Next Article in Journal
OMOP CDM Can Facilitate Data-Driven Studies for Cancer Prediction: A Systematic Review
Next Article in Special Issue
Purinergic Signalling in the Cochlea
Previous Article in Journal
SERPINE1 DNA Methylation Levels Quantified in Blood Cells at Five Years of Age Are Associated with Adiposity and Plasma PAI-1 Levels at Five Years of Age
Previous Article in Special Issue
Alternatively Spliced Isoforms of the P2X7 Receptor: Structure, Function and Disease Associations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Adenosine, Schizophrenia and Cancer: Does the Purinergic System Offer a Pathway to Treatment?

1
Department of Neurosciences, University of Toledo, Toledo, OH 43614, USA
2
Neurosciences Institute, ProMedica, Toledo, OH 43606, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(19), 11835; https://doi.org/10.3390/ijms231911835
Submission received: 9 August 2022 / Revised: 23 September 2022 / Accepted: 29 September 2022 / Published: 5 October 2022
(This article belongs to the Special Issue Purinergic Signalling in Physiology and Pathophysiology)

Abstract

:
For over a century, a complex relationship between schizophrenia diagnosis and development of many cancers has been observed. Findings from epidemiological studies are mixed, with reports of increased, reduced, or no difference in cancer incidence in schizophrenia patients. However, as risk factors for cancer, including elevated smoking rates and substance abuse, are commonly associated with this patient population, it is surprising that cancer incidence is not higher. Various factors may account for the proposed reduction in cancer incidence rates including pathophysiological changes associated with disease. Perturbations of the adenosine system are hypothesized to contribute to the neurobiology of schizophrenia. Conversely, hyperfunction of the adenosine system is found in the tumor microenvironment in cancer and targeting the adenosine system therapeutically is a promising area of research in this disease. We outline the current biochemical and pharmacological evidence for hypofunction of the adenosine system in schizophrenia, and the role of increased adenosine metabolism in the tumor microenvironment. In the context of the relatively limited literature on this patient population, we discuss whether hypofunction of this system in schizophrenia, may counteract the immunosuppressive role of adenosine in the tumor microenvironment. We also highlight the importance of studies examining the adenosine system in this subset of patients for the potential insight they may offer into these complex disorders.

1. Introduction

Epidemiological observations dating back over one hundred years suggest that incidence rates of some cancers may be lower in patients with schizophrenia [1,2,3,4,5]. Numerous studies have been conducted in the decades since then but have failed to reach a consensus; there are reports of increased, reduced, or no difference in incidence rates of most cancers in schizophrenia patients compared to the general population (see reviews [2,3,4,5] and epidemiology studies [5,6,7,8,9,10,11]). However, as many of the risk factors for cancer, including smoking, alcohol use, increased body weight, physical inactivity, and reduced access to medical care, are also found in schizophrenia populations, it is surprising that cancer incidence rates are not higher amongst these patients [4,6,12,13,14,15,16,17]. As has previously been observed, this unexpected finding lends support to the idea that a diagnosis of schizophrenia may be protective against development of (some) cancers [18].

2. Cancer Incidence in Schizophrenia

A limitation of many epidemiological studies of cancer incidence in schizophrenia is that they do not control for cancer-related confounding variables. Age, for example, is a major confounding variable, as patients with schizophrenia typically have a shorter lifespan [19], making comparisons of cancer incidence with control populations a challenge. It is also crucial to precisely define control groups (general population, patients in inpatient care or outpatient care, non-schizophrenia, non-psychiatric, etc.) and to match study subjects based on age and sex. However, when risk factors like age and smoking are considered in analyses, the reported cancer incidence rates are lower than would be expected in this patient population [6,7]. It is also important to note that cancer incidence rates, and not cancer mortality rates, are proposed to be lower for some cancers in schizophrenia patients. Survival rates of schizophrenia patients who develop cancer are typically lower than the general public [20,21]. This is especially interesting when considering separate biological mechanisms regulate cancer initiation, proliferation and metastasis [22]. In Table 1 we summarize the results of recent epidemiology meta-analyses reporting cancer rates in schizophrenia patients. Overall, a limited number of prospective cohort studies were available for inclusion in these meta-analyses, missing medication history and detailed demographic information is common, variable data collection methods and the use of inconsistently defined control groups contribute to challenges in interpreting the data and determining whether cancer incidence rates are significantly reduced in schizophrenia patients [3,23]. Interestingly, breast cancer incidence rates are consistently higher in studies of schizophrenia populations (Table 1). Recent genome wide association studies (GWAS) have identified a shared risk locus associated with schizophrenia and breast cancer [24] and increased risk for breast, ovarian and thyroid cancers, but not other cancers, in schizophrenia patients [25]. Conversely, lung, liver and prostate cancer incidence appears to be lower in schizophrenia populations (Table 1), suggesting that incidence rates of some cancers may be reduced, or unchanged, in these patients. Additional appropriately powered studies that consider different cancer types and account for potentially confounding variables (age, BMI, smoking, medications, genetics, and geography) will be required to ascertain whether a diagnosis of schizophrenia protects against the development of some cancers.
Chronic antipsychotic use, genetic factors, and pathophysiological changes have been posited to explain the potentially reduced risk of cancer development in schizophrenia patients. For example, there is a growing body of evidence that antipsychotics may protect against the development of cancer; antipsychotics are now being repurposed as potential cancer therapies, with anticancer mechanisms of action in peripheral tissues still poorly understood [6,28,29,30,31]. Epidemiological studies have found that unaffected family members of patients with schizophrenia have reduced rates of cancer [4,32], suggesting that genetic factors contributing to schizophrenia risk may also reduce the risk of developing cancer. However, much less is known about how pathophysiological changes associated with schizophrenia may contribute to the reduced risk of cancer development in some patients [33,34].
The potential protective effects of the adenosine system against cancer development in schizophrenia patients was first suggested over 20 years ago in the “Purinergic Hypothesis of Schizophrenia” by Lara and Souza [35,36]. The authors noted the reduced rates of cancer in schizophrenia patients, and proposed a role for the purinergic system in this phenomenon, although little was known about the function of the adenosine system in cancer at that time [37,38]. Today, the purinergic hypothesis has evolved, with a growing body of evidence suggesting that reduced availability of extracellular adenosine, and the resulting effects on neuromodulation, and immune system and bioenergetic regulation, may contribute to the onset of schizophrenia [39]. Conversely, increased adenosine metabolism is now recognized as a characteristic of many types of cancer. Elevated adenosine, and its metabolic precursor adenosine triphosphate (ATP), promote cancer growth and play a role in host immunosuppression in the tumor microenvironment (TME) [36,40,41,42,43].
Herein, we discuss the biochemical and pharmacological evidence for hypofunction of the adenosine system in schizophrenia and hyperfunction in cancer (Figure 1). We draw primarily from postmortem and clinical studies to explore whether perturbation of the adenosine system may contribute to reduced risk for cancer development in some schizophrenia patients. We also review the literature showing that augmentation or attenuation of the adenosine system can have therapeutic benefits for these disorders.

3. The Adenosine System: Extracellular Adenosine Generating Pathways

The ratio of ATP:adenosine is tightly regulated by enzymatic pathways that modulate the generation of extracellular adenosine (Figure 2). Adenosine can be directly released from the cell by nucleoside transporters [44] where it is rapidly catabolized to inosine by the purine salvage pathway enzyme adenosine deaminase (ADA) [45]. Inosine is further metabolized via a series of enzymes to the end-product of adenosine metabolism, uric acid. Alternatively, adenosine can be generated from the sequential breakdown of ATP [46]. ATP can be released from the cell into the extracellular space via several different mechanisms including vesicular release, lysosomes, cell lysis, and nucleotide-permeable channels [47,48,49,50,51]. ATP is then rapidly catabolized to adenosine by the extracellular enzymes ectonucleoside triphosphate diphosphohydrolases (ENTPD 1-3, 8; Table 2) [52] which hydrolyze ATP to ADP/AMP. Ecto-5′nucleotidase (NT5E; Table 2) is the primary rate-limiting enzyme that converts AMP to adenosine [53,54,55]. During pathophysiological states, ATP and adenosine are released into the extracellular space to fine-tune the immune system [56]. ENTPD1 and NT5E play significant roles in the regulation of immunity and inflammation [57,58]. This enzyme cascade modulates purinergic signaling by driving a shift from a pro-inflammatory ATP state to an adenosine-induced anti-inflammatory environment, a role that is facilitated by their widespread expression on almost all tissue cell types [59]. Thus, adenosine and ATP levels are closely interrelated and the ratio of adenosine to ATP acts as a significant modulator of their biological effects.

