherto unclear functional significance) correlated with limited overall survival in patients with colorectal carcinoma (Tokunaga et al, 2019), as did tumor infiltration by CD8+CD73+ cells in prostate cancer patients (Leclerc et al, 2016). Finally, ADORA2A expression or signaling was linked to poor disease outcome in individuals with diffuse large B‐cell lymphoma (Wang et al, 2019), renal carcinoma (Kamai et al, 2021), and a variety of tumors from The Cancer Genome Atlas (Sidders et al, 2020), as was the ADORA2B polymorphism rs2015353 in patients with colorectal cancer (Tokunaga et al, 2019), although the functional implications of rs2015353 on ADORA2B activity remain to be elucidated.

Table 2.

Pathophysiological relevance of adenosinergic signaling in human cancer.

Cancer No. patients Variable Technology Impact References
Breast cancer 122 CD73 IHC Decreased OS Buisseret et al (2018)
>6,000 NT5E Gene expression profiling Reduced pCR Loi et al (2013)
CLL 34 Circulating CD4+CD39+ and CD8+CD39+ cells Flow cytometry Disease stage Pulte et al (2011)
62 Circulating CD4+CD39+ T cells Flow cytometry Disease stage Perry et al (2012)
Colorectal cancer 107 ADORA2B rs2015353 SNP analysis Decreased OS Tokunaga et al (2019)

107

215

ENTPD1 rs11188513 SNP analysis Decreased OS Tokunaga et al (2019)
215 ENTPD1 rs2226163 SNP analysis Decreased OS Tokunaga et al (2019)
107 NT5E rs2229523 SNP analysis Decreased OS Tokunaga et al (2019)
129 ENTPD1, NT5E, and ADORA2B SNP analysis No correlation Tokunaga et al (2019)
60 Circulating CD8+CD39+ T cells Flow cytometry Disease progression Gallerano et al (2020)
60 ENTPD1 rs10748643 SNP analysis Disease progression Gallerano et al (2020)
215 CD73 IHC Disease progression Decreased OS Messaoudi et al (2020)
193 Circulating CD73 ELISA Decreased OS Messaoudi et al (2020)
DLBCL 91 ADORA2A on TILs IHC Decreased OS Wang et al (2019)
91 CD73 IHC Decreased OS Wang et al (2019)
Endometrial tumors 29 CD39 IHC Tumor grade Aliagas et al (2014)
Glioma 500 NT5E Gene expression profiling Limited DFS Xu et al (2013)
HNSCC 19 Circulating CD8+CD39+ T cells Flow cytometry Disease progression Gallerano et al (2020)
Melanoma 546 Circulating CD73 AMPase activity

Disease progression

Decreased OS

Turiello et al (2020)
NSCLC 132 Circulating CD8+CD39+ T cells Flow cytometry Decreased OS Koh et al (2020)
24 Circulating CD8+CD39+ MDSCs Flow cytometry Tumor infiltration by MDSCs Li et al (2017)
Ovarian cancer 208 CD73 IHC Decreased OS Turcotte et al (2015)
Pancreatic cancer 28 ENTPD1 RT–PCR Decreased OS Künzli et al (2007)
3 Circulating CD8+CD39+ T cells Flow cytometry Disease progression Gallerano et al (2020)
110 CD73 IHC Decreased OS Tahkola et al (2021)
168 NT5E Gene expression profiling

Disease progression

Decreased OS

Chen et al (2020a)
Prostate cancer 285 Circulating CD8+CD73+ T cells IHC Disease progression Leclerc et al (2016)
Renal cell carcinoma 60 ADORA2A expression IHC

Metastatic dissemination

Decreased OS

Kamai et al (2021)
138 CD73 IHC Decreased OS Tripathi et al (2020)
243 CD8+CD39+ T cells IHC Disease progression Qi et al (2020)
367 CD39 IHC and RT–PCR

Disease stage

Disease progression

Wu et al (2020a)
Various tumor types N/A ADORA2A‐regulated gene expression Gene expression profiling Decreased OS Sidders et al (2020)

CLL, chronic lymphocytic leukemia; DBLCL, diffuse large B‐cell lymphoma; DFS, disease‐free survival; ELISA, enzyme‐linked immunosorbent assay; IHC, immunohistochemistry; MDSC, myeloid‐derived suppressor cell; N/A, not available; OS, overall survival; pCR, pathological complete response; SNP, single nucleotide polymorphism; TIL, tumor‐infiltrating lymphocyte.

Of note, the discovery that the TME contains unusually high levels of extracellular ATP has prompted innovative therapeutic approaches that specifically harness this biochemical feature. For instance, a monoclonal antibody specific for the immunostimulatory receptor TNF receptor superfamily member 9 (TNFRSF9, best known as CD137) has been engineered to drive CD137 signaling only in the presence of nearly millimolar ATP levels, thus inducing a potent anticancer immune response in the absence of adverse effects due to extratumoral activity (Kamata‐Sakurai et al, 2020). This agent is currently being tested in combination with the immune checkpoint inhibitor (ICI) atezolizumab (a PD‐L1‐blocking antibody) for safety and preliminary efficacy in a phase I clinical trial enrolling patients with solid tumors (JapicCTI‐205153). Along similar lines, it has recently been shown that a novel positive allosteric modulator of P2RX7 (which only acts in the presence of high extracellular ATP levels) potentiates the therapeutic effects of an ICI specific for programmed cell death 1 (PDCD1, best known as PD‐1) by stimulating DCs to release IL18 in support of the effector functions of tumor‐infiltrating NK cells and CTLs (Douguet et al, 2021). Incidentally, these therapeutic applications provide an independent and convincing demonstration (mutatis mutandis, almost an “ex adiuvantibus” proof) of the accuracy of early measurements of extracellular ATP concentration in the TME (Pellegatti et al, 2008).

Taken together, these observations point to CD39/CD73‐dependent adenosine generation and consequent adenosinergic signaling via ADORA2A and ADORA2B as a key immunosuppressive mechanism supporting the progression and metastatic dissemination multiple tumors (Fig 3). Of note, while additional adenosine receptors including ADORA1 and ADORA3 (which inhibit cAMP signaling) are expressed by malignant cells and some tumor‐infiltrating immune cells (Stagg & Smyth, 2010), their role in immunosurveillance remains poorly investigated.

Figure 3. Immunosuppression by extracellular ATP metabolites.

Figure 3

Extracellular adenosine (ADO) not only favors metastatic dissemination by binding adenosine A2b receptor (ADORA2B) on malignant cells, hence promoting invasiveness, and endothelial cells (ECs), thus promoting neovascularization, but also modulates the functions of multiple immune cells upon interaction with ADORA2B or adenosine A2a receptor (ADORA2A). Specifically, adenosine signaling inhibits dendritic cell (DC) maturation and interferes with the effector functions of cytotoxic T lymphocytes (CTLs), B cells, and natural killer (NK) cells, while favoring immunosuppression by M2‐like tumor‐associated macrophages (TAMs), myeloid‐derived suppressor cells (MDSCs), and regulatory T (TREG) cells.

Targeting purinergic signaling for cancer therapy

Consistent with the key role of extracellular ATP and its degradation products in the control of immunosurveillance, a variety of pharmacological and genetic approaches designed to boost ATP‐driven immunostimulation or inhibit adenosine‐dependent immunosuppression (alone or combined with other treatments) have been shown to mediate prominent antineoplastic effects in immunocompetent mouse models of cancer.

Boosting ATP secretion with autophagy‐activating maneuvers, including weekly cycles of nutrient deprivation and administration of so‐called caloric restriction mimetics (CRMs, i.e., molecules that induce autophagy and cause other biochemical correlates of nutrient deprivation in the absence of sizeable weight loss), has been linked to improved therapeutic responses to immunogenic chemotherapies in immunocompetent mouse models of fibrosarcoma, correlating with decreased infiltration by TREG cells, via a mechanism that depends on expression of the essential autophagy gene autophagy‐related 5 (ATG5) in cancer cells and intact immune responses (Pietrocola et al, 2016; Castoldi et al, 2020; Wu et al, 2020b). Along similar lines, short‐term starvation reportedly boosts the responsiveness of mouse breast cancer cells growing in immunocompetent syngeneic hosts to radiation therapy (RT) (Saleh et al, 2013; Simone et al, 2016). However, proficient autophagic responses in mouse breast cancer cells limit the efficacy of RT in vivo as a consequence of an improved disposal of permeabilized mitochondria that would otherwise release mitochondrial DNA in the cytosol and trigger cyclic GMP‐AMP synthase (CGAS) signaling coupled to type I interferon (IFN) secretion (Medler et al, 2019; Yamazaki et al, 2020). This suggests that whole‐body autophagy activation by short‐term fasting may support the efficacy of RT by mechanisms unrelated to ATP secretion in the TME, potentially linked to improved autophagic responses in immune cells, most of which rely on autophagy for optimal functions (Clarke & Simon, 2019). That said, even though cyclic/short‐term nutrient deprivation could be safely implemented in at least some cancer patients (Krstic et al, 2020), clinicians remain cautious on implementing clinical trials involving such a nutritional measure. Similarly, while many CRMs with robust anticancer activity in preclinical models are currently available as over‐the‐counter medications (e.g., aspirin) (Castoldi et al, 2020) or vitamin supplementations (e.g., nicotinamide) (Buqué et al, 2020), clinical development remains at bay, at least in some cases reflecting potential dosing issues.

Blocking CD39 or CD73 has also been associated with robust antineoplastic effects in immunocompetent mouse models of cancer. For instance, whole‐body Entpd1 deletion as well as reconstitution of radiosensitive hematopoietic cells with Entpd1 −/− precursors have been shown to inhibit in vivo growth and metastatic dissemination of mouse melanoma and colorectal carcinoma cells (Sun et al, 2010; Perrot et al, 2019). Similar findings have been obtained in Entpd1 −/− mice xenografted with mouse melanoma or fibrosarcoma cells and treated with immunogenic chemotherapy or ICIs (Perrot et al, 2019), as well as with (i) monoclonal antibodies targeting human CD39 in human ENTPD1 knock‐in mice bearing mouse fibrosarcoma cells (Perrot et al, 2019), (ii) monoclonal antibodies targeting mouse CD39 in wild‐type mice used as hosts for mouse melanoma, fibrosarcoma, or colorectal carcinoma cells (Li et al, 2019c; Yan et al, 2020), (iii) antisense oligonucleotides targeting Entpd1 in wild‐type mice bearing syngeneic breast cancer cells (Kashyap et al, 2019), and (iv) pharmacological CD39 inhibitors in wild‐type mice xenografted with mouse colorectal carcinoma cells (Michaud et al, 2011). Of note, in the latter models, the anticancer effects of CD39 blockage could be abrogated by P2rx7 or Nlrp3 whole‐body deletion, as well as by co‐administration of monoclonal antibodies targeting IL18 (Li et al, 2019c; Yan et al, 2020), formally linking therapeutic activity to accrued extracellular ATP signaling (rather than to mere adenosine depletion). Mice lacking Nt5e have been shown to be poorly permissive to the growth of syngeneic glioblastoma, lymphoma, melanoma, ovarian cancer, colorectal carcinoma, and breast cancer cells (Stagg et al, 2011; Wang et al, 2011; Yegutkin et al, 2011; Young et al, 2016; Yan et al, 2019). Moreover, blockage of CD73 with monoclonal antibodies or pharmacological agents mediates standalone therapeutic effects or enhances the efficacy of other treatments (encompassing chemotherapeutics, RT, and ICIs) in immunocompetent mouse models of breast cancer (Loi et al, 2013; Allard et al, 2014; Young et al, 2016; Wennerberg et al, 2020), colorectal carcinoma (Allard et al, 2013; Hay et al, 2016; Tsukui et al, 2020), melanoma (Iannone et al, 2014; Young et al, 2016), ovarian cancer (Häusler et al, 2014; Li et al, 2019b), head and neck squamous cell carcinoma (Deng et al, 2018), and prostate cancer (Allard et al, 2013). In line with such an abundant preclinical literature, several monoclonal antibodies targeting CD39 or CD73 are currently being tested for their safety and anticancer efficacy, either as standalone therapeutics or combined with ICIs, in clinical trials (Table 3). Preliminary findings from such studies point to an acceptable safety profile and promising clinical activity encompassing disease stabilization and (at least in some patients) partial or complete responses (Mobasher et al, 2019; Bendell et al, 2020).