4. The Adenosine System: Purinergic Receptors

Adenosine and ATP activate two main families of receptors; the purinergic P1 and P2 receptors; respectively (see Supplementary Table 1 for receptor nomenclature; ligands; G-protein; and ions) [60,61,62,63,64,65,66,67,68]. The G-protein coupled P1 adenosine receptors are inhibitory (A1 and A3) and excitatory (A2AR and A2BR); with high affinity (A1R and A2AR) (0.1–0.3 μM) and low affinity (A2BR; A3R) (15–25 μM) for adenosine [36,69,70], although some studies using functional assays report that adenosine is approximately equipotent at its receptors [71,72,73]. Purinergic receptor activation is a tightly regulated process. A1Rs are typically tonically activated by endogenous extracellular adenosine whereas A2ARs are selectively recruited by adenosine produced through extracellular catabolism of ATP by ectonucleotidases [74,75,76,77].
Due to the widespread constitutive expression of purinergic receptors in the body and the ability of P1Rs to form homo- and heterodimers with other neurotransmitter receptor types [68], adenosine and ATP serve significant roles in modulating neural processes, immune response, energy metabolism, and sleep [69,70,71]. Presynaptic A1 receptor activation inhibits excitatory synaptic transmission by reducing calcium influx and glutamate release, while post-synaptic A1 receptor activation reduces ionotropic glutamate receptor and voltage-sensitive calcium channel activation [78] leading to a decrease in excitatory synapse activity. Conversely, the actions of facilitatory A2ARs increase the release of glutamate and ionotropic glutamate receptor function in different brain regions. Their expression on astrocytes regulates glutamate uptake and Na+/K+ ATPase, contributing to the role of A2ARs as selective mediators of synaptic plasticity [78]. Increasing the complexity of adenosine’s neuromodulatory action [79,80], adenosine receptors can also form heterodimers with other G-protein coupled receptors [81]. Activation of antagonistic A2A-D2 receptor heteromers, first identified in striatal membrane preparations [82] and found in GABAergic striatopallidal neurons [83], results in reduced D2 receptor affinity for agonists. A2A-D2 receptors expressed on striatal astrocytes also functionally interact to modulate glutamate gliotransmitter release [84]. Inhibitory A1 receptors dimerize with excitatory dopamine D1 receptors to form receptor heteromers that modulate striatonigral neuronal function [83]. Adenosine’s ability to modulate glutamatergic and dopaminergic pathways is of relevance in the pathophysiology of schizophrenia.
ATP and adenosine also regulate immune responses via activation of purinergic receptors, leading to immune cell activation, signal amplification, chemotaxis, and phagocytosis [56,85,86,87]. ATP acts as a pro-inflammatory danger associated molecular pattern (DAMP) when it is increased in extracellular fluids, usually indicating inflammation, ischemia, or injury [36,88]. In contrast, adenosine displays potent anti-inflammatory effects [89,90,91]. Modulation of purinergic receptors and ectonucleotidases allows for complex feedback regulation. During periods of injury or stress, cells respond by increasing ATP release into the extracellular space, inducing a pro-inflammatory response upon binding to P2 receptors. Most immune cells express several types of P2XRs and P2YRs [92]. Injured or stressed cells release ATP to recruit immune cells for phagocytosis and clearance. Immune cells can also release ATP to amplify cellular activation and regulate chemotaxis [93]. Prolonged states of inflammation can be toxic to tissues and cells. Thus, to prevent excessive inflammation and tissue damage, ATP is hydrolyzed by ectonucleotidases to generate adenosine. The decreased extracellular ratio of ATP:adenosine reduces P2 signaling and increases P1 signaling, specifically the upregulation and activation of A2ARs and A2BRs. Engagement of A2ARs allows for the inhibition of lymphocyte activation [94] while activation of A2BR increases the production of vascular endothelial growth factor (VEGF) and interleukin-6 (IL-6) [95]. A2BRs, which are mainly expressed on macrophages, dendritic cells, and mast cells, may then contribute to the resolution of inflammation by promoting healing [96]. The endogenous anti-inflammatory role of adenosine is utilized in therapies for acute and chronic inflammatory diseases like chronic obstructive pulmonary disease and rheumatoid arthritis [97,98].

5. The Adenosine System: Perturbed Adenosine Metabolism in Disease: Uric Acid

The final product of adenosine metabolism, uric acid, is used clinically to assess purine catabolism. Altered levels of uric acid in the blood of schizophrenia patients implicates impairment of the adenosine system in the pathophysiology of this disorder. Reduced plasma uric acid levels are reported in clinically stable and relapsed schizophrenia patients [99] and in first-episode neuroleptic-naïve schizophrenia patients [100] compared to healthy controls. Although schizophrenia patients are not hypouricemic (generally defined as <2 mg/dL), they report lower uric acid plasma levels (schizophrenia male—5.1 mg/dL, schizophrenia female—4.4 mg/dL; healthy control male—6 mg/dL, healthy control female—5.1 mg/dL), suggesting perturbed purinergic signaling [100]. In contrast, higher levels of uric acid are reported during the acute phase in schizophrenia patients [101]. Similarly, during psychosis relapse, elevated uric acid levels have been found in schizophrenia patients that were as high as in bipolar disorder patients during manic episodes and higher than in the healthy control group [102]. These inconsistent findings of uric acid levels may be attributed to the relatively small study sizes, differences in uric acid levels at different stages of this heterogeneous disorder, or the effects of antipsychotic medications. A recent meta-analysis of 17 studies addressed these limitations, and in subgroup analyses He et al., found that uric acid levels are significantly reduced in first episode psychosis (Weighed mean difference −40.61 μmol/L, CI95% −59.47–−21.76, p < 0.0001) in schizophrenia patients but not in those with chronic schizophrenia (Weighted mean difference = −3.09 μmol/L, CI95% −35.21–29.04, p = 0.85) [103]. They also reported uric acid levels are reduced in male (Weighed mean difference = −34.83 μmol/L, CI95% −54.97–−14.69, p = 0.0007) and American schizophrenia patients (Weighed mean difference = −39.90 μmol/L, CI95% −61.47–−18.33, p = 0.0003). These findings suggest disease-stage and sex-dependent differences in uric acid levels indicating an alteration in purine catabolism early in the course of the disease. Hypofunction of the adenosine system in schizophrenia lends one possible explanation for these decreased uric acid levels in patients, as a lowered availability of adenosine restricts uric acid production.
Uric acid also plays a dual role in the body’s antioxidant defense system [104]. Low levels of uric acid decrease the body’s ability to prevent free radical damage, leading to cellular damage and death [105]. Conversely, high levels of uric acid (hyperuricemia, generally defined as >6 mg/dL) contribute to an inflammatory response, as seen in disorders such as gout and metabolic syndrome [106,107]. Uric acid acts as a powerful antioxidant to neutralize free radicals, such as peroxynitrite and hydroxyl, making its levels in the blood a strong indicator of oxidative stress. There is robust evidence supporting an increased oxidative stress status in schizophrenia [108,109], while the use of antioxidant supplementation, like vitamin E and C, improves some of the psychopathological symptoms of schizophrenia [110,111,112]. Thus, as well as reflecting changes in adenosine metabolism, baseline levels of purine metabolites are a good predictor of clinical and neurological symptoms in schizophrenia [113].
Hyperuricemia is associated with cancer incidence and mortality [114,115]. Elevated serum uric acid (6.8 mg/dL or 404 uM without the presence of gout) is positively correlated with risk for gastrointestinal cancers and an increased overall mortality risk for cancers [116]. High serum uric acid (>6 mg/dL) is used as a prognostic predictor of colorectal cancer, pancreatic cancer, large B-cell lymphoma, and esophageal squamous cell carcinoma [117,118,119,120]. A study examining hyperuricemia in cancer development showed spontaneous hepatocellular carcinoma and hepatomegaly in mice following knockout of the enzyme, Urahplt2/plt2, responsible for uric acid metabolism [121]. These findings point to altered adenosine catabolism as contributing to cancer incidence and mortality. Adenosine levels are increased in the TME but not in plasma of patients with cancer [122,123]. Equally, expression and activity of the enzyme ADA, which metabolizes adenosine to inosine, is also increased in tumors [124], but similar increases in serum may originate from other sources [125]. Increased uric acid levels likely reflect the increased abundance of adenosine in the TME and are an indicator of oxidative stress in response to cancer.
We posit that the adenosine system is hypofunctional in patients with schizophrenia resulting in reduced uric acid generation, in line with the reports of lower peripheral uric acid levels, which potentially mitigates the increased risk for cancer associated with hyperuricemia. Hypo- and hyperuricemia are associated with disease and although animal models indicate a potential causative role for hyperuricemia in hepatic cancers [121], the etiology of dysregulated uric acid levels in these diseases is not yet known. Rather, uric acid acts as a marker of purine catabolism, suggesting significant perturbation of the adenosine system in both schizophrenia and cancer.

6. Schizophrenia: Evidence for Adenosine System Perturbation

Schizophrenia is a severe, debilitating mental illness characterized by paranoid delusional thinking, hallucinations and alterations in volition, neurocognition, and affective regulation. While the global incidence of schizophrenia is relatively small, estimated at 0.30–0.66%, there is a substantial burden to the individual afflicted with the disease and to society [126,127]. One evolving theory describing the etiology of schizophrenia is dysfunction of the adenosine system. As a neuromodulator of both the glutamate and dopamine neurotransmitter systems, perturbation of adenosine may reconcile the hyperdopaminergic and hypoglutamatergic states that underlie the positive, negative, and cognitive symptoms found in patients with schizophrenia. This hypofunction of adenosine hypothesis developed from observation of several phenomena: (1) presynaptic activation of adenosine A1 receptors inhibits the release of glutamate, (2) postsynaptic activation reduces NMDA-receptor functioning, and (3) adenosine signaling through postsynaptic A2A receptors inhibits D2 receptor signaling through a heteromeric complex, as reviewed [128]. Both dysfunction of the glutamate and the dopamine systems have been attributed to the etiology of schizophrenia.
The enzymes responsible for the generation and regulation of extracellular adenosine are significantly altered in the brain in schizophrenia. Protein expression of the equilibrative nucleoside transporter (ENT1), which transports adenosine, is reduced in SCZ in the superior temporal gyrus [129]. Protein expression and enzyme activity of ENTPD1 and NT5E are significantly reduced in the striatum in schizophrenia subjects [130]. ENPTD1 and ENTPD2 transcript levels were also significantly reduced in enriched populations of astrocytes, but not neurons, in the dorsolateral prefrontal cortex (DLPFC) in schizophrenia [131]. Conversely, transcript levels of adenosine deaminase (ADA), the enzyme responsible for metabolizing adenosine to inosine, were significantly increased in an enriched population of pyramidal neurons in the same study, suggesting region- and cell-subtype specific dysregulation in adenosine metabolism. Although it has been proposed that increased expression of the enzyme adenosine kinase may drive adenosine hypofunction [39,132], recent findings show no significant changes in adenosine kinase expression in the brain in schizophrenia [133]. This suggests that hypofunction of ectonucleotidase and ADA pathways are likely responsible for the altered availability of extracellular adenosine.
Purinergic receptors are also significantly dysregulated in schizophrenia. Polymorphisms in the A1 receptor, but not the A2A receptor, are associated with schizophrenia [128]. In the striatum, a reduction in the transcript and the protein expression of the A2A receptor, attributed to hypermethylation of the A2A coding region, was found in schizophrenia [134]. Increases in striatal A2A receptor density [135] and caudate nucleus A2A and D2 receptor protein expression [136] are reported in schizophrenia patients. Conversely, A2A-D2 receptor heterodimers were significantly reduced (almost 60%) in the same subjects [136]. These results lend support to the adenosine hypothesis of schizophrenia and deficits in the neuromodulatory action of adenosine on the dopamine system via the A2A-D2 receptor heteromers. No changes were found in A1 receptor expression in the striatum [133], but there was a reduction in the A1 receptor transcript in an enriched population of pyramidal neurons from the DLPFC in schizophrenia subjects [131]. Little is known about A2B and A3 receptor expression in the brain in schizophrenia, likely due to their low levels of expression in this tissue. As a modulator of the dopamine and glutamate neurotransmitter systems, dysfunction of the adenosine system is hypothesized to contribute to the dopaminergic hyperfunction and glutamatergic hypofunction that contributes to the onset of schizophrenia symptoms.

7. Schizophrenia: The Adenosine System as a Therapeutic Target

The clinical treatment for schizophrenia is mainly centered on antipsychotics, specifically dopamine D2 receptor antagonists. However, a number of small clinical studies demonstrate the utility of the adenosine modulators allopurinol, a xanthine oxidase inhibitor [137,138], and dipyramidole, an inhibitor of the adenosine transporter (ENT) [139]. Used as add-on therapies to antipsychotics, they improved both positive (hallucinations, delusions, mania) and negative symptoms (avolition, flat affect, asociality, alogia) of schizophrenia, as assessed by the Positive and Negative Syndrome Scale (PANSS) [140,141]. However, others report no differences between adjuvant allopurinol and placebo [142]. A recent study found that administering adenosine modulators as add-on therapies significantly reduced the rate of psychiatric rehospitalization and all-cause mortality among schizophrenia patients in a Finnish cohort, particularly in younger patients (<45 years) [143]. In animal models of schizophrenia-like behaviors, augmenting adenosine levels in mice that overexpress the enzyme ADK [37] and antagonism of the P2X7R in the sub-chronic phencyclidine-induced model of schizophrenia [144], significantly improved cognitive deficits. Further investigation of the adenosine system, including therapies that directly target purinergic receptors and ATP-adenosine metabolic pathway components, may offer new avenues for the treatment of schizophrenia [145].

8. Cancer: Extracellular Adenosine Generation and Cancer

The TME is a multicellular complex composed of various immune cells, cancer associated fibroblasts, endothelial cells and cancer stem cells [146]. The TME suppresses immune response and supports a complex network of signals between multiple cell types that work in concert to permit and sustain tumor growth and vascularization [147,148,149]. Accumulation of extracellular ATP is observed in the TME and under hypoxic, chronic and acute inflammation conditions [122,123]. Extracellular ATP levels are low (nanomolar range) in healthy tissue however upon cellular stress, damage or cell death, extracellular ATP can increase significantly (micromolar range) [123,150]. As ATP is released from cancer cells, it is metabolized to immunosuppressive AMP and adenosine, which act as key modulators of immune cells [36]. As in the brain, adenosine in the TME is primarily produced by extracellular metabolism of ATP via a cascade of ectonucleotidase enzymes (Figure 2) [151]. Studies examining ex vivo adenosine levels in rodent models found adenosine to be twice as high in tumors as compared to other tissues and at least 30% higher in the tumor core [123]. The increased presence of adenosine in the TME serves to regulate cancer cell growth and immune cell activity following adenosine receptor activation [123,152,153].
Enzymatic pathways that regulate the production of extracellular adenosine are also of importance in cancer development. The purine salvage pathway enzyme, ADA, modulates ATP:adenosine levels. ADA activity is increased in breast, kidney, and colorectal cancers as well as lymphocytes, a potential compensatory mechanism to offset elevated adenosine production [125,154,155,156]. Conversely, ADA activity was decreased in prostate and gastric tumors, as well as Hodgkin’s lymphoma [157,158,159]. This highlights the dynamic metabolomic demands of cancer tissues and perturbation of adenosine metabolic processes in cancer.
Increased production of extracellular adenosine promotes both cancer cell proliferation and suppression of immune cells in the TME. There is abundant evidence that ENTPD1 and NT5E expression are increased in cancer including in gastric cancer, non-small cell lung cancer and prostate cancer [160,161,162,163]. ENTPD1 is elevated in intratumoral immune cells of non-small cell lung cancer [164]. Adult T-Cell leukemia/lymphoma cells that express high levels of ENTPD1 effectively evade antitumor immunity [165]. Similar findings have been reported in human follicular lymphoma, glioblastoma multiforme, breast cancer, rectal adenocarcinoma, non-small cell lung cancer, head and neck cancer, and medulloblastoma [166,167,168,169,170,171,172]. In immunocompetent rats, a NT5E siRNA was used to assess NT5E contribution to immune cell evasion and tumor growth in glioblastoma. In this model, decreased T lymphocyte infiltration and tumor cell apoptosis was observed [173]. Similarly, knockdown of ENTPD1 (CD39) in cancer cells and in mice yielded lower ATP consumption and increased T-Cell infiltration in tumors [174]. Additionally, a meta-analysis of 13 studies analyzing the prognostic value of NT5E, indicates NT5E expression correlates with poor overall survival and disease-free survival in solid-tumor cancer patients [175]. ENTPD1 and NT5E, as important regulators of extracellular adenosine availability, facilitate evasion of the immune system by cancer cells. These findings illustrate widespread upregulation of ATP-adenosine levels and metabolism by ectonucleotidases, resulting in cancer cell proliferation and immunosuppression in cancer. Targeting elevated adenosine levels and ATP-adenosine metabolism, may increase immune response in cancer.
Extracellular adenosine binds to the A2A receptor expressed on macrophages, monocytes, dendritic cells, natural killer cells and T-Cells [41,123,176,177,178] and the A2B receptor on macrophages and dendritic cells [96,179]. This increases intracellular cAMP leading to cAMP/PKA signaling that suppresses T-cell response [180]. However, adenosine receptor activation has pleiotropic effects on other immune cells. It prevents dendritic cell activation but also increases the release of VEGF, a signaling protein that promotes angiogenesis thereby promoting tumor growth [96,181]. In tissues where adenosine concentrations are low, adenosine activation of A1Rs can enhance neutrophil activity [85]. Adenosine can also induce chemotaxis in immature dendritic cells via A1R activation [182], whereas mature dendritic cells are sensitive to adenosine-mediated inhibition through the A1 receptor [181]. Overall, adenosine receptor activation has an immunosuppressive effect on different mature (and immature) immune cells and aids the progression of cancer by facilitating immune-surveillance evasion.
All adenosine receptors subtypes seem to contribute to cancer cell proliferation via activation of different kinase signaling pathways including AKT, ERK1/2, p38, JNK and PKC-δ pathways [42,183,184,185,186,187,188,189]. Colon cancer cells, where A2A and A3 are the most abundantly expressed receptor subtypes, undergo increased cell proliferation when treated with high (micromolar) concentrations of adenosine [189]. The A2AR contributes to angiogenesis and wound healing indicating that the downstream targets of adenosine receptors, including ERK, JNK, p38, and AKT, contribute to tumor growth via multiple signaling pathways [190,191]. Bladder urothelial carcinoma tissues and cells express A2BR at higher levels than other adenosine receptor subtypes. High expression of A2BR is correlated with poor prognosis, and suppression of A2BR inhibited proliferation, invasion, and migration of bladder urothelial carcinoma cells with arrest at the G1 phase [188]. Oral squamous cell carcinoma cells (OSCC) are inhibited following knockdown of A2BR. Interestingly, the P2 purinergic receptor P2X7, which is expressed on tumor-associated immune cells, also contributes to immunosuppression of cancer in the tumor microenvironment [192,193,194,195]. Additionally, knockdown of the P2X7 receptor suppresses TGFβ-1 induced cell migration in A549 lung cancer cells and actin remodeling, illustrating that activation of P2X7 contributes to cell migration [196].
In summary, purinergic signaling is important to cancer cell progression and immune response through a variety of pathways. Elevated extracellular ATP acts on P2 receptors, which are abundantly expressed in many cancer subtypes and immune cells which contribute to increased cancer cell proliferation and immune evasion. ATP is metabolized to adenosine which acts on P1 receptors, whose immunosuppressive role similarly helps tumors evade immune cells.