Table 3.

Ongoing clinical trials targeting purinergic signaling for cancer therapy. a

Agent Target Indications Phase Status Notes Ref.

AZD4635

(AstraZeneca)

ADORA2A CRPC II Recruiting In combination with oleclumab (anti‐CD73), durvalumab (anti‐PD‐L1) NCT04089553
NSCLC I/II Active In combination with oleclumab (anti‐CD73) NCT03381274
Solid tumors I Active Single agent and in combination with durvalumab (anti‐PD‐L1), oleclumab (anti‐CD73), docetaxel, abiraterone acetate, enzalutamide NCT02740985
Solid tumors I Recruiting Single agent NCT03980821

Ciforadenant

CPI‐444/V81444 (Corvus Pharmaceuticals)

ADORA2A NSCLC I/II Recruiting Single agent and in combination with atezolizumab (anti‐PD‐L1) NCT03337698

CPRC

Renal cell carcinoma

I Recruiting Single agent and in combination with atezolizumab (anti‐PD‐L1) NCT02655822
EOS100850 (iTeos Therapeutics) ADORA2A Solid tumors I Recruiting Single agent NCT03873883

Etrumadenant

AB928 (Arcus Biosciences)

ADORA2A

ADORA2B

CRC

GEC

I Active In combination with mFOLFOX NCT03720678
NSCLC I/I Recruiting In combination with carboplatin, pemetrexed and pembrolizumab NCT03846310

Ovarian cancer

TNBC

I Recruiting In combination with IPI‐549 (PI3γ inhibitor), doxorubicin, paclitaxel NCT03719326
Solid tumors I Active In combination with zimberelimab (AB122, anti‐PD1) NCT03629756
Taminadenant NIR178/PBF‐509 (Pablobio/Novartis) ADORA2A NSCLC I Active Single agent and in combination with spartalizumab (anti‐PD‐1) NCT02403193
TNBC I Recruiting In combination with spartalizumab (anti‐PD‐1) and LAG525 (anti‐LAG‐3) NCT03742349
Solid tumors I Recruiting Single agent and in combination with NZV930 (anti‐CD73), spartalizumab (anti‐PD1) NCT03549000
Solid tumors I Recruiting Single agent and in combination with spartalizumab (anti‐PD‐1); NZV930 (anti‐CD73), KAZ954 NCT04237649
Solid tumors II Recruiting Single agent and in combination with spartalizumab (anti‐PD‐1) NCT03207867
SRF617 (Surface Oncology) CD39 Solid tumors I Recruiting Single agent and in combination with paclitaxel, gemcitabine, pembrolizumab (anti‐PD‐L1) NCT04336098
TTX‐030 (AbbVie) CD39

Lymphoma

Solid tumors

I/I Recruiting Pembrolizumab (anti‐PD‐L1), docetaxel, gemcitabine NCT03884556
Solid tumors I Recruiting In combination with budigalimab (anti‐PD‐1), mFOLFOX NCT04306900
BMS986179 (Bristol Myers Squibb) CD73 Solid tumors I/II Active Single agent and in combination with nivolumab (anti‐PD‐1) NCT02754141
CPI‐006 (Corvus Pharmaceuticals) CD73

NHL

Solid tumors

I/I Recruiting Single agent and in combination with ciforadenant (ADORA2A antagonist), pembrolizumab (anti‐PD‐L1) NCT03454451
LY3475070 (Eli‐Lilly) CD73 Solid tumors I Recruiting Single agent or in combination with pembrolizumab (anti‐PD‐L1) NCT04148937
NZV930 (Surface Oncology) CD73 Solid tumors I Recruiting Single agent or in combination with spartalizumab (anti‐PD‐1), NIR178 (ADORA2A antagonist) NCT03549000

Oleclumab

MEDI9447 (AstraZeneca)

CD73 Bladder cancer I Recruiting In combination with durvalumab (anti‐PD‐L1) NCT03773666
NSCLC II Recruiting In combination with durvalumab (anti‐PD‐L1) NCT03334617
Ovarian cancer II Recruiting Single agent or in combination with durvalumab (anti‐PD‐L1) NCT03267589
Pancreatic cancer I/II Recruiting In combination with gemcitabine, paclitaxel, durvalumab (anti‐PD‐L1), FOLFOX NCT03611556
TNBC I/II Recruiting In combination with durvalumab (anti‐PD‐L1), paclitaxel NCT03742102

TJ004309

TJD5 (Tracon Pharmaceuticals)

CD73 Solid tumors I Recruiting In combination with atezolizumab (anti‐PD‐L1) NCT03835949

CRC, colorectal cancer; CRPC, castration‐resistant prostate cancer; GEC, gastroesophageal cancer; NHL, non‐Hodgkin lymphoma; NSCLC, non‐small cell lung cancer; TNBC, triple‐negative breast cancer.

a

Restricted to active and recruiting studies, as per www.clinicaltrials.gov on February 15, 2021.

Genetic and pharmacological strategies for ADORA2A and ADORA2B inhibition have also been shown to mediate anticancer effects in preclinical cancer models. For instance, (whole‐body or myeloid cell‐specific) Adora2a or Adora2b deletion has been demonstrated to inhibit tumor growth in immunocompetent mice bearing syngeneic melanoma (Ohta et al, 2006; Cekic et al, 2014; Chen et al, 2020b), lymphoma (Waickman et al, 2012; Nakamura et al, 2020), breast carcinoma (Beavis et al, 2013), or lung cancer cells (Chen et al, 2020b). Consistent with this notion, pharmacological ADORA2A (e.g., CPI‐444, SCH58261, AZD4635) or ADORA2B (e.g., PSB‐1115) inhibitors have been attributed robust antineoplastic properties in syngeneic mouse models of melanoma (Willingham et al, 2018), fibrosarcoma (Beavis et al, 2017), breast cancer (Beavis et al, 2013; Mittal et al, 2014; Beavis et al, 2015; Beavis et al, 2017), lymphoma (Nakamura et al, 2020), colorectal carcinoma (Beavis et al, 2015; Willingham et al, 2018), multiple myeloma (Yang et al, 2020b), and renal cell carcinoma (Willingham et al, 2018), especially when combined with ICIs or other immunotherapies. Importantly, at least in some of these models, co‐inhibition of ADORA2A and CD73 mediated superior tumor control as compared to inhibiting ADORA2A or CD73 alone (Young et al, 2016), suggesting a non‐redundant role for these two factors in the establishment of local immunosuppression. Consistent with these preclinical observations, a number of ADORA2A or dual ADORA2A/ADORA2B inhibitors are currently being investigated for safety and activity in clinical trials (Table 3). Preliminary findings from these studies indicate that many ADORA2A/ADORA2B inhibitors exhibit an acceptable safety profile and at least some degree of clinical activity (Chiappori et al, 2018; Fong et al, 2020; Lim et al, 2020).

Additional approaches that may be harnessed to target adenosine signaling in the TME encompass the use of CD38 or ENPP1 blockers and strategies that revert tumor hypoxia (e.g., respiratory hyperoxygenation) (Hatfield et al, 2014; Hatfield & Sitkovsky, 2020). However, CD38‐specific agents (including the FDA‐approved monoclonal antibody daratumumab) are currently used with the aim of eradicating CD38‐expressing myeloma cells (Facon et al, 2019), and ENPP1 blockers are still in preclinical development (Carozza et al, 2020). Similarly, hyperoxygenation has been employed in the past to improve radiosensitivity (because the ability of radiation therapy to cause DNA damage in cancer cell depends on local oxygen tension) but is no longer used for this purpose.

In summary, although a variety of strategies have been successfully used to target purinergic signaling in preclinical tumor models, CD73, ADORA2A, and (less so) CD39 blockers are the only drugs currently in clinical development as anticancer agents, for the most part in combination with standard of care therapeutics or ICIs (Table 3).

Concluding remarks

In summary, extracellular ATP and its degradation products play a key role in the regulation of the tumor immune contexture, hence have a major influence on the propensity of human neoplasms to respond to therapy. While various agents aimed at boosting extracellular ATP concentrations and/or limiting adenosine signaling are already in clinical development, multiple questions to be addressed and avenues to be explored remain. First, it will be important to determine the contribution of P2RX7 and/or P2RY2 signaling in cancer cells to tumor growth and metastatic dissemination in specific settings. Accumulating evidence indicates that some cancer cells can harness extracellular ATP in support of disease progression and resistance to therapy (Martin et al, 2017), suggesting that CD39 and/or CD73 inhibition may not mediate optimal anticancer effects in some settings, and calling for the identification of cancer cell‐targeted P2RX7 or P2RY2 inhibitors. Second, it will be crucial to develop combinatorial approaches based on the dual blockage of ATP degradation and adenosine signaling, potentially in the context of ICI‐based immunotherapy. Indeed, it appears that the efficacy of ADORA2A and/or ADORA2B blockers can be significantly boosted by CD73 inhibition (Young et al, 2016), pointing to a functional non‐redundancy that may be harnessed for therapeutic purposes. Also, while inhibiting ATP degradation or adenosine signaling mediates anticancer effects per se (at least in various models), blocking additional immunosuppressive pathways such as those controlled by co‐inhibitory receptors appears to provide improved disease control in most cases (Beavis et al, 2017; Leone et al, 2018; Goswami et al, 2020). Finally, it will be interesting to elucidate whether and how purinergic signaling can be targeted in patients with innate or acquired resistance to ICI‐based immunotherapy, reflecting the fact that exhausted T cells generally express high levels of CD39 (Canale et al, 2018). Irrespective of these and other incognita, purinergic signaling stands out as a particularly promising target for the development of novel anticancer agents.