9. Cancer: Adenosine System as a Therapeutic Target in Cancer

A number of different anti-cancer therapies that target the adenosine system are currently in development (Table 3). Small molecules (See drug formulas Table 3 and structures in Supplementary Figure 1) have been developed to target the adenosine receptors to improve cancer immune response and inhibit proliferation and metastasis of cancer cells. A2A receptor antagonists currently in clinical trials include compounds CPI-444, MK-3814, NIR178, and AZD4635 that are administered as monotherapies or in combination with PD-1 and PD-L1 antagonists [197]. A2AR antagonists like AZD-4635, CPI-444, and NIR-178 are under investigation for their ability to improve T-Cell infiltration in tumors. Lesser studied A2B and A3 receptor antagonists, like PBF-1129 and CF-102, respectively, are also undergoing clinical trials (Table 3, Figure 1).
A promising but relatively understudied strategy is targeting adenosine-generating enzymes. Due to their higher specificity and longer half-life, monoclonal antibody-directed chemotherapies are used to target the adenosine generating enzyme NT5E (see Table 3) [198,199]. Internalization of NT5E following treatment with monoclonal antibody therapies prevents adenosine-mediated action on tumors, as well as preventing metastasis [199]. Therapies targeting ENTPD1 are predominately still exploratory. By reducing adenosine availability in the extracellular space, ENTPD1-targeted therapies seek to prevent metastasis and improve cancer immune response by restoring T-reg function [200,201,202]. In breast cancer, reducing ENTPD1 activity and adenosine generation prevents A2B receptor activation, reducing metastasis-inducing transcription factor FRA1/FOSL1 expression [203].
Adenosine system targeting cancer therapies currently being investigated in clinical trials highlight the importance of elucidating the role of adenosine system dysfunction in the development of novel cancer therapeutics.

10. Summary and Conclusions

One of the most surprising findings from epidemiological studies of schizophrenia is the reportedly low incidence of cancer relative to the general population. Conflicting reports of cancer rates in schizophrenia have been published for decades, even though this population are considered at increased risk of developing cancer, due to the prevalence of risk factors like smoking and access to medical care [4]. Such controversial findings lend support for the idea that understanding the pathophysiology of schizophrenia may offer unique insight into the etiology of cancer.
Overall, there is a paucity of data available on cancer-related markers in schizophrenia patients who develop cancer. The available literature on this unique population has breadth but lacks depth, as the publicly available epidemiological data sets are limited to diagnoses and procedures (e.g., National Inpatient Sample). Several studies consider genetic risk variants that may be relevant to schizophrenia and cancer, but there is little data available from clinical laboratory tests reporting on biological markers for cancer in schizophrenia patients [204,205,206]. Thus, we consider reports from postmortem and clinical studies that assess the purinergic system in these disorders.
The adenosine system is hyperfunctional in cancer, facilitating tumor growth, survival and proliferation while evidence suggests this system is hypofunctional in schizophrenia. While it is challenging to determine if hypofunction of the adenosine system in the brain in schizophrenia extends systemically (and may impact cancer development throughout the body), corresponding changes in peripheral uric acid levels in a subset of the schizophrenia population suggest that this may be the case. Perturbation of ATP and adenosine metabolism, resulting in reduced availability of extracellular adenosine and dysregulated purinergic receptor activation, may attenuate the pathological oncogenic processes driven by elevated purine levels. Adenosine and ATP contribute to tumorigenesis through P1 and P2 receptor mediated signaling. Importantly, several P1 receptor antagonists are under investigation as therapies for different cancers. In schizophrenia, modulating adenosine catabolism, rather than directly targeting P1 receptors (due to associated side effects) is a moderately successful strategy at alleviating symptoms of schizophrenia. This approach is also used in cancer treatment, and drugs targeting ENTPD and NT5E are under exploration.
Much work is required to determine why and how a schizophrenia diagnosis appears to protect against the development of cancer. Schizophrenia and cancer are complex, heterogeneous disorders; multiple genetic and environmental factors interact leading to the onset of these disorders. Perturbation of neurobiological processes, like the adenosine system, may underlie the lower incidence of cancer reported in some schizophrenia patients, and lends support for targeting this system therapeutically in these disorders.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms231911835/s1.

Author Contributions

Conceptualization, A.-R.H. and S.M.O.; writing—original draft preparation, A.-R.H., K.B., O.K., N.H., X.W. and S.M.O.; writing—review and editing, A.-R.H., K.B., R.E.M. and S.M.O.; supervision, S.M.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by RO1 MH107487 and MH121102 (REM). This project was supported by Grant YIG-1-139-20 awarded to SMOD from the American Foundation for Suicide Prevention. The content is solely the responsibility of the authors and does not necessarily represent the official views of the American Foundation for Suicide Prevention.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. HMSO; The Board of Control of the Commissioners in Lunacy. Sixty-Fourth Report of the Commissioners in Lunacy to the Lord Chancellor; His Majety’s Stationery Office: London, UK, 1910.
  2. Berchuck, J.E.; Meyer, C.S.; Zhang, N.; Berchuck, C.M.; Trivedi, N.N.; Cohen, B.; Wang, S. Association of Mental Health Treatment with Outcomes for US Veterans Diagnosed with Non–Small Cell Lung Cancer. JAMA Oncol. 2020, 6, 1055–1062. [Google Scholar] [CrossRef] [PubMed]
  3. Bushe, C.J.; Hodgson, R. Schizophrenia and cancer: In 2010 do we understand the connection? Can. J. Psychiatry 2010, 55, 761–767. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Catts, V.S.; Catts, S.V.; O’Toole, B.I.; Frost, A.D. Cancer incidence in patients with schizophrenia and their first-degree relatives—A meta-analysis. Acta Psychiatr. Scand. 2008, 117, 323–336. [Google Scholar] [CrossRef] [PubMed]
  5. Li, H.; Li, J.; Yu, X.; Zheng, H.; Sun, X.; Lu, Y.; Zhang, Y.; Li, C.; Bi, X. The incidence rate of cancer in patients with schizophrenia: A meta-analysis of cohort studies. Schizophr. Res. 2018, 195, 519–528. [Google Scholar] [CrossRef]
  6. Mortensen, P.B. The occurrence of cancer in first admitted schizophrenic patients. Schizophr. Res. 1994, 12, 185–194. [Google Scholar] [CrossRef]
  7. Mortensen, P.B. The incidence of cancer in schizophrenic patients. J. Epidemiol. Community Health 1989, 43, 43–47. [Google Scholar] [CrossRef] [Green Version]
  8. Goldacre, M.J.; Kurina, L.M.; Wotton, C.J.; Yeates, D.; Seagroat, V. Schizophrenia and cancer: An epidemiological study. Br. J. Psychiatry 2005, 187, 334–338. [Google Scholar] [CrossRef] [Green Version]
  9. Lichtermann, D.; Ekelund, J.; Pukkala, E.; Tanskanen, A.; Lonnqvist, J. Incidence of cancer among persons with schizophrenia and their relatives. Arch. Gen. Psychiatry 2001, 58, 573–578. [Google Scholar] [CrossRef] [Green Version]
  10. Xu, D.; Chen, G.; Kong, L.; Zhang, W.; Hu, L.; Chen, C.; Li, J.; Zhuo, C. Lower risk of liver cancer in patients with schizophrenia: A systematic review and meta-analysis of cohort studies. Oncotarget 2017, 8, 102328–102335. [Google Scholar] [CrossRef] [Green Version]
  11. Zhuo, C.; Zhuang, H.; Gao, X.; Triplett, P.T. Lung cancer incidence in patients with schizophrenia: Meta-analysis. Br. J. Psychiatry 2019, 215, 704–711. [Google Scholar] [CrossRef]
  12. Hippisley-Cox, J.; Vinogradova, Y.; Coupland, C.; Parker, C. Risk of Malignancy in Patients with Schizophrenia or Bipolar Disorder: Nested Case-Control Study. Arch. Gen. Psychiatry 2007, 64, 1368–1376. [Google Scholar] [CrossRef] [Green Version]
  13. Osborn, D.P.; Limburg, H.; Walters, K.; Petersen, I.; King, M.; Green, J.; Watson, J.; Nazareth, I. Relative incidence of common cancers in people with severe mental illness. Cohort study in the United Kingdom THIN primary care database. Schizophr. Res. 2013, 143, 44–49. [Google Scholar] [CrossRef]
  14. Lasser, K.; Boyd, J.W.; Woolhandler, S.; Himmelstein, D.U.; McCormick, D.; Bor, D.H. Smoking and mental illness: A population-based prevalence study. J. Am. Med. Assoc. 2000, 284, 2606–2610. [Google Scholar] [CrossRef]
  15. Shinozaki, Y.; Nakao, M.; Takeuchi, T.; Yano, E. Smoking rates among schizophrenia patients in Japan. Psychiatry Res. 2011, 186, 165–169. [Google Scholar] [CrossRef]
  16. Campo-Arias, A.; Diaz-Martinez, L.A.; Rueda-Jaimes, G.E.; Rueda-Sanchez, M.; Farelo-Palacin, D.; Diaz, F.J.; de Leon, J. Smoking is associated with schizophrenia, but not with mood disorders, within a population with low smoking rates: A matched case-control study in Bucaramanga, Colombia. Schizophr. Res. 2006, 83, 269–276. [Google Scholar] [CrossRef]
  17. Murphy, K.A.; Stone, E.M.; Presskreischer, R.; McGinty, E.E.; Daumit, G.L.; Pollack, C.E. Cancer Screening Among Adults with and without Serious Mental Illness: A Mixed Methods Study. Med. Care 2021, 59, 327–333. [Google Scholar] [CrossRef]
  18. Hodgson, R.; Wildgust, H.J.; Bushe, C.J. Cancer and schizophrenia: Is there a paradox? J. Psychopharmacol. 2010, 24 (Suppl. 4), 51–60. [Google Scholar] [CrossRef]
  19. Hjorthoj, C.; Sturup, A.E.; McGrath, J.J.; Nordentoft, M. Years of potential life lost and life expectancy in schizophrenia: A systematic review and meta-analysis. Lancet Psychiatry 2017, 4, 295–301. [Google Scholar] [CrossRef]
  20. Kisely, S.; Crowe, E.; Lawrence, D. Cancer-related mortality in people with mental illness. JAMA Psychiatry 2013, 70, 209–217. [Google Scholar] [CrossRef] [Green Version]
  21. Ni, L.; Wu, J.; Long, Y.; Tao, J.; Xu, J.; Yuan, X.; Yu, N.; Wu, R.; Zhang, Y. Mortality of site-specific cancer in patients with schizophrenia: A systematic review and meta-analysis. BMC Psychiatry 2019, 19, 323. [Google Scholar] [CrossRef]
  22. Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target Ther. 2020, 5, 28. [Google Scholar] [CrossRef] [Green Version]
  23. Xiping, Z.; Shuai, Z.; Feijiang, Y.; Bo, C.; Shifeng, Y.; Qihui, C. Meta-analysis of the Correlation Between Schizophrenia and Breast Cancer. Clin. Breast Cancer 2019, 19, e172–e185. [Google Scholar] [CrossRef] [Green Version]
  24. Lu, D.; Song, J.; Lu, Y.; Fall, K.; Chen, X.; Fang, F.; Landen, M.; Hultman, C.M.; Czene, K.; Sullivan, P.; et al. A shared genetic contribution to breast cancer and schizophrenia. Nat. Commun. 2020, 11, 4637. [Google Scholar] [CrossRef]
  25. Yuan, K.; Song, W.; Liu, Z.; Lin, G.N.; Yu, S. Mendelian Randomization and GWAS Meta Analysis Revealed the Risk-Increasing Effect of Schizophrenia on Cancers. Biology 2022, 11, 1345. [Google Scholar] [CrossRef]
  26. Wootten, J.C.; Wiener, J.C.; Blanchette, P.S.; Anderson, K.K. Cancer incidence and stage at diagnosis among people with psychotic disorders: Systematic review and meta-analysis. Cancer Epidemiol. 2022, 80, 102233. [Google Scholar] [CrossRef]
  27. Ge, F.; Huo, Z.; Liu, Y.; Du, X.; Wang, R.; Lin, W.; Wang, R.; Chen, J.; Lu, Y.; Wen, Y.; et al. Association between schizophrenia and prostate cancer risk: Results from a pool of cohort studies and Mendelian randomization analysis. Compr. Psychiatry 2022, 115, 152308. [Google Scholar] [CrossRef]
  28. Durand, N.; Simsir, M.; Signetti, L.; Labbal, F.; Ballotti, R.; Mus-Veteau, I. Methiothepin Increases Chemotherapy Efficacy against Resistant Melanoma Cells. Molecules 2021, 26, 1867. [Google Scholar] [CrossRef]
  29. Matteoni, S.; Matarrese, P.; Ascione, B.; Buccarelli, M.; Ricci-Vitiani, L.; Pallini, R.; Villani, V.; Pace, A.; Paggi, M.G.; Abbruzzese, C. Anticancer Properties of the Antipsychotic Drug Chlorpromazine and Its Synergism with Temozolomide in Restraining Human Glioblastoma Proliferation In Vitro. Front. Oncol. 2021, 11, 635472. [Google Scholar] [CrossRef]
  30. Weissenrieder, J.S.; Reed, J.L.; Moldovan, G.L.; Johnson, M.T.; Trebak, M.; Neighbors, J.D.; Mailman, R.B.; Hohl, R.J. Antipsychotic drugs elicit cytotoxicity in glioblastoma multiforme in a calcium-dependent, non-D2 receptor-dependent, manner. Pharmacol. Res. Perspect. 2021, 9, e00689. [Google Scholar] [CrossRef]
  31. Zhuo, C.; Xun, Z.; Hou, W.; Ji, F.; Lin, X.; Tian, H.; Zheng, W.; Chen, M.; Liu, C.; Wang, W.; et al. Surprising Anticancer Activities of Psychiatric Medications: Old Drugs Offer New Hope for Patients with Brain Cancer. Front. Pharmacol. 2019, 10, 1262. [Google Scholar] [CrossRef]
  32. Ji, J.; Sundquist, K.; Ning, Y.; Kendler, K.S.; Sundquist, J.; Chen, X. Incidence of cancer in patients with schizophrenia and their first-degree relatives: A population-based study in Sweden. Schizophr. Bull. 2013, 39, 527–536. [Google Scholar] [CrossRef] [PubMed]