Author contributions

LG conceived the article. OK and LG wrote the first version of the manuscript with critical input from LB, TY, FDV, MJS, and GK. OK, TY, and LB prepared display items under supervision from LG. All authors approved the final version of the article.

Conflict of interest

OK reports research funding from Daiichi Sankyo and a co‐founder role of Samsara therapeutics. FDV is a member of the Scientific Advisory Board of Biosceptre, Ltd. GK reports research funding from Bayer Healthcare, Eleor, Genentech, Glaxo Smyth Kline, Institut Mérieux, Lytix, PharmaMar, Sotio, and Vasculox (completed), funding from Samsara, consulting/advisory honoraria from The Longevity Labs and Lytix, membership of the Executive Board of Bristol Myers Squibb Foundation France, co‐founder role of everImmune, Samsara therapeutics and Therafast‐Bio. LG reports research funding from Lytix and Phosplatin (completed), consulting/advisory honoraria from Boehringer Ingelheim, AstraZeneca, OmniSEQ, The Longevity Labs, Inzen, Onxeo, and the Luke Heller TECPR2 Foundation. All other authors declare that they have no conflict to interest.

Acknowledgements

We are indebted to Giulia Petroni (Weill Cornell Medicine) for help with figure preparation. FDV is supported by the Italian Association for Cancer Research (AIRC) grant # IG13025 and by institutional funds from the University of Ferrara (Italy). MJS is supported by a National Health and Medical Research Council Investigator (1173958) and Program Grant (1132519). GK is supported by the Ligue contre le Cancer (équipe labellisée); Agence National de la Recherche (ANR)—Projets blancs; ANR under the frame of E‐Rare‐2, the ERA‐Net for Research on Rare Diseases; Association pour la recherche sur le cancer (ARC); Association Ruban Rose; Cancéropôle Ile‐de‐France; Fondation pour la Recherche Médicale (FRM); a donation by Elior; Gustave Roussy Odyssea, the European Union Horizon 2020 Project Oncobiome; Fondation Carrefour; High‐end Foreign Expert Program in China (GDW20171100085 and GDW20181100051), Institut National du Cancer (INCa); Inserm (HTE); Institut Universitaire de France; LeDucq Foundation; the LabEx Immuno‐Oncology; the RHU Torino Lumière; the Seerave Foundation; the SIRIC Stratified Oncology Cell DNA Repair and Tumor Immune Elimination (SOCRATE); and the SIRIC Cancer Research and Personalized Medicine (CARPEM). The LG laboratory is supported by a Breakthrough Level 2 grant from the US Department of Defense (DoD), Breast Cancer Research Program (BRCP) (#BC180476P1), by the 2019 Laura Ziskin Prize in Translational Research (#ZP‐6177, PI: Formenti) from the Stand Up to Cancer (SU2C), by a Mantle Cell Lymphoma Research Initiative (MCL‐RI, PI: Chen‐Kiang) grant from the Leukemia and Lymphoma Society (LLS), by a startup grant from the Dept. of Radiation Oncology at Weill Cornell Medicine (New York, US), by a Rapid Response Grant from the Functional Genomics Initiative (New York, US), by industrial collaborations with Lytix (Oslo, Norway) and Phosplatin (New York, US), and by donations from Phosplatin (New York, US), the Luke Heller TECPR2 Foundation (Boston, US), and Sotio a.s. (Prague, Czech Republic).

The EMBO Journal (2021) 40: e108130.

This article is part of the Cancer Reviews 2021 series.

Contributor Information

Guido Kroemer, Email: kroemer@orange.fr.

Lorenzo Galluzzi, Email: deadoc80@gmail.com.