  33. Platten, M.; Nollen, E.A.A.; Rohrig, U.F.; Fallarino, F.; Opitz, C.A. Tryptophan metabolism as a common therapeutic target in cancer, neurodegeneration and beyond. Nat. Rev. Drug Discov. 2019, 18, 379–401. [Google Scholar] [CrossRef] [PubMed]
  34. Sarkar, C.; Chakroborty, D.; Basu, S. Neurotransmitters as regulators of tumor angiogenesis and immunity: The role of catecholamines. J. Neuroimmune Pharmacol. 2013, 8, 7–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Lara, D.R.; Souza, D.O. Schizophrenia: A purinergic hypothesis. Med. Hypotheses 2000, 54, 157–166. [Google Scholar] [CrossRef]
  36. Vigano, S.; Alatzoglou, D.; Irving, M.; Menetrier-Caux, C.; Caux, C.; Romero, P.; Coukos, G. Targeting Adenosine in Cancer Immunotherapy to Enhance T-Cell Function. Front. Immunol. 2019, 10, 925. [Google Scholar] [CrossRef] [Green Version]
  37. Shen, H.Y.; Singer, P.; Lytle, N.; Wei, C.J.; Lan, J.Q.; Williams-Karnesky, R.L.; Chen, J.F.; Yee, B.K.; Boison, D. Adenosine augmentation ameliorates psychotic and cognitive endophenotypes of schizophrenia. J. Clin. Investig. 2012, 122, 2567–2577. [Google Scholar] [CrossRef] [Green Version]
  38. Dunwiddie, T.V.; Masino, S.A. The role and regulation of adenosine in the central nervous system. Annu. Rev. Neurosci. 2001, 24, 31–55. [Google Scholar] [CrossRef] [Green Version]
  39. Boison, D.; Singer, P.; Shen, H.Y.; Feldon, J.; Yee, B.K. Adenosine hypothesis of schizophrenia—Opportunities for pharmacotherapy. Neuropharmacology 2012, 62, 1527–1543. [Google Scholar] [CrossRef] [Green Version]
  40. Blay, J.; White, T.D.; Hoskin, D.W. The extracellular fluid of solid carcinomas contains immunosuppressive concentrations of adenosine. Cancer Res. 1997, 57, 2602–2605. [Google Scholar]
  41. Cekic, C.; Day, Y.J.; Sag, D.; Linden, J. Myeloid expression of adenosine A2A receptor suppresses T and NK cell responses in the solid tumor microenvironment. Cancer Res. 2014, 74, 7250–7259. [Google Scholar] [CrossRef] [Green Version]
  42. Gessi, S.; Bencivenni, S.; Battistello, E.; Vincenzi, F.; Colotta, V.; Catarzi, D.; Varano, F.; Merighi, S.; Borea, P.A.; Varani, K. Inhibition of A2A Adenosine Receptor Signaling in Cancer Cells Proliferation by the Novel Antagonist TP455. Front. Pharmacol. 2017, 8, 888. [Google Scholar] [CrossRef]
  43. Huang, Y.; Gu, Z.; Fan, Y.; Zhai, G.; Zhao, X.; Sun, Q.; Shi, Y.; Lin, G. Inhibition of the adenosinergic pathway: The indispensable part of oncological therapy in the future. Purinergic Signal. 2019, 15, 53–67. [Google Scholar] [CrossRef]
  44. Boswell-Casteel, R.C.; Hays, F.A. Equilibrative nucleoside transporters—A review. Nucleosides Nucleotides Nucleic Acids 2017, 36, 7–30. [Google Scholar] [CrossRef]
  45. Cristalli, G.; Costanzi, S.; Lambertucci, C.; Lupidi, G.; Vittori, S.; Volpini, R.; Camaioni, E. Adenosine deaminase: Functional implications and different classes of inhibitors. Med. Res. Rev. 2001, 21, 105–128. [Google Scholar] [CrossRef]
  46. Zimmermann, H.; Zebisch, M.; Strater, N. Cellular function and molecular structure of ecto-nucleotidases. Purinergic Signal. 2012, 8, 437–502. [Google Scholar] [CrossRef] [Green Version]
  47. Li, A. Mechanisms of ATP release, the enabling step in purinergic dynamics. Cell. Physiol. Biochem. 2011, 28, 1135–1144. [Google Scholar] [CrossRef]
  48. Lohman, A.W. Mechanisms of ATP release and signaling in the blood vessel wall. Cardiovasc. Res. 2012, 95, 269–280. [Google Scholar] [CrossRef] [Green Version]
  49. Lazarowski, E.R. Molecular mechanisms of purine and pyrimidine nucleotide release. Adv. Pharmacol. 2011, 61, 2211–2261. [Google Scholar]
  50. Chavan, V.; Willis, J.; Walker, S.K.; Clark, H.R.; Liu, X.; Fox, M.A.; Srivastava, S.; Mukherjee, K. Correction: Central Presynaptic Terminals Are Enriched in ATP but the Majority Lack Mitochondria. PLoS ONE 2017, 12, e0181140. [Google Scholar] [CrossRef] [Green Version]
  51. Lindberg, D.; Shan, D.; Ayers-Ringler, J.; Oliveros, A.; Benitez, J.; Prieto, M.; McCullumsmith, R.; Choi, D.S. Purinergic signaling and energy homeostasis in psychiatric disorders. Curr. Mol. Med. 2015, 15, 275–295. [Google Scholar] [CrossRef] [Green Version]
  52. Mizumoto, N.; Kumamoto, T.; Robson, S.C.; Sévigny, J.; Matsue, H.; Enjyoji, K.; Takashima, A. CD39 is the dominant Langerhans cell-associated ecto-NTPDase: Modulatory roles in inflammation and immune responsiveness. Nat. Med. 2002, 8, 358–365. [Google Scholar] [CrossRef] [PubMed]
  53. Tozzi, M.G.; Pesi, R.; Allegrini, S. On the physiological role of cytosolic 5′-nucleotidase II (cN-II): Pathological and therapeutical implications. Curr. Med. Chem. 2013, 34, 4285–4291. [Google Scholar] [CrossRef] [PubMed]
  54. Romio, M.; Reinbeck, B.; Bongardt, S.; Hüls, S.; Burghoff, S.; Schrader, J. Extracellular purine metabolism and signaling of CD73-derived adenosine in murine Treg and Teff cells. Am. J. Physiol. Cell Physiol. 2011, 301, 530–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Robson, S. The E-NTPDase family of ectonucleotidases: Structure function relationships and pathophysiological significance. Purinergic Signal. 2006, 2, 409–430. [Google Scholar] [CrossRef] [Green Version]
  56. Cekic, G. Purinergic regulation of the immune system. Nature Rev. Immunol. 2016, 16, 177–192. [Google Scholar] [CrossRef]
  57. Friedman, D.J.; Kunzli, B.M.; Yi, A.R.; Sevigny, J.; Berberat, P.O.; Enjyoji, K.; Csizmadia, E.; Friess, H.; Robson, S.C. From the Cover: CD39 deletion exacerbates experimental murine colitis and human polymorphisms increase susceptibility to inflammatory bowel disease. Proc. Natl. Acad. Sci. USA 2009, 106, 16788–16793. [Google Scholar] [CrossRef] [Green Version]
  58. Louis, N.A.; Robinson, A.M.; MacManus, C.F.; Karhausen, J.; Scully, M.; Colgan, S.P. Control of IFN-alphaA by CD73: Implications for mucosal inflammation. J. Immunol. 2008, 180, 4246–4255. [Google Scholar] [CrossRef] [Green Version]
  59. Yegutkin, G.G. Nucleotide- and nucleoside-converting ectoenzymes: Important modulators of purinergic signalling cascade. Biochim. Biophys. Acta—Mol. Cell Res. 2008, 1783, 673–694. [Google Scholar] [CrossRef] [Green Version]
  60. Kopp, R.; Krautloher, A.; Ramirez-Fernandez, A.; Nicke, A. P2 × 7 Interactions and Signaling—Making Head or Tail of It. Front. Mol. Neurosci. 2019, 12, 183. [Google Scholar] [CrossRef]
  61. Samways, D.S.; Li, Z.; Egan, T.M. Principles and properties of ion flow in P2X receptors. Front. Cell. Neurosci. 2014, 8, 6. [Google Scholar] [CrossRef] [Green Version]
  62. Ormond, S.J.; Barrera, N.P.; Qureshi, O.S.; Henderson, R.M.; Edwardson, J.M.; Murrell-Lagnado, R.D. An uncharged region within the N terminus of the P2 × 6 receptor inhibits its assembly and exit from the endoplasmic reticulum. Mol. Pharmacol. 2006, 69, 1692–1700. [Google Scholar] [CrossRef] [Green Version]
  63. Jeong, Y.H.; Walsh, M.C.; Yu, J.; Shen, H.; Wherry, E.J.; Choi, Y. Mice Lacking the Purinergic Receptor P2 × 5 Exhibit Defective Inflammasome Activation and Early Susceptibility to Listeria monocytogenes. J. Immunol. 2020, 205, 760–766. [Google Scholar] [CrossRef]
  64. Stokes, L.; Layhadi, J.A.; Bibic, L.; Dhuna, K.; Fountain, S.J. P2 × 4 Receptor Function in the Nervous System and Current Breakthroughs in Pharmacology. Front. Pharmacol. 2017, 8, 291. [Google Scholar] [CrossRef] [Green Version]
  65. Fabbretti, E. ATP P2 × 3 receptors and neuronal sensitization. Front. Cell. Neurosci. 2013, 7, 236. [Google Scholar] [CrossRef] [Green Version]
  66. Richler, E.; Shigetomi, E.; Khakh, B.S. Neuronal P2 × 2 receptors are mobile ATP sensors that explore the plasma membrane when activated. J. Neurosci. 2011, 31, 16716–16730. [Google Scholar] [CrossRef] [Green Version]
  67. Mahaut-Smith, M.P.; Jones, S.; Evans, R.J. The P2 × 1 receptor and platelet function. Purinergic Signal. 2011, 7, 341–356. [Google Scholar] [CrossRef] [Green Version]
  68. Erb, L.; Weisman, G.A. Coupling of P2Y receptors to G proteins and other signaling pathways. Wiley Interdiscip. Rev. Membr. Transp. Signal. 2012, 1, 789–803. [Google Scholar] [CrossRef] [Green Version]