References

  1. Adinolfi E, Melchiorri L, Falzoni S, Chiozzi P, Morelli A, Tieghi A, Cuneo A, Castoldi G, Di Virgilio F, Baricordi OR (2002) P2X7 receptor expression in evolutive and indolent forms of chronic B lymphocytic leukemia. Blood 99: 706–708 [DOI] [PubMed] [Google Scholar]
  2. Aliagas E, Vidal A, Texidó L, Ponce J, Condom E, Martín‐Satué M (2014) High expression of ecto‐nucleotidases CD39 and CD73 in human endometrial tumors. Mediators Inflamm 2014: 509027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Allard B, Allard D, Buisseret L, Stagg J (2020) The adenosine pathway in immuno‐oncology. Nat Rev Clin Oncol 17: 611–629 [DOI] [PubMed] [Google Scholar]
  4. Allard B, Cousineau I, Allard D, Buisseret L, Pommey S, Chrobak P, Stagg J (2019) Adenosine A2a receptor promotes lymphangiogenesis and lymph node metastasis. Oncoimmunology 8: 1601481 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Allard B, Pommey S, Smyth MJ, Stagg J (2013) Targeting CD73 enhances the antitumor activity of anti‐PD‐1 and anti‐CTLA‐4 mAbs. Clin Cancer Res 19: 5626–5635 [DOI] [PubMed] [Google Scholar]
  6. Allard B, Turcotte M, Spring K, Pommey S, Royal I, Stagg J (2014) Anti‐CD73 therapy impairs tumor angiogenesis. Int J Cancer 134: 1466–1473 [DOI] [PubMed] [Google Scholar]
  7. Anderson CM, Macleod KF (2019) Autophagy and cancer cell metabolism. Int Rev Cell Mol Biol 347: 145–190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Aquea G, Bresky G, Lancellotti D, Madariaga JA, Zaffiri V, Urzua U, Haberle S, Bernal G (2014) Increased expression of P2RY2, CD248 and EphB1 in gastric cancers from Chilean patients. Asian Pac J Cancer Prev 15: 1931–1936 [DOI] [PubMed] [Google Scholar]
  9. Aymeric L, Apetoh L, Ghiringhelli F, Tesniere A, Martins I, Kroemer G, Smyth MJ, Zitvogel L (2010) Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res 70: 855–858 [DOI] [PubMed] [Google Scholar]
  10. Bao L, Locovei S, Dahl G (2004) Pannexin membrane channels are mechanosensitive conduits for ATP. FEBS Lett 572: 65–68 [DOI] [PubMed] [Google Scholar]
  11. Beavis PA, Divisekera U, Paget C, Chow MT, John LB, Devaud C, Dwyer K, Stagg J, Smyth MJ, Darcy PK (2013) Blockade of A2A receptors potently suppresses the metastasis of CD73+ tumors. Proc Natl Acad Sci USA 110: 14711–14716 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Beavis PA, Henderson MA, Giuffrida L, Mills JK, Sek K, Cross RS, Davenport AJ, John LB, Mardiana S, Slaney CY et al (2017) Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J Clin Invest 127: 929–941 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Beavis PA, Milenkovski N, Henderson MA, John LB, Allard B, Loi S, Kershaw MH, Stagg J, Darcy PK (2015) Adenosine receptor 2A blockade increases the efficacy of anti‐PD‐1 through enhanced antitumor T‐cell responses. Cancer Immunol Res 3: 506–517 [DOI] [PubMed] [Google Scholar]
  14. Beckel JM, Gómez NM, Lu W, Campagno KE, Nabet B, Albalawi F, Lim JC, Boesze‐Battaglia K, Mitchell CH (2018) Stimulation of TLR3 triggers release of lysosomal ATP in astrocytes and epithelial cells that requires TRPML1 channels. Sci Rep 8: 5726 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bendell JC, Manji GA, Pant S, Lai DW, Colabella J, Berry W, Paoloni MC, Grossman WJ, O'Reilly EM (2020) A phase I study to evaluate the safety and tolerability of AB680 combination therapy in participants with gastrointestinal malignancies. J Clin Oncol 38: TPS788 [Google Scholar]
  16. Bergers G, Fendt SM (2021) The metabolism of cancer cells during metastasis. Nat Rev Cancer 21: 162–180 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bidula S, Dhuna K, Helliwell R, Stokes L (2019) Positive allosteric modulation of P2X7 promotes apoptotic cell death over lytic cell death responses in macrophages. Cell Death Dis 10: 882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Borsellino G, Kleinewietfeld M, Di Mitri D, Sternjak A, Diamantini A, Giometto R, Höpner S, Centonze D, Bernardi G, Dell'Acqua ML et al (2007) Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood 110: 1225–1232 [DOI] [PubMed] [Google Scholar]
  19. Brand A, Singer K, Koehl G, Kolitzus M, Schoenhammer G, Thiel A, Matos C, Bruss C, Klobuch S, Peter K et al (2016) LDHA‐associated lactic acid production blunts tumor immunosurveillance by T and NK cells. Cell Metab 24: 657–671 [DOI] [PubMed] [Google Scholar]
  20. Buisseret L, Pommey S, Allard B, Garaud S, Bergeron M, Cousineau I, Ameye L, Bareche Y, Paesmans M, Crown J et al (2018) Clinical significance of CD73 in triple‐negative breast cancer: multiplex analysis of a phase III clinical trial. Ann Oncol 29: 1056–1062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Buqué A, Bloy N, Perez‐Lanzón M, Iribarren K, Humeau J, Pol JG, Levesque S, Mondragon L, Yamazaki T, Sato Ai et al (2020) Immunoprophylactic and immunotherapeutic control of hormone receptor‐positive breast cancer. Nat Commun 11: 3819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Canale FP, Ramello MC, Núñez N, Araujo Furlan CL, Bossio SN, Gorosito Serrán M, Tosello Boari J, Del Castillo A, Ledesma M, Sedlik C et al (2018) CD39 expression defines cell exhaustion in tumor‐infiltrating CD8(+) T cells. Cancer Res 78: 115–128 [DOI] [PubMed] [Google Scholar]
  23. Cao Q, Zhao K, Zhong XZ, Zou Y, Yu H, Huang P, Xu TL, Dong XP (2014) SLC17A9 protein functions as a lysosomal ATP transporter and regulates cell viability. J Biol Chem 289: 23189–23199 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Carozza JA, Brown JA, Bohnert V, Fernandez D, AlSaif Y, Mardjuki RE, Smith M, Li L (2020) Structure‐aided development of small‐molecule inhibitors of ENPP1, the extracellular phosphodiesterase of the immunotransmitter cGAMP. Cell Chem Biol 27: 1347–1358 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Castoldi F, Humeau J, Martins I, Lachkar S, Loew D, Dingli F, Durand S, Enot D, Bossut N, Chery A et al (2020) Autophagy‐mediated metabolic effects of aspirin. Cell Death Discov 6: 129 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Cekic C, Day YJ, Sag D, Linden J (2014) Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res 74: 7250–7259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chang C‐H, Qiu J, O’Sullivan D, Buck M, Noguchi T, Curtis J, Chen Q, Gindin M, Gubin M, van der Windt G et al (2015) Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162: 1229–1241 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chekeni FB, Elliott MR, Sandilos JK, Walk SF, Kinchen JM, Lazarowski ER, Armstrong AJ, Penuela S, Laird DW, Salvesen GS et al (2010) Pannexin 1 channels mediate 'find‐me' signal release and membrane permeability during apoptosis. Nature 467: 863–867 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chen G, Xie W, Nah J, Sauvat A, Liu P, Pietrocola F, Sica V, Carmona‐Gutierrez D, Zimmermann A, Pendl T et al (2019) 3,4‐Dimethoxychalcone induces autophagy through activation of the transcription factors TFE3 and TFEB. EMBO Mol Med 11: e10469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Chen Q, Pu N, Yin H, Zhang J, Zhao G, Lou W, Wu W (2020a) CD73 acts as a prognostic biomarker and promotes progression and immune escape in pancreatic cancer. J Cell Mol Med 24: 8674–8686 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Chen S, Akdemir I, Fan J, Linden J, Zhang B, Cekic C (2020b) The expression of adenosine A2B receptor on antigen‐presenting cells suppresses CD8(+) T‐cell responses and promotes tumor growth. Cancer Immunol Res 8: 1064–1074 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Chen X, Sun Y, Wang B, Wang H (2020c) Prognostic significance of autophagy‐related genes Beclin1 and LC3 in ovarian cancer: a meta‐analysis. J Int Med Res 48: 300060520968299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Chiappori A, Williams CC, Creelan BC, Tanvetyanon T, Gray JE, Haura EB, Thapa R, Chen D‐T, Beg AA, Boyle TA et al (2018) Phase I/II study of the A2AR antagonist NIR178 (PBF‐509), an oral immunotherapy, in patients (pts) with advanced NSCLC. J Clin Oncol 36: 9089 [Google Scholar]
  34. Clarke AJ, Simon AK (2019) Autophagy in the renewal, differentiation and homeostasis of immune cells. Nat Rev Immunol 19: 170–183 [DOI] [PubMed] [Google Scholar]
  35. Cohen HB, Ward A, Hamidzadeh K, Ravid K, Mosser DM (2015) IFN‐γ prevents adenosine receptor (A2bR) upregulation to sustain the macrophage activation response. J Immunol 195: 3828–3837 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Cui L, Wang X, Zhao X, Kong C, Li Z, Liu Y, Jiang X, Zhang X (2019) The autophagy‐related genes Beclin1 and LC3 in the prognosis of pancreatic cancer. Int J Clin Exp Pathol 12: 2989–2996 [PMC free article] [PubMed] [Google Scholar] [Retracted]
  37. d’Almeida SM, Kauffenstein G, Roy C, Basset L, Papargyris L, Henrion D, Catros V, Ifrah N, Descamps P, Croue A et al (2016) The ecto‐ATPDase CD39 is involved in the acquisition of the immunoregulatory phenotype by M‐CSF‐macrophages and ovarian cancer tumor‐associated macrophages: regulatory role of IL‐27. Oncoimmunology 5: e1178025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Dahl G (2015) ATP release through pannexon channels. Philos Trans R Soc Lond B Biol Sci 370: 20140191 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Dardano A, Falzoni S, Caraccio N, Polini A, Tognini S, Solini A, Berti P, Di Virgilio F, Monzani F (2009) 1513A>C polymorphism in the P2X7 receptor gene in patients with papillary thyroid cancer: correlation with histological variants and clinical parameters. J Clin Endocrinol Metab 94: 695–698 [DOI] [PubMed] [Google Scholar]
  40. de Lourdes Mora‐García M, López‐Cisneros S, Gutiérrez‐Serrano V, García‐Rocha R, Weiss‐Steider B, Hernández‐Montes J, Sánchez‐Peña HI, Ávila‐Ibarra LR, Don‐López CA, Muñóz‐Godínez R et al (2019) HPV‐16 Infection is associated with a high content of CD39 and CD73 Ectonucleotidases in cervical samples from patients with CIN‐1. Mediators Inflamm 2019: 4651627 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. De Marchi E, Orioli E, Pegoraro A, Sangaletti S, Portararo P, Curti A, Colombo MP, Di Virgilio F, Adinolfi E (2019) The P2X7 receptor modulates immune cells infiltration, ectonucleotidases expression and extracellular ATP levels in the tumor microenvironment. Oncogene 38: 3636–3650 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. De Sousa EMF, Vermeulen L, Fessler E, Medema JP (2013) Cancer heterogeneity–a multifaceted view. EMBO Rep 14: 686–695 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Deng WW, Li YC, Ma SR, Mao L, Yu GT, Bu LL, Kulkarni AB, Zhang WF, Sun ZJ (2018) Specific blockade CD73 alters the "exhausted" phenotype of T cells in head and neck squamous cell carcinoma. Int J Cancer 143: 1494–1504 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Dersh D, Hollý J, Yewdell JW (2021) A few good peptides: MHC class I‐based cancer immunosurveillance and immunoevasion. Nat Rev Immunol 21: 116–128 [DOI] [PubMed] [Google Scholar]
  45. Di Virgilio F, Sarti AC, Falzoni S, De Marchi E, Adinolfi E (2018) Extracellular ATP and P2 purinergic signalling in the tumour microenvironment. Nat Rev Cancer 18: 601–618 [DOI] [PubMed] [Google Scholar]
  46. Dosch M, Gerber J, Jebbawi F, Beldi G (2018) Mechanisms of ATP release by inflammatory cells. Int J Mol Sci 19: 1222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Dosch M, Zindel J, Jebbawi F, Melin N, Sanchez‐Taltavull D, Stroka D, Candinas D, Beldi G (2019) Connexin‐43‐dependent ATP release mediates macrophage activation during sepsis. Elife 8: e42670 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Douguet L, Janho dit Hreich S, Benzaquen J, Seguin L, Juhel T, Dezitter X, Duranton C, Ryffel B, Kanellopoulos J, Delarasse C et al (2021) A small‐molecule P2RX7 activator promotes anti‐tumor immune responses and sensitizes lung tumor to immunotherapy. Nat Commun 12: 653 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Duan S, Nordmeier S, Byrnes AE, Buxton ILO (2021) Extracellular vesicle‐mediated purinergic signaling contributes to host microenvironment plasticity and metastasis in triple negative breast cancer. Int J Mol Sci 22: 597 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Dunn GP, Bruce AT, Ikeda H, Old LJ, Schreiber RD (2002) Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 3: 991–998 [DOI] [PubMed] [Google Scholar]
  51. Elliott MR, Chekeni FB, Trampont PC, Lazarowski ER, Kadl A, Walk SF, Park D, Woodson RI, Ostankovich M, Sharma P et al (2009) Nucleotides released by apoptotic cells act as a find‐me signal to promote phagocytic clearance. Nature 461: 282–286 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Eltzschig HK, Eckle T, Mager A, Küper N, Karcher C, Weissmüller T, Boengler K, Schulz R, Robson SC, Colgan SP (2006) ATP release from activated neutrophils occurs via connexin 43 and modulates adenosine‐dependent endothelial cell function. Circ Res 99: 1100–1108 [DOI] [PubMed] [Google Scholar]
  53. Facon T, Kumar S, Plesner T, Orlowski RZ, Moreau P, Bahlis N, Basu S, Nahi H, Hulin C, Quach H et al (2019) Daratumumab plus Lenalidomide and Dexamethasone for Untreated Myeloma. N Engl J Med 380: 2104–2115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Fong L, Hotson A, Powderly JD, Sznol M, Heist RS, Choueiri TK, George S, Hughes BGM, Hellmann MD, Shepard DR et al (2020) Adenosine 2A receptor blockade as an immunotherapy for treatment‐refractory renal cell cancer. Cancer Discov 10: 40–53 [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Fridman WH, Zitvogel L, Sautès‐Fridman C, Kroemer G (2017) The immune contexture in cancer prognosis and treatment. Nat Rev Clin Oncol 14: 717–734 [DOI] [PubMed] [Google Scholar]
  56. Gaertner F, Massberg S (2019) Patrolling the vascular borders: platelets in immunity to infection and cancer. Nat Rev Immunol 19: 747–760 [DOI] [PubMed] [Google Scholar]
  57. Gallerano D, Ciminati S, Grimaldi A, Piconese S, Cammarata I, Focaccetti C, Pacella I, Accapezzato D, Lancellotti F, Sacco L et al (2020) Genetically driven CD39 expression shapes human tumor‐infiltrating CD8(+) T‐cell functions. Int J Cancer 147: 2597–2610 [DOI] [PubMed] [Google Scholar]
  58. Galluzzi L, Chan TA, Kroemer G, Wolchok JD, Lopez‐Soto A (2018) The hallmarks of successful anticancer immunotherapy. Sci Transl Med 10: eaat7807 [DOI] [PubMed] [Google Scholar]
  59. Galluzzi L, Humeau J, Buqué A, Zitvogel L, Kroemer G (2020) Immunostimulation with chemotherapy in the era of immune checkpoint inhibitors. Nat Rev Clin Oncol 17: 725–741 [DOI] [PubMed] [Google Scholar]
  60. Gao ZW, Wang HP, Lin F, Wang X, Long M, Zhang HZ, Dong K (2017) CD73 promotes proliferation and migration of human cervical cancer cells independent of its enzyme activity. BMC Cancer 17: 135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Garner H, de Visser KE (2020) Immune crosstalk in cancer progression and metastatic spread: a complex conversation. Nat Rev Immunol 20: 483–497 [DOI] [PubMed] [Google Scholar]
  62. Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, Vermaelen K, Panaretakis T, Mignot G, Ullrich E et al (2009) Activation of the NLRP3 inflammasome in dendritic cells induces IL‐1beta‐dependent adaptive immunity against tumors. Nat Med 15: 1170–1178 [DOI] [PubMed] [Google Scholar]
  63. Giatromanolaki A, Kouroupi M, Pouliliou S, Mitrakas A, Hasan F, Pappa A, Koukourakis MI (2020) Ectonucleotidase CD73 and CD39 expression in non‐small cell lung cancer relates to hypoxia and immunosuppressive pathways. Life Sci 259: 118389 [DOI] [PubMed] [Google Scholar]
  64. Goswami S, Walle T, Cornish AE, Basu S, Anandhan S, Fernandez I, Vence L, Blando J, Zhao H, Yadav SS et al (2020) Immune profiling of human tumors identifies CD73 as a combinatorial target in glioblastoma. Nat Med 26: 39–46 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next generation. Cell 144: 646–674 [DOI] [PubMed] [Google Scholar]
  66. Hatfield SM, Kjaergaard J, Lukashev D, Belikoff B, Schreiber TH, Sethumadhavan S, Abbott R, Philbrook P, Thayer M, Shujia D et al (2014) Systemic oxygenation weakens the hypoxia and hypoxia inducible factor 1α‐dependent and extracellular adenosine‐mediated tumor protection. J Mol Med (Berl) 92: 1283–1292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Hatfield SM, Sitkovsky MV (2020) Antihypoxic oxygenation agents with respiratory hyperoxia to improve cancer immunotherapy. J Clin Invest 130: 5629–5637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Häusler SF, Del Barrio IM, Diessner J, Stein RG, Strohschein J, Hönig A, Dietl J, Wischhusen J (2014) Anti‐CD39 and anti‐CD73 antibodies A1 and 7G2 improve targeted therapy in ovarian cancer by blocking adenosine‐dependent immune evasion. Am J Transl Res 6: 129–139 [PMC free article] [PubMed] [Google Scholar]
  69. Häusler SFM, Montalbán del Barrio I, Strohschein J, Anoop Chandran P, Engel JB, Hönig A, Ossadnik M, Horn E, Fischer B, Krockenberger M et al (2011) Ectonucleotidases CD39 and CD73 on OvCA cells are potent adenosine‐generating enzymes responsible for adenosine receptor 2A‐dependent suppression of T cell function and NK cell cytotoxicity. Cancer Immunol Immunother 60: 1405–1418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Hay CM, Sult E, Huang Q, Mulgrew K, Fuhrmann SR, McGlinchey KA, Hammond SA, Rothstein R, Rios‐Doria J, Poon E et al (2016) Targeting CD73 in the tumor microenvironment with MEDI9447. Oncoimmunology 5: e1208875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Ho WJ, Jaffee EM, Zheng L (2020) The tumour microenvironment in pancreatic cancer ‐ clinical challenges and opportunities. Nat Rev Clin Oncol 17: 527–540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Husain Z, Huang Y, Seth P, Sukhatme VP (2013) Tumor‐derived lactate modifies antitumor immune response: effect on myeloid‐derived suppressor cells and NK cells. J Immunol 191: 1486–1495 [DOI] [PubMed] [Google Scholar]
  73. Iannone R, Miele L, Maiolino P, Pinto A, Morello S (2014) Adenosine limits the therapeutic effectiveness of anti‐CTLA4 mAb in a mouse melanoma model. Am J Cancer Res 4: 172–181 [PMC free article] [PubMed] [Google Scholar]
  74. Imura Y, Morizawa Y, Komatsu R, Shibata K, Shinozaki Y, Kasai H, Moriishi K, Moriyama Y, Koizumi S (2013) Microglia release ATP by exocytosis. Glia 61: 1320–1330 [DOI] [PubMed] [Google Scholar]
  75. Jiang L, Huang S, Li W, Zhang D, Zhang S, Zhang W, Zheng P, Chen Z (2012) Expression of autophagy and ER stress‐related proteins in primary salivary adenoid cystic carcinoma. Pathol Res Pract 208: 635–641 [DOI] [PubMed] [Google Scholar]
  76. Kamai T, Kijima T, Tsuzuki T, Nukui A, Abe H, Arai K, Yoshida KI (2021) Increased expression of adenosine 2A receptors in metastatic renal cell carcinoma is associated with poorer response to anti‐vascular endothelial growth factor agents and anti‐PD‐1/Anti‐CTLA4 antibodies and shorter survival. Cancer Immunol Immunother [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kamata‐Sakurai M, Narita Y, Hori Y, Nemoto T, Uchikawa R, Honda M, Hironiwa N, Taniguchi K, Shida‐Kawazoe M, Metsugi S et al (2020) Antibody to CD137 activated by extracellular adenosine triphosphate is tumor selective and broadly effective in vivo without systemic immune activation. Cancer Discov 11: 158–175 [DOI] [PubMed] [Google Scholar]
  78. Kang J, Kang N, Lovatt D, Torres A, Zhao Z, Lin J, Nedergaard M (2008) Connexin 43 hemichannels are permeable to ATP. J Neurosci 28: 4702–4711 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Kashyap AS, Thelemann T, Klar R, Kallert SM, Festag J, Buchi M, Hinterwimmer L, Schell M, Michel S, Jaschinski F et al (2019) Antisense oligonucleotide targeting CD39 improves anti‐tumor T cell immunity. J Immunother Cancer 7: 67 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Kayagaki N, Kornfeld OS, Lee BL, Stowe IB, O’Rourke K, Li Q, Sandoval W, Yan D, Kang J, Xu M et al (2021) NINJ1 mediates plasma membrane rupture during lytic cell death. Nature 591: 131–136 [DOI] [PubMed] [Google Scholar]
  81. Kayhan M, Koyas A, Akdemir I, Savas AC, Cekic C (2019) Adenosine receptor signaling targets both PKA and Epac pathways to polarize dendritic cells to a suppressive phenotype. J Immunol 203: 3247–3255 [DOI] [PubMed] [Google Scholar]
  82. Kepp O, Kroemer G (2020) Autophagy induction by thiostrepton for the improvement of anticancer therapy. Autophagy 16: 1166–1167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kim JS, Bae GE, Kim KH, Lee SI, Chung C, Lee D, Lee TH, Kwon IS, Yeo MK (2019) Prognostic significance of LC3B and p62/SQSTM1 expression in gastric adenocarcinoma. Anticancer Res 39: 6711–6722 [DOI] [PubMed] [Google Scholar]
  84. Kjaergaard J, Hatfield S, Jones G, Ohta A, Sitkovsky M (2018) A(2A) adenosine receptor gene deletion or synthetic A(2A) antagonist liberate tumor‐reactive CD8(+) T cells from tumor‐induced immunosuppression. J Immunol 201: 782–791 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Klein M, Bopp T (2016) Cyclic AMP represents a crucial component of Treg cell‐mediated immune regulation. Front Immunol 7: 315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Ko YH, Cho YS, Won HS, Jeon EK, An HJ, Hong SU, Park JH, Lee MA (2013) Prognostic significance of autophagy‐related protein expression in resected pancreatic ductal adenocarcinoma. Pancreas 42: 829–835 [DOI] [PubMed] [Google Scholar]
  87. Koh J, Kim Y, Lee KY, Hur JY, Kim MS, Kim B, Cho HJ, Lee YC, Bae YH, Ku BM et al (2020) MDSC subtypes and CD39 expression on CD8(+) T cells predict the efficacy of anti‐PD‐1 immunotherapy in patients with advanced NSCLC. Eur J Immunol 50: 1810–1819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Kroemer G, Senovilla L, Galluzzi L, André F, Zitvogel L (2015) Natural and therapy‐induced immunosurveillance in breast cancer. Nat Med 21: 1128–1138 [DOI] [PubMed] [Google Scholar]