  69. Olah, M.E. Adenosine receptor subtypes: Characterization and therapeutic regulation. Annu. Rev. Pharmacol. Toxicol. 1995, 35, 581–606. [Google Scholar] [CrossRef]
  70. Ralevic, V.; Burnstock, G. Receptors for purines and pyrimidines. Pharmacol. Rev. 1998, 50, 413–492. [Google Scholar]
  71. Klotz, K.N.; Hessling, J.; Hegler, J.; Owman, C.; Kull, B.; Fredholm, B.B.; Lohse, M.J. Comparative pharmacology of human adenosine receptor subtypes—Characterization of stably transfected receptors in CHO cells. Naunyn-Schmiedeberg Arch. Pharmacol. 1998, 357, 1–9. [Google Scholar] [CrossRef]
  72. Fredholm, B.B.; Irenius, E.; Kull, B.; Schulte, G. Comparison of the potency of adenosine as an agonist at human adenosine receptors expressed in Chinese hamster ovary cells. Biochem. Pharmacol. 2001, 61, 443–448. [Google Scholar] [CrossRef]
  73. Schulte, G.; Fredholm, B.B. Human adenosine A(1), A(2A), A(2B), and A(3) receptors expressed in Chinese hamster ovary cells all mediate the phosphorylation of extracellular-regulated kinase 1/2. Mol. Pharmacol. 2000, 58, 477–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Haskó, G. Adenosine receptor signaling in the brain immune system. Trends Pharmacol. Sci. 2005, 26, 511–516. [Google Scholar] [CrossRef] [PubMed]
  75. Augusto, E.; Matos, M.; Sevigny, J.; El-Tayeb, A.; Bynoe, M.S.; Muller, C.E.; Cunha, R.A.; Chen, J.F. Ecto-5’-nucleotidase (CD73)-mediated formation of adenosine is critical for the striatal adenosine A2A receptor functions. J. Neurosci. 2013, 33, 11390–11399. [Google Scholar] [CrossRef] [Green Version]
  76. Rebola, N.; Lujan, R.; Cunha, R.A.; Mulle, C. Adenosine A2A receptors are essential for long-term potentiation of NMDA-EPSCs at hippocampal mossy fiber synapses. Neuron 2008, 57, 121–134. [Google Scholar] [CrossRef] [Green Version]
  77. Cunha, R.A.; Correia-de-Sa, P.; Sebastiao, A.M.; Ribeiro, J.A. Preferential activation of excitatory adenosine receptors at rat hippocampal and neuromuscular synapses by adenosine formed from released adenine nucleotides. Br. J. Pharmacol. 1996, 119, 253–260. [Google Scholar] [CrossRef] [Green Version]
  78. Cunha, R.A. How does adenosine control neuronal dysfunction and neurodegeneration? J. Neurochem. 2016, 139, 1019–1055. [Google Scholar] [CrossRef]
  79. Cunha, R.A. Neuroprotection by adenosine in the brain: From A(1) receptor activation to A (2A) receptor blockade. Purinergic Signal. 2005, 1, 111–134. [Google Scholar] [CrossRef] [Green Version]
  80. Lopes, C.R.; Lourenco, V.S.; Tome, A.R.; Cunha, R.A.; Canas, P.M. Use of knockout mice to explore CNS effects of adenosine. Biochem. Pharmacol. 2021, 187, 114367. [Google Scholar] [CrossRef]
  81. Agnati, L.F.; Ferre, S.; Lluis, C.; Franco, R.; Fuxe, K. Molecular mechanisms and therapeutical implications of intramembrane receptor/receptor interactions among heptahelical receptors with examples from the striatopallidal GABA neurons. Pharmacol. Rev. 2003, 55, 509–550. [Google Scholar] [CrossRef] [Green Version]
  82. Ferre, S.; von Euler, G.; Johansson, B.; Fredholm, B.B.; Fuxe, K. Stimulation of high-affinity adenosine A2 receptors decreases the affinity of dopamine D2 receptors in rat striatal membranes. Proc. Natl. Acad. Sci. USA 1991, 88, 7238–7241. [Google Scholar] [CrossRef] [Green Version]
  83. Ferre, S.; Fredholm, B.B.; Morelli, M.; Popoli, P.; Fuxe, K. Adenosine-dopamine receptor-receptor interactions as an integrative mechanism in the basal ganglia. Trends Neurosci. 1997, 20, 482–487. [Google Scholar] [CrossRef]
  84. Cervetto, C.; Venturini, A.; Passalacqua, M.; Guidolin, D.; Genedani, S.; Fuxe, K.; Borroto-Esquela, D.O.; Cortelli, P.; Woods, A.; Maura, G.; et al. A2A-D2 receptor-receptor interaction modulates gliotransmitter release from striatal astrocyte processes. J. Neurochem. 2017, 140, 268–279. [Google Scholar] [CrossRef]
  85. Hasko, G.; Cronstein, B.N. Adenosine: An endogenous regulator of innate immunity. Trends Immunol. 2004, 25, 33–39. [Google Scholar] [CrossRef]
  86. Panther, E.; Corinti, S.; Idzko, M.; Herouy, Y.; Napp, M.; La Sala, A.; Girolomoni, G.; Norgauer, J. Adenosine affects expression of membrane molecules, cytokine and chemokine release, and the T-cell stimulatory capacity of human dendritic cells. Blood 2003, 101, 3985–3990. [Google Scholar] [CrossRef] [Green Version]
  87. Chekeni, F.B.; Elliott, M.R.; Sandilos, J.K.; Walk, S.F.; Kinchen, J.M.; Lazarowski, E.R.; Armstrong, A.J.; Penuela, S.; Laird, D.W.; Salvesen, G.S.; et al. Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis. Nature 2010, 467, 863–867. [Google Scholar] [CrossRef] [Green Version]
  88. Krysko, D.V.; Garg, A.D.; Kaczmarek, A.; Krysko, O.; Agostinis, P.; Vandenabeele, P. Immunogenic cell death and DAMPs in cancer therapy. Nat. Rev. Cancer 2012, 12, 860–875. [Google Scholar] [CrossRef]
  89. Dosch, M. Mechanisms of ATP Release by Inflammatory Cells. Int. J. Mol. Sci. 2018, 19, 1222. [Google Scholar] [CrossRef] [Green Version]
  90. Borea, P.A.; Gessi, S.; Merighi, S.; Varani, K. Adenosine as a multi-signalling guardian angel in human diseases: When, where and how does it exert its protective effects? Trends Pharmacol. Sci. 2016, 37, 419–434. [Google Scholar] [CrossRef]
  91. Ernst, P.B.; Garrison, J.C.; Thompson, L.F. Much ado about adenosine: Adenosine synthesis and function in regulatory T cell biology. J. Immunol. 2010, 185, 1993–1998. [Google Scholar] [CrossRef] [Green Version]
  92. Di Virgilio, F.; Vuerich, M. Purinergic signaling in the immune system. Auton. Neurosci. 2015, 191, 117–123. [Google Scholar] [CrossRef] [Green Version]
  93. Wang, X.; Qin, W.; Xu, X.; Xiong, Y.; Zhang, Y.; Zhang, H.; Sun, B. Endotoxin-induced autocrine ATP signaling inhibits neutrophil chemotaxis through enhancing myosin light chain phosphorylation. Proc. Natl. Acad. Sci. USA 2017, 114, 4483–4488. [Google Scholar] [CrossRef] [Green Version]
  94. Koshiba, M.; Kojima, H.; Huang, S.; Apasov, S.; Sitkovsky, M.V. Memory of extracellular adenosine A2A purinergic receptor-mediated signalling in murine T cells. J. Biol. Chem. 1997, 272, 25881–25889. [Google Scholar] [CrossRef]
  95. Ernens, I. Adenosine up-regulates vascular endothelial growth factor in human macrophages. Biochem. Biophys. Res. Commun. 2010, 392, 351–356. [Google Scholar] [CrossRef]
  96. Novitskiy, S.V. Adenosine receptors in regulation of dendritic cell differentiation and function. Blood 2008, 112, 1822–1831. [Google Scholar] [CrossRef]
  97. Abbas, M.; Verville, J.; Mroue, K. Resolution of bronchoconstriction with positive airway pressure after intravenous adenosine. Am. J. Emerg. Med. 2021, 44, e481–e482. [Google Scholar] [CrossRef]
  98. Shams, S.; Martinez, J.M.; Dawson, J.R.D.; Flores, J.; Gabriel, M.; Garcia, G.; Guevara, A.; Murray, K.; Pacifici, N.; Vargas, M.V.; et al. The Therapeutic Landscape of Rheumatoid Arthritis: Current State and Future Directions. Front. Pharmacol. 2021, 12, 680043. [Google Scholar] [CrossRef]
  99. Yao, J.K.; Reddy, R.; van Kammen, D.P. Reduced level of plasma antioxidant uric acid in schizophrenia. Psychiatry Res. 1998, 80, 29–39. [Google Scholar] [CrossRef]
  100. Reddy, R.; Keshavan, M.; Yao, J.K. Reduced plasma antioxidants in first-episode patients with schizophrenia. Schizophr. Res. 2003, 62, 205–212. [Google Scholar] [CrossRef]
  101. Nagamine, T. Abnormal laboratory values during the acute and recovery phases in schizophrenic patients: A retrospective study. Neuropsychiatr. Dis. Treat. 2010, 6, 281–288. [Google Scholar] [CrossRef] [Green Version]
  102. Gültekin, B.K.; Kesebir, S.; Kabak, S.G.; Ergün, F.F.; Yaylaci, E.T. Are Uric Acid Levels Different from Healthy Subjects in Bipolar Affective Disorder and Schizophrenia? Relationship Between Clinical Improvement and Episode Severity in Male Patients. Nöro Psikiyatr. Arşivi 2014, 51, 229–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. He, Q.; You, Y.; Yu, L.; Yao, L.; Lu, H.; Zhou, X.; Wu, S.; Chen, L.; Chen, Y.; Zhao, X. Uric acid levels in subjects with schizophrenia: A systematic review and meta-analysis. Psychiatry Res. 2020, 292, 113305. [Google Scholar] [CrossRef] [PubMed]
  104. Kang, D.H.; Ha, S.K. Uric Acid Puzzle: Dual Role as Anti-oxidantand Pro-oxidant. Electrolyte Blood Press. 2014, 12, 1–6. [Google Scholar] [CrossRef] [PubMed]