  89. Krstic J, Pieber TR, Prokesch A (2020) Stratifying nutritional restriction in cancer therapy: next stop, personalized medicine. Int Rev Cell Mol Biol 354: 231–259 [DOI] [PubMed] [Google Scholar]
  90. Künzli BM, Berberat PO, Giese T, Csizmadia E, Kaczmarek E, Baker C, Halaceli I, Büchler MW, Friess H, Robson SC (2007) Upregulation of CD39/NTPDases and P2 receptors in human pancreatic disease. Am J Physiol Gastrointest Liver Physiol 292: G223–G230 [DOI] [PubMed] [Google Scholar]
  91. Ladoire S, Enot D, Senovilla L, Ghiringhelli F, Poirier‐Colame V, Chaba K, Semeraro M, Chaix M, Penault‐Llorca F, Arnould L et al (2016) The presence of LC3B puncta and HMGB1 expression in malignant cells correlate with the immune infiltrate in breast cancer. Autophagy 12: 864–875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Ladoire S, Penault‐Llorca F, Senovilla L, Dalban C, Enot D, Locher C, Prada N, Poirier‐Colame V, Chaba K, Arnould L et al (2015) Combined evaluation of LC3B puncta and HMGB1 expression predicts residual risk of relapse after adjuvant chemotherapy in breast cancer. Autophagy 11: 1878–1890 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Lan J, Lu H, Samanta D, Salman S, Lu Y, Semenza GL (2018) Hypoxia‐inducible factor 1‐dependent expression of adenosine receptor 2B promotes breast cancer stem cell enrichment. Proc Natl Acad Sci USA 115: E9640–E9648 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Lappas CM, Rieger JM, Linden J (2005) A2A adenosine receptor induction inhibits IFN‐gamma production in murine CD4+ T cells. J Immunol 174: 1073–1080 [DOI] [PubMed] [Google Scholar]
  95. Leclerc BG, Charlebois R, Chouinard G, Allard B, Pommey S, Saad F, Stagg J (2016) CD73 expression is an independent prognostic factor in prostate cancer. Clin Cancer Res 22: 158–166 [DOI] [PubMed] [Google Scholar]
  96. Lee YJ, Ha YJ, Na Kang Yu, Kang KJ, Hwang JS, Chung WJ, Cho KB, Park KS, Kim ES, Seo H‐Y et al (2013) The autophagy‐related marker LC3 can predict prognosis in human hepatocellular carcinoma. PLoS One 8: e81540 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Lee YS, Radford KJ (2019) The role of dendritic cells in cancer. Int Rev Cell Mol Biol 348: 123–178 [DOI] [PubMed] [Google Scholar]
  98. Leone RD, Powell JD (2020) Metabolism of immune cells in cancer. Nat Rev Cancer 20: 516–531 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Leone RD, Sun IM, Oh MH, Sun IH, Wen J, Englert J, Powell JD (2018) 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 67: 1271–1284 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Li C, Chen Q, Tian Z, Li S, Gong Z, Lin Z, Wang B, Liu H (2019a) Expression of MIF, Beclin1, and LC3 in human salivary gland adenoid cystic carcinoma and its prognostic value. Medicine 98: e15402 [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Li H, Lv M, Qiao B, Li X (2019b) Blockade pf CD73/adenosine axis improves the therapeutic efficacy of docetaxel in epithelial ovarian cancer. Arch Gynecol Obstet 299: 1737–1746 [DOI] [PubMed] [Google Scholar]
  102. Li J, Wang L, Chen X, Li L, Li Yu, Ping Yu, Huang L, Yue D, Zhang Z, Wang F et al (2017) CD39/CD73 upregulation on myeloid‐derived suppressor cells via TGF‐β‐mTOR‐HIF‐1 signaling in patients with non‐small cell lung cancer. Oncoimmunology 6: e1320011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Li JX, Yan Q, Liu N, Zheng WJ, Hu M, Yu ZM, Zhou YD, Wang XW, Liang FX, Chen R (2020) The prognostic value of autophagy‐related markers Bclin‐1 and LC‐3 in colorectal cancers: a systematic review and meta‐analysis. Evid Based Complement Alternat Med 2020: 8475840 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Li X‐Y, Moesta AK, Xiao C, Nakamura K, Casey M, Zhang H, Madore J, Lepletier A, Aguilera AR, Sundarrajan A et al (2019c) Targeting CD39 in cancer reveals an extracellular ATP‐ and inflammasome‐driven tumor immunity. Cancer Discov 9: 1754–1773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Lim EA, Bauer TM, Patel MR, Falchook GS, Karlix JL, Choe JH, George DJ, Mugundu GM, Pilling E, Chen H et al (2020) A phase I, open‐label, multicenter study to assess the safety, pharmacokinetics, and preliminary antitumor activity of AZD4635 both as monotherapy and in combination in patients with advanced solid malignancies: Results from prostate cancer patients (NCT02740985). J Clin Oncol 38: 5518 [Google Scholar]
  106. Lim To WK, Kumar P, Marshall JM (2015) Hypoxia is an effective stimulus for vesicular release of ATP from human umbilical vein endothelial cells. Placenta 36: 759–766 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Loi S, Pommey S, Haibe‐Kains B, Beavis PA, Darcy PK, Smyth MJ, Stagg J (2013) CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc Natl Acad Sci USA 110: 11091–11096 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Lopez‐Otin C, Kroemer G (2021) Hallmarks of health. Cell 184: 33–63 [DOI] [PubMed] [Google Scholar]
  109. Ludwig N, Yerneni SS, Azambuja JH, Gillespie DG, Menshikova EV, Jackson EK, Whiteside TL (2020) Tumor‐derived exosomes promote angiogenesis via adenosine A(2B) receptor signaling. Angiogenesis 23: 599–610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Ma Y, Adjemian S, Mattarollo S, Yamazaki T, Aymeric L, Yang H, Portela Catani J, Hannani D, Duret H, Steegh K et al (2013) Anticancer chemotherapy‐induced intratumoral recruitment and differentiation of antigen‐presenting cells. Immunity 38: 729–741 [DOI] [PubMed] [Google Scholar]
  111. Ma Y, Aymeric L, Locher C, Mattarollo SR, Delahaye NF, Pereira P, Boucontet L, Apetoh L, Ghiringhelli F, Casares N et al (2011) Contribution of IL‐17‐producing gamma delta T cells to the efficacy of anticancer chemotherapy. J Exp Med 208: 491–503 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Mantovani A, Marchesi F, Malesci A, Laghi L, Allavena P (2017) Tumour‐associated macrophages as treatment targets in oncology. Nat Rev Clin Oncol 14: 399–416 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Martin S, Dudek‐Peric AM, Garg AD, Roose H, Demirsoy S, Van Eygen S, Mertens F, Vangheluwe P, Vankelecom H, Agostinis P (2017) An autophagy‐driven pathway of ATP secretion supports the aggressive phenotype of BRAF(V600E) inhibitor‐resistant metastatic melanoma cells. Autophagy 13: 1512–1527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Martinek J, Wu TC, Cadena D, Banchereau J, Palucka K (2019) Interplay between dendritic cells and cancer cells. Int Rev Cell Mol Biol 348: 179–215 [DOI] [PubMed] [Google Scholar]
  115. Martinez‐Outschoorn UE, Peiris‐Pagés M, Pestell RG, Sotgia F, Lisanti MP (2017) Cancer metabolism: a therapeutic perspective. Nat Rev Clin Oncol 14: 113 [DOI] [PubMed] [Google Scholar]
  116. Martins I, Wang Y, Michaud M, Ma Y, Sukkurwala AQ, Shen S, Kepp O, Métivier D, Galluzzi L, Perfettini J‐l et al (2014) Molecular mechanisms of ATP secretion during immunogenic cell death. Cell Death Differ 21: 79–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Medina CB, Mehrotra P, Arandjelovic S, Perry JSA, Guo Y, Morioka S, Barron B, Walk SF, Ghesquière B, Krupnick AS et al (2020) Metabolites released from apoptotic cells act as tissue messengers. Nature 580: 130–135 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Medler T, Patel JM, Alice A, Baird JR, Hu HM, Gough MJ (2019) Activating the nucleic acid‐sensing machinery for anticancer immunity. Int Rev Cell Mol Biol 344: 173–214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Messaoudi N, Cousineau I, Arslanian E, Henault D, Stephen D, Vandenbroucke‐Menu F, Dagenais M, Létourneau R, Plasse M, Roy A et al (2020) Prognostic value of CD73 expression in resected colorectal cancer liver metastasis. Oncoimmunology 9: 1746138 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Michaud M, Martins I, Sukkurwala Aq, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G et al (2011) Autophagy‐dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 334: 1573–1577 [DOI] [PubMed] [Google Scholar]
  121. Minguet S, Huber M, Rosenkranz L, Schamel WW, Reth M, Brummer T (2005) Adenosine and cAMP are potent inhibitors of the NF‐kappa B pathway downstream of immunoreceptors. Eur J Immunol 35: 31–41 [DOI] [PubMed] [Google Scholar]
  122. Mittal D, Sinha D, Barkauskas D, Young A, Kalimutho M, Stannard K, Caramia F, Haibe‐Kains B, Stagg J, Khanna KK et al (2016) Adenosine 2B receptor expression on cancer cells promotes metastasis. Cancer Res 76: 4372–4382 [DOI] [PubMed] [Google Scholar]
  123. Mittal D, Young A, Stannard K, Yong M, Teng MW, Allard B, Stagg J, Smyth MJ (2014) Antimetastatic effects of blocking PD‐1 and the adenosine A2A receptor. Cancer Res 74: 3652–3658 [DOI] [PubMed] [Google Scholar]
  124. Mobasher M, Miller RA, Kwei L, Strahs D, Das V, Luciano G, Powderly JD, Merchan JR, Barve MA, LoRusso P et al (2019) A phase I/Ib multicenter study to evaluate the humanized anti‐CD73 antibody, CPI‐006, as a single agent, in combination with CPI‐444, and in combination with pembrolizumab in adult patients with advanced cancers. J Clin Oncol 37: TPS2646‐TPS2646 [Google Scholar]
  125. Moesta AK, Li XY, Smyth MJ (2020) Targeting CD39 in cancer. Nat Rev Immunol 20: 739–755 [DOI] [PubMed] [Google Scholar]
  126. Morandi F, Morandi B, Horenstein AL, Chillemi A, Quarona V, Zaccarello G, Carrega P, Ferlazzo G, Mingari MC, Moretta L et al (2015) A non‐canonical adenosinergic pathway led by CD38 in human melanoma cells induces suppression of T cell proliferation. Oncotarget 6: 25602–25618 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Murata N, Ito S, Furuya K, Takahara N, Naruse K, Aso H, Kondo M, Sokabe M, Hasegawa Y (2014) Ca2+ influx and ATP release mediated by mechanical stretch in human lung fibroblasts. Biochem Biophys Res Commun 453: 101–105 [DOI] [PubMed] [Google Scholar]
  128. Nagate Y, Ezoe S, Fujita J, Okuzaki D, Motooka D, Ishibashi T, Ichii M, Tanimura A, Kurashige M, Morii E et al (2021) Ectonucleotidase CD39 is highly expressed on ATLL cells and is responsible for their immunosuppressive function. Leukemia 35: 107–118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Nakamura K, Casey M, Oey H, Vari F, Stagg J, Gandhi MK, Smyth MJ (2020) Targeting an adenosine‐mediated "don't eat me signal" augments anti‐lymphoma immunity by anti‐CD20 monoclonal antibody. Leukemia 34: 2708–2721 [DOI] [PubMed] [Google Scholar]
  130. Narahari AK, Kreutzberger AJ, Gaete PS, Chiu YH, Leonhardt SA, Medina CB, Jin X, Oleniacz PW, Kiessling V, Barrett PQ et al (2021) ATP and large signaling metabolites flux through caspase‐activated Pannexin 1 channels. Elife 10: e64787 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Németh ZH, Lutz CS, Csóka B, Deitch EA, Leibovich SJ, Gause WC, Tone M, Pacher P, Vizi ES, Haskó G (2005) Adenosine augments IL‐10 production by macrophages through an A2B receptor‐mediated posttranscriptional mechanism. J Immunol 175: 8260–8270 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Neo SY, Yang Y, Record J, Ma R, Chen X, Chen Z, Tobin NP, Blake E, Seitz C, Thomas R et al (2020) CD73 immune checkpoint defines regulatory NK cells within the tumor microenvironment. J Clin Invest 130: 1185–1198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Nückel H, Frey UH, Dürig J, Dührsen U, Siffert W (2004) 1513A/C polymorphism in the P2X7 receptor gene in chronic lymphocytic leukemia: absence of correlation with clinical outcome. Eur J Haematol 72: 259–263 [DOI] [PubMed] [Google Scholar]
  134. Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong M, Huang X, Caldwell S, Liu K, Smith P et al (2006) A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci USA 103: 13132–13137 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. O'Sullivan D, Sanin DE, Pearce EJ, Pearce EL (2019) Metabolic interventions in the immune response to cancer. Nat Rev Immunol 19: 324–335 [DOI] [PubMed] [Google Scholar]
  136. Paneesha S, Starczynski J, Pepper C, Delgado J, Hooper L, Fegan C, Pratt G (2006) The P2X7 receptor gene polymorphism 1513 A–>C has no effect on clinical prognostic markers and survival in multiple myeloma. Leuk Lymphoma 47: 281–284 [DOI] [PubMed] [Google Scholar]
  137. Pellegatti P, Raffaghello L, Bianchi G, Piccardi F, Pistoia V, Di Virgilio F (2008) Increased level of extracellular ATP at tumor sites: in vivo imaging with plasma membrane luciferase. PLoS One 3: e2599 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Perregaux DG, McNiff P, Laliberte R, Conklyn M, Gabel CA (2000) ATP acts as an agonist to promote stimulus‐induced secretion of IL‐1 beta and IL‐18 in human blood. J Immunol 165: 4615–4623 [DOI] [PubMed] [Google Scholar]
  139. Perrot I, Michaud H‐A, Giraudon‐Paoli M, Augier S, Docquier A, Gros L, Courtois R, Déjou C, Jecko D, Becquart O et al (2019) Blocking antibodies targeting the CD39/CD73 immunosuppressive pathway unleash immune responses in combination cancer therapies. Cell Rep 27: 2411–2425 [DOI] [PubMed] [Google Scholar]
  140. Perry C, Hazan‐Halevy I, Kay S, Cipok M, Grisaru D, Deutsch V, Polliack A, Naparstek E, Herishanu Y (2012) Increased CD39 expression on CD4(+) T lymphocytes has clinical and prognostic significance in chronic lymphocytic leukemia. Ann Hematol 91: 1271–1279 [DOI] [PubMed] [Google Scholar]
  141. Petroni G, Buque A, Zitvogel L, Kroemer G, Galluzzi L (2021) Immunomodulation by targeted anticancer agents. Cancer Cell 39: 310–345 [DOI] [PubMed] [Google Scholar]
  142. Pietrocola F, Pol J, Vacchelli E, Rao S, Enot D, Baracco E, Levesque S, Castoldi F, Jacquelot N, Yamazaki T et al (2016) Caloric restriction mimetics enhance anticancer immunosurveillance. Cancer Cell 30: 147–160 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Pinheiro AR, Paramos‐de‐Carvalho D, Certal M, Costa MA, Costa C, Magalhães‐Cardoso MT, Ferreirinha F, Sévigny J, Correia‐de‐Sá P (2013) Histamine induces ATP release from human subcutaneous fibroblasts, via pannexin‐1 hemichannels, leading to Ca2+ mobilization and cell proliferation. J Biol Chem 288: 27571–27583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Pulte D, Furman RR, Broekman MJ, Drosopoulos JH, Ballard HS, Olson KE, Kizer JR, Marcus AJ (2011) CD39 expression on T lymphocytes correlates with severity of disease in patients with chronic lymphocytic leukemia. Clin Lymphoma Myeloma Leuk 11: 367–372 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Qi Yu, Xia Yu, Lin Z, Qu Y, Qi Y, Chen Y, Zhou Q, Zeng H, Wang J, Chang Y et al (2020) Tumor‐infiltrating CD39(+)CD8(+) T cells determine poor prognosis and immune evasion in clear cell renal cell carcinoma patients. Cancer Immunol Immunother 69: 1565–1576 [DOI] [PMC free article] [PubMed] [Google Scholar]
  146. Qin Z, Yu X, Lin M, Wu J, Ma S, Wang N (2018) Prognostic and clinicopathological value of Beclin‐1 expression in hepatocellular carcinoma: a meta‐analysis. World J Surg Oncol 16: 170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Rao S, Gharib K, Han A (2019) Cancer immunosurveillance by T cells. Int Rev Cell Mol Biol 342: 149–173 [DOI] [PubMed] [Google Scholar]
  148. Raskovalova T, Huang X, Sitkovsky M, Zacharia LC, Jackson EK, Gorelik E (2005) Gs protein‐coupled adenosine receptor signaling and lytic function of activated NK cells. J Immunol 175: 4383–4391 [DOI] [PubMed] [Google Scholar]
  149. Rodriguez‐Ruiz ME, Vitale I, Harrington KJ, Melero I, Galluzzi L (2020) Immunological impact of cell death signaling driven by radiation on the tumor microenvironment. Nat Immunol 21: 120–134 [DOI] [PubMed] [Google Scholar]
  150. Rybstein MD, Bravo‐San Pedro JM, Kroemer G, Galluzzi L (2018) The autophagic network and cancer. Nat Cell Biol 20: 243–251 [DOI] [PubMed] [Google Scholar]
  151. Sáez JC, Contreras‐Duarte S, Gómez GI, Labra VC, Santibañez CA, Gajardo‐Gómez R, Avendaño BC, Díaz EF, Montero TD, Velarde V et al (2018) Connexin 43 hemichannel activity promoted by pro‐inflammatory cytokines and high glucose alters endothelial cell function. Front Immunol 9: 1899 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Saez PJ, Vargas P, Shoji KF, Harcha PA, Lennon‐Dumenil AM, Saez JC (2017) ATP promotes the fast migration of dendritic cells through the activity of pannexin 1 channels and P2X7 receptors. Sci Signal 10: eaah7107 [DOI] [PubMed] [Google Scholar]
  153. Saleh A, Simone B, Palazzo J, Savage JE, Sano Y, Dan Tu, Jin L, Champ C, Zhao S, Lim M et al (2013) Caloric restriction augments radiation efficacy in breast cancer. Cell Cycle 12: 1955–1963 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Schuler PJ, Saze Z, Hong CS, Muller L, Gillespie DG, Cheng D, Harasymczuk M, Mandapathil M, Lang S, Jackson EK et al (2014) Human CD4+ CD39+ regulatory T cells produce adenosine upon co‐expression of surface CD73 or contact with CD73+ exosomes or CD73+ cells. Clin Exp Immunol 177: 531–543 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Schumacher D, Strilic B, Sivaraj KK, Wettschureck N, Offermanns S (2013) Platelet‐derived nucleotides promote tumor‐cell transendothelial migration and metastasis via P2Y2 receptor. Cancer Cell 24: 130–137 [DOI] [PubMed] [Google Scholar]
  156. Shi L, Feng M, Du S, Wei X, Song H, Yixin X, Song J, Wenxian G (2019) Adenosine generated by regulatory T cells induces CD8(+) T cell exhaustion in gastric cancer through A2aR pathway. Biomed Res Int 2019: 4093214 [DOI] [PMC free article] [PubMed] [Google Scholar]
  157. Sidders B, Zhang P, Goodwin K, O'Connor G, Russell DL, Borodovsky A, Armenia J, McEwen R, Linghu B, Bendell JC et al (2020) Adenosine signaling is prognostic for cancer outcome and has predictive utility for immunotherapeutic response. Clin Cancer Res 26: 2176–2187 [DOI] [PubMed] [Google Scholar]
  158. Simone BA, Dan Tu, Palagani A, Jin L, Han SY, Wright C, Savage JE, Gitman R, Lim MK, Palazzo J et al (2016) Caloric restriction coupled with radiation decreases metastatic burden in triple negative breast cancer. Cell Cycle 15: 2265–2274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Sitkovsky MV, Hatfield S, Abbott R, Belikoff B, Lukashev D, Ohta A (2014) Hostile, hypoxia‐A2‐adenosinergic tumor biology as the next barrier to overcome for tumor immunologists. Cancer Immunol Res 2: 598–605 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Smyth LA, Ratnasothy K, Tsang JY, Boardman D, Warley A, Lechler R, Lombardi G (2013) CD73 expression on extracellular vesicles derived from CD4+ CD25+ Foxp3+ T cells contributes to their regulatory function. Eur J Immunol 43: 2430–2440 [DOI] [PubMed] [Google Scholar]
  161. Sprooten J, Agostinis P, Garg AD (2019) Type I interferons and dendritic cells in cancer immunotherapy. Int Rev Cell Mol Biol 348: 217–262 [DOI] [PubMed] [Google Scholar]
  162. Stagg J, Divisekera U, McLaughlin N, Sharkey J, Pommey S, Denoyer D, Dwyer KM, Smyth MJ (2010) Anti‐CD73 antibody therapy inhibits breast tumor growth and metastasis. Proc Natl Acad Sci USA 107: 1547–1552 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Stagg J, Divisekera U, Duret H, Sparwasser T, Teng MW, Darcy PK, Smyth MJ (2011) CD73‐deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res 71: 2892–2900 [DOI] [PubMed] [Google Scholar]
  164. Stagg J, Smyth MJ (2010) Extracellular adenosine triphosphate and adenosine in cancer. Oncogene 29: 5346–5358 [DOI] [PubMed] [Google Scholar]
  165. Starczynski J, Pepper C, Pratt G, Hooper L, Thomas A, Hoy T, Milligan D, Bentley P, Fegan C (2003) The P2X7 receptor gene polymorphism 1513 A–>C has no effect on clinical prognostic markers, in vitro sensitivity to fludarabine, Bcl‐2 family protein expression or survival in B‐cell chronic lymphocytic leukaemia. Br J Haematol 123: 66–71 [DOI] [PubMed] [Google Scholar]
  166. Stout CE, Costantin JL, Naus CC, Charles AC (2002) Intercellular calcium signaling in astrocytes via ATP release through connexin hemichannels. J Biol Chem 277: 10482–10488 [DOI] [PubMed] [Google Scholar]
  167. Sun X, Wu Y, Gao W, Enjyoji K, Csizmadia E, Müller CE, Murakami T, Robson SC (2010) CD39/ENTPD1 expression by CD4+Foxp3+ regulatory T cells promotes hepatic metastatic tumor growth in mice. Gastroenterology 139: 1030–1040 [DOI] [PMC free article] [PubMed] [Google Scholar]
  168. Synnestvedt K, Furuta GT, Comerford KM, Louis N, Karhausen J, Eltzschig HK, Hansen KR, Thompson LF, Colgan SP (2002) Ecto‐5'‐nucleotidase (CD73) regulation by hypoxia‐inducible factor‐1 mediates permeability changes in intestinal epithelia. J Clin Invest 110: 993–1002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Tahkola K, Ahtiainen M, Kellokumpu I, Mecklin JP, Laukkarinen J, Laakkonen J, Kenessey I, Jalkanen S, Salmi M, Bohm J (2021) Prognostic impact of CD73 expression and its relationship to PD‐L1 in patients with radically treated pancreatic cancer. Virchows Arch 478: 209–217 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Talmadge JE, Gabrilovich DI (2013) History of myeloid‐derived suppressor cells. Nat Rev Cancer 13: 739–752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Tang H, Sebti S, Titone R, Zhou Y, Isidoro C, Ross TS, Hibshoosh H, Xiao G, Packer M, Xie Y et al (2015) Decreased BECN1 mRNA expression in human breast cancer is associated with estrogen receptor‐negative subtypes and poor prognosis. EBioMedicine 2: 255–263 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Tappe KA, Budida R, Stankov MV, Frenz T, R Shah H, Volz A, Sutter G, Kalinke U, Behrens GMN (2018) Immunogenic cell death of dendritic cells following modified vaccinia virus Ankara infection enhances CD8(+) T cell proliferation. Eur J Immunol 48: 2042–2054 [DOI] [PubMed] [Google Scholar]
  173. Tatsuno K, Yamazaki T, Hanlon D, Han P, Robinson E, Sobolev O, Yurter A, Rivera‐Molina F, Arshad N, Edelson RL et al (2019) Extracorporeal photochemotherapy induces bona fide immunogenic cell death. Cell Death Dis 10: 578 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Thunberg U, Tobin G, Johnson A, Söderberg O, Padyukov L, Hultdin M, Klareskog L, Enblad G, Sundström C, Roos G et al (2002) Polymorphism in the P2X7 receptor gene and survival in chronic lymphocytic leukaemia. Lancet 360: 1935–1939 [DOI] [PubMed] [Google Scholar]
  175. Timp W, Feinberg AP (2013) Cancer as a dysregulated epigenome allowing cellular growth advantage at the expense of the host. Nat Rev Cancer 13: 497–510 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Togashi Y, Shitara K, Nishikawa H (2019) Regulatory T cells in cancer immunosuppression ‐ implications for anticancer therapy. Nat Rev Clin Oncol 16: 356–371 [DOI] [PubMed] [Google Scholar]
  177. Tokunaga A, Tsukimoto M, Harada H, Moriyama Y, Kojima S (2010) Involvement of SLC17A9‐dependent vesicular exocytosis in the mechanism of ATP release during T cell activation. J Biol Chem 285: 17406–17416 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Tokunaga R, Cao S, Naseem M, Lo JH, Battaglin F, Puccini A, Berger MD, Soni S, Millstein J, Zhang W et al (2019) Prognostic Effect of adenosine‐related genetic variants in metastatic colorectal cancer treated with bevacizumab‐based chemotherapy. Clin Colorectal Cancer 18: e8–e19 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Torres‐Pineda DB, Mora‐García MdL, García‐Rocha R, Hernández‐Montes J, Weiss‐Steider B, Montesinos‐Montesinos JJ, Don‐López CA, Marín‐Aquino LA, Muñóz‐Godínez R, Ibarra LRÁ et al (2020) Adenosine augments the production of IL‐10 in cervical cancer cells through interaction with the A(2B) adenosine receptor, resulting in protection against the activity of cytotoxic T cells. Cytokine 130: 155082 [DOI] [PubMed] [Google Scholar]
  180. Tripathi A, Lin E, Xie W, Flaifel A, Steinharter JA, Stern Gatof EN, Bouchard G, Fleischer JH, Martinez‐Chanza N, Gray C et al (2020) Prognostic significance and immune correlates of CD73 expression in renal cell carcinoma. J Immunother Cancer 8: e001467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  181. Tsukui H, Horie H, Koinuma K, Ohzawa H, Sakuma Y, Hosoya Y, Yamaguchi H, Yoshimura K, Lefor AK, Sata N et al (2020) CD73 blockade enhances the local and abscopal effects of radiotherapy in a murine rectal cancer model. BMC Cancer 20: 411 [DOI] [PMC free article] [PubMed] [Google Scholar]
  182. Turcotte M, Spring K, Pommey S, Chouinard G, Cousineau I, George J, Chen GM, Gendoo DMA, Haibe‐Kains B, Karn T et al (2015) CD73 is associated with poor prognosis in high‐grade serous ovarian cancer. Cancer Res 75: 4494–4503 [DOI] [PubMed] [Google Scholar]
  183. Turiello R, Capone M, Giannarelli D, Morretta E, Monti MC, Madonna G, Mallardo D, Festino L, Azzaro R, Levesque MP et al (2020) Serum CD73 is a prognostic factor in patients with metastatic melanoma and is associated with response to anti‐PD‐1 therapy. J Immunother Cancer 8: e001689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  184. Vesely MD, Schreiber RD (2013) Cancer immunoediting: antigens, mechanisms, and implications to cancer immunotherapy. Ann N Y Acad Sci 1284: 1–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  185. Vitale I, Shema E, Loi S, Galluzzi L (2021) Intratumoral heterogeneity in cancer progression and response to immunotherapy. Nat Med 27: 212–224 [DOI] [PubMed] [Google Scholar]
  186. Waickman AT, Alme A, Senaldi L, Zarek PE, Horton M, Powell JD (2012) Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol Immunother 61: 917–926 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Wang L, Fan J, Thompson LF, Zhang Y, Shin T, Curiel TJ, Zhang B (2011) CD73 has distinct roles in nonhematopoietic and hematopoietic cells to promote tumor growth in mice. J Clin Invest 121: 2371–2382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  188. Wang M, Jing J, Li H, Liu J, Yuan Y, Sun L (2021) The expression characteristics and prognostic roles of autophagy‐related genes in gastric cancer. PeerJ 9: e10814 [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Wang X, Zhang T, Song Z, Li L, Zhang X, Liu J, Liu X, Qiu L, Qian Z, Zhou S et al (2019) Tumor CD73/A2aR adenosine immunosuppressive axis and tumor‐infiltrating lymphocytes in diffuse large B‐cell lymphoma: correlations with clinicopathological characteristics and clinical outcome. Int J Cancer 145: 1414–1422 [DOI] [PubMed] [Google Scholar]
  190. Wang Y, Xie W, Humeau J, Chen G, Liu P, Pol J, Zhang Z, Kepp O, Kroemer G (2020) Autophagy induction by thiostrepton improves the efficacy of immunogenic chemotherapy. J Immunother Cancer 8: e000462 [DOI] [PMC free article] [PubMed] [Google Scholar]
  191. Watson MJ, Vignali PDA, Mullett SJ, Overacre‐Delgoffe AE, Peralta RM, Grebinoski S, Menk AV, Rittenhouse NL, DePeaux K, Whetstone RD et al (2021) Metabolic support of tumour‐infiltrating regulatory T cells by lactic acid. Nature 591: 645–651 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Wennerberg E, Spada S, Rudqvist N‐P, Lhuillier C, Gruber S, Chen Q, Zhang F, Zhou XK, Gross SS, Formenti SC et al (2020) CD73 blockade promotes dendritic cell infiltration of irradiated tumors and tumor rejection. Cancer Immunol Res 8: 465–478 [DOI] [PMC free article] [PubMed] [Google Scholar]
  193. Wiley JS, Dao‐Ung LP, Gu BJ, Sluyter R, Shemon AN, Li C, Taper J, Gallo J, Manoharan A (2002) A loss‐of‐function polymorphic mutation in the cytolytic P2X7 receptor gene and chronic lymphocytic leukaemia: a molecular study. Lancet 359: 1114–1119 [DOI] [PubMed] [Google Scholar]
  194. Willingham SB, Ho PY, Hotson A, Hill C, Piccione EC, Hsieh J, Liu L, Buggy JJ, McCaffery I, Miller RA (2018) A2AR antagonism with CPI‐444 induces antitumor responses and augments efficacy to anti‐PD‐(L)1 and anti‐CTLA‐4 in preclinical models. Cancer Immunol Res 6: 1136–1149 [DOI] [PubMed] [Google Scholar]
  195. Wu J, Wang YC, Xu WH, Luo WJ, Wan FN, Zhang HL, Ye DW, Qu YY, Zhu YP (2020a) High expression of CD39 is associated with poor prognosis and immune infiltrates in clear cell renal cell carcinoma. Onco Targets Ther 13: 10453–10464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Wu Qi, Tian A‐L, Durand S, Aprahamian F, Nirmalathasan N, Xie W, Liu P, Zhao L, Zhang S, Pan H et al (2020b) Isobacachalcone induces autophagy and improves the outcome of immunogenic chemotherapy. Cell Death Dis 11: 1015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  197. Xiao Z, Dai Z, Locasale JW (2019) Metabolic landscape of the tumor microenvironment at single cell resolution. Nat Commun 10: 3763 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Xu S, Shao Q‐Q, Sun J‐T, Yang N, Xie Qi, Wang D‐H, Huang Q‐B, Huang B, Wang X‐Y, Li X‐G et al (2013) Synergy between the ectoenzymes CD39 and CD73 contributes to adenosinergic immunosuppression in human malignant gliomas. Neuro Oncol 15: 1160–1172 [DOI] [PMC free article] [PubMed] [Google Scholar]
  199. Yamazaki T, Kirchmair A, Sato Ai, Buqué A, Rybstein M, Petroni G, Bloy N, Finotello F, Stafford L, Navarro Manzano E et al (2020) Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat Immunol 21: 1160–1171 [DOI] [PubMed] [Google Scholar]
  200. Yan A, Joachims ML, Thompson LF, Miller AD, Canoll PD, Bynoe MS (2019) CD73 promotes glioblastoma pathogenesis and enhances its chemoresistance via A(2B) adenosine receptor signaling. J Neurosci 39: 4387–4402 [DOI] [PMC free article] [PubMed] [Google Scholar]
  201. Yan J, Li X‐Y, Roman Aguilera A, Xiao C, Jacoberger‐Foissac C, Nowlan B, Robson SC, Beers C, Moesta AK, Geetha N et al (2020) Control of metastases via myeloid CD39 and NK cell effector function. Cancer Immunol Res 8: 356–367 [DOI] [PubMed] [Google Scholar]
  202. Yang H, Geng YH, Wang P, Yang H, Zhou YT, Zhang HQ, He HY, Fang WG, Tian XX (2020a) Extracellular ATP promotes breast cancer invasion and chemoresistance via SOX9 signaling. Oncogene 39: 5795–5810 [DOI] [PubMed] [Google Scholar]
  203. Yang R, Elsaadi S, Misund K, Abdollahi P, Vandsemb EN, Moen SH, Kusnierczyk A, Slupphaug G, Standal T, Waage A et al (2020b) Conversion of ATP to adenosine by CD39 and CD73 in multiple myeloma can be successfully targeted together with adenosine receptor A2A blockade. J Immuno Ther Cancer 8: e000610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  204. Yang X, Lu Y, Hang J, Zhang J, Zhang T, Huo Y, Liu J, Lai S, Luo D, Wang L et al (2020c) Lactate‐modulated immunosuppression of myeloid‐derived suppressor cells contributes to the radioresistance of pancreatic cancer. Cancer Immunol Res 8: 1440–1451 [DOI] [PubMed] [Google Scholar]
  205. Yang Y, Delalio LJ, Best AK, Macal E, Milstein J, Donnelly I, Miller AM, McBride M, Shu X, Koval M et al (2020d) Endothelial Pannexin 1 channels control inflammation by regulating intracellular calcium. J Immunol 204: 2995–3007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  206. Yeaman MR (2014) Platelets: at the nexus of antimicrobial defence. Nat Rev Microbiol 12: 426–437 [DOI] [PubMed] [Google Scholar]
  207. Yegutkin GG, Marttila‐Ichihara F, Karikoski M, Niemelä J, Laurila JP, Elima K, Jalkanen S, Salmi M (2011) Altered purinergic signaling in CD73‐deficient mice inhibits tumor progression. Eur J Immunol 41: 1231–1241 [DOI] [PubMed] [Google Scholar]
  208. Young A, Ngiow S, Barkauskas D, Sult E, Hay C, Blake S, Huang Q, Liu J, Takeda K, Teng M et al (2016) Co‐inhibition of CD73 and A2AR adenosine signaling improves anti‐tumor immune responses. Cancer Cell 30: 391–403 [DOI] [PubMed] [Google Scholar]
  209. Young A, Ngiow SF, Gao Y, Patch A‐M, Barkauskas DS, Messaoudene M, Lin G, Coudert JD, Stannard KA, Zitvogel L et al (2018) A2AR adenosine signaling suppresses natural killer cell maturation in the tumor microenvironment. Cancer Res 78: 1003–1016 [DOI] [PubMed] [Google Scholar]
  210. Yu M, Guo G, Huang L, Deng L, Chang C‐S, Achyut BR, Canning M, Xu N, Arbab AS, Bollag RJ et al (2020) CD73 on cancer‐associated fibroblasts enhanced by the A2B‐mediated feedforward circuit enforces an immune checkpoint. Nat Commun 11: 515 [DOI] [PMC free article] [PubMed] [Google Scholar]
  211. Zhang LY, Ibbotson RE, Orchard JA, Gardiner AC, Seear RV, Chase AJ, Oscier DG, Cross NC (2003) P2X7 polymorphism and chronic lymphocytic leukaemia: lack of correlation with incidence, survival and abnormalities of chromosome 12. Leukemia 17: 2097–2100 [DOI] [PubMed] [Google Scholar]
  212. Zhou P, Zhi X, Zhou T, Chen S, Li X, Wang L, Yin L, Shao Z, Ou Z (2007) Overexpression of Ecto‐5'‐nucleotidase (CD73) promotes T‐47D human breast cancer cells invasion and adhesion to extracellular matrix. Cancer Biol Ther 6: 426–431 [DOI] [PubMed] [Google Scholar]

Articles from The EMBO Journal are provided here courtesy of Nature Publishing Group

RESOURCES