  105. Ames, B.N.; Cathcart, R.; Schwiers, E.; Hochstein, P. Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: A hypothesis. Proc. Natl. Acad. Sci. USA 1981, 78, 6858–6862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Godin, O.; Leboyer, M.; Gaman, A.; Aouizerate, B.; Berna, F.; Brunel, L.; Capdevielle, D.; Chereau, I.; Dorey, J.M.; Dubertret, C.; et al. Metabolic syndrome, abdominal obesity and hyperuricemia in schizophrenia: Results from the FACE-SZ cohort. Schizophr. Res. 2015, 168, 388–394. [Google Scholar] [CrossRef]
  107. Rajan, S.; Zalpuri, I.; Harrington, A.; Cimpeanu, C.; Song, X.; Fan, X. Relationship between serum uric acid level and cardiometabolic risks in nondiabetic patients with schizophrenia. Int. Clin. Psychopharmacol. 2016, 31, 51–56. [Google Scholar] [CrossRef]
  108. Flatow, J.; Buckley, P.; Miller, B.J. Meta-analysis of oxidative stress in schizophrenia. Biol. Psychiatry 2013, 74, 400–409. [Google Scholar] [CrossRef] [Green Version]
  109. Ermakov, E.A.; Dmitrieva, E.M.; Parshukova, D.A.; Kazantseva, D.V.; Vasilieva, A.R.; Smirnova, L.P. Oxidative Stress-Related Mechanisms in Schizophrenia Pathogenesis and New Treatment Perspectives. Oxid. Med. Cell. Longev. 2021, 2021, 8881770. [Google Scholar] [CrossRef]
  110. Arvindakshan, M.; Ghate, M.; Ranjekar, P.K.; Evans, D.R.; Mahadik, S.P. Supplementation with a combination of omega-3 fatty acids and antioxidants (vitamins E and C) improves the outcome of schizophrenia. Schizophr. Res. 2003, 62, 195–204. [Google Scholar] [CrossRef]
  111. Mahadik, S.P.; Pillai, A.; Joshi, S.; Foster, A. Prevention of oxidative stress-mediated neuropathology and improved clinical outcome by adjunctive use of a combination of antioxidants and omega-3 fatty acids in schizophrenia. Int. Rev. Psychiatry 2006, 18, 119–131. [Google Scholar] [CrossRef]
  112. Ng, F.; Berk, M.; Dean, O.; Bush, A.I. Oxidative stress in psychiatric disorders: Evidence base and therapeutic implications. Int. J. Neuropsychopharmacol. 2008, 11, 851–876. [Google Scholar] [CrossRef] [Green Version]
  113. Yao, J.K.; Condray, R.; Dougherty, G.G., Jr.; Keshavan, M.S.; Montrose, D.M.; Matson, W.R.; McEvoy, J.; Kaddurah-Daouk, R.; Reddy, R.D. Associations between purine metabolites and clinical symptoms in schizophrenia. PLoS ONE 2012, 7, e42165. [Google Scholar] [CrossRef] [Green Version]
  114. Ben Salem, C.; Slim, R.; Fathallah, N.; Hmouda, H. Drug-induced hyperuricaemia and gout. Rheumatology 2017, 56, 679–688. [Google Scholar] [CrossRef]
  115. Mi, S.; Gong, L.; Sui, Z. Friend or Foe? An Unrecognized Role of Uric Acid in Cancer Development and the Potential Anticancer Effects of Uric Acid-lowering Drugs. J. Cancer 2020, 11, 5236–5244. [Google Scholar] [CrossRef]
  116. Yang, J.; Wang, Y.; Zhao, Q.; Zhang, X.; Wang, X.; Qin, X.; Zhang, R.; Shen, L.; Jiang, X.; Jiang, H.; et al. Association of serum uric acid with increased risk of cancer among hypertensive Chinese. Int. J. Cancer 2017, 141, 112–120. [Google Scholar] [CrossRef] [Green Version]
  117. Mao, L.; Guo, C.; Zheng, S. Elevated urinary 8-oxo-7,8-dihydro-2’-deoxyguanosine and serum uric acid are associated with progression and are prognostic factors of colorectal cancer. Onco Targets Ther. 2018, 11, 5895–5902. [Google Scholar] [CrossRef] [Green Version]
  118. Chen, Y.F.; Li, Q.; Chen, D.T.; Pan, J.H.; Chen, Y.H.; Wen, Z.S.; Zeng, W.A. Prognostic value of pre-operative serum uric acid levels in esophageal squamous cell carcinoma patients who undergo R0 esophagectomy. Cancer Biomark. 2016, 17, 89–96. [Google Scholar] [CrossRef]
  119. Yuan, C.; Xu, X.H.; Wang, X.L.; Xu, L.; Chen, Z.; Li, Y.Q. Relationship between serum uric acid and metastatic and nonmetastatic rectal cancer patients with undergoing no chemotherapy. Medicine 2016, 95, e5463. [Google Scholar] [CrossRef]
  120. Stotz, M.; Szkandera, J.; Seidel, J.; Stojakovic, T.; Samonigg, H.; Reitz, D.; Gary, T.; Kornprat, P.; Schaberl-Moser, R.; Hoefler, G.; et al. Evaluation of uric acid as a prognostic blood-based marker in a large cohort of pancreatic cancer patients. PLoS ONE 2014, 9, e104730. [Google Scholar] [CrossRef]
  121. Stevenson, W.S.; Hyland, C.D.; Zhang, J.G.; Morgan, P.O.; Willson, T.A.; Gill, A.; Hilton, A.A.; Viney, E.M.; Bahlo, M.; Masters, S.L.; et al. Deficiency of 5-hydroxyisourate hydrolase causes hepatomegaly and hepatocellular carcinoma in mice. Proc. Natl. Acad. Sci. USA 2010, 107, 16625–16630. [Google Scholar] [CrossRef] [Green Version]
  122. Serra, S.; Horenstein, A.L.; Vaisitti, T.; Brusa, D.; Rossi, D.; Laurenti, L.; D’Arena, G.; Coscia, M.; Tripodo, C.; Inghirami, G.; et al. CD73-generated extracellular adenosine in chronic lymphocytic leukemia creates local conditions counteracting drug-induced cell death. Blood 2011, 118, 6141–6152. [Google Scholar] [CrossRef]
  123. Ohta, A.; Gorelik, E.; Prasad, S.J.; Ronchese, F.; Lukashev, D.; Wong, M.K.; Huang, X.; Caldwell, S.; Liu, K.; Smith, P.; et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc. Natl. Acad. Sci. USA 2006, 103, 13132–13137. [Google Scholar] [CrossRef] [Green Version]
  124. Roberts, E.L.; Roberts, O.T. Plasma adenosine deaminase isoform 2 in cancer patients undergoing chemotherapy. Br. J. Biomed. Sci. 2012, 69, 11–13. [Google Scholar] [CrossRef]
  125. Aghaei, M.; Karami-Tehrani, F.; Salami, S.; Atri, M. Adenosine deaminase activity in the serum and malignant tumors of breast cancer: The assessment of isoenzyme ADA1 and ADA2 activities. Clin. Biochem. 2005, 38, 887–891. [Google Scholar] [CrossRef] [PubMed]
  126. McGrath, J.; Saha, S.; Welham, J.; El Saadi, O.; MacCauley, C.; Chant, D. A systematic review of the incidence of schizophrenia: The distribution of rates and the influence of sex, urbanicity, migrant status and methodology. BMC Med. 2004, 2, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Saha, S.; Chant, D.; Welham, J.; McGrath, J. A systematic review of the prevalence of schizophrenia. PLoS Med. 2005, 2, e141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Krugel, U. Purinergic receptors in psychiatric disorders. Neuropharmacology 2016, 104, 212–225. [Google Scholar] [CrossRef] [PubMed]
  129. Shan, D.; Haroutunian, V.; Meador-Woodruff, J.H.; McCullumsmith, R.E. Expression of equilibrative nucleoside transporter type 1 protein in elderly patients with schizophrenia. Neuroreport 2012, 23, 224–227. [Google Scholar] [CrossRef] [Green Version]
  130. Aliagas, E.; Villar-Menendez, I.; Sevigny, J.; Roca, M.; Romeu, M.; Ferrer, I.; Martin-Satue, M.; Barrachina, M. Reduced striatal ecto-nucleotidase activity in schizophrenia patients supports the “adenosine hypothesis”. Purinergic Signal. 2013, 9, 599–608. [Google Scholar] [CrossRef] [Green Version]
  131. O’Donovan, S.M.; Sullivan, C.; Koene, R.; Devine, E.; Hasselfeld, K.; Moody, C.L.; McCullumsmith, R.E. Cell-subtype-specific changes in adenosine pathways in schizophrenia. Neuropsychopharmacology 2018, 43, 1667–1674. [Google Scholar] [CrossRef] [Green Version]
  132. Boison, D. Adenosine kinase: Exploitation for therapeutic gain. Pharmacol. Rev. 2013, 65, 906–943. [Google Scholar] [CrossRef] [Green Version]
  133. Villar-Menendez, I.; Diaz-Sanchez, S.; Blanch, M.; Albasanz, J.L.; Pereira-Veiga, T.; Monje, A.; Planchat, L.M.; Ferrer, I.; Martin, M.; Barrachina, M. Reduced striatal adenosine A2A receptor levels define a molecular subgroup in schizophrenia. J. Psychiatr. Res. 2014, 51, 49–59. [Google Scholar] [CrossRef]
  134. Villar-Menendez, I.; Porta, S.; Buira, S.P.; Pereira-Veiga, T.; Diaz-Sanchez, S.; Albasanz, J.L.; Ferrer, I.; Martin, M.; Barrachina, M. Increased striatal adenosine A2A receptor levels is an early event in Parkinson’s disease-related pathology and it is potentially regulated by miR-34b. Neurobiol. Dis. 2014, 69, 206–214. [Google Scholar] [CrossRef]
  135. Deckert, J.; Brenner, M.; Durany, N.; Zochling, R.; Paulus, W.; Ransmayr, G.; Tatschner, T.; Danielczyk, W.; Jellinger, K.; Riederer, P. Up-regulation of striatal adenosine A(2A) receptors in schizophrenia. Neuroreport 2003, 14, 313–316. [Google Scholar] [CrossRef]
  136. Valle-Leon, M.; Callado, L.F.; Aso, E.; Cajiao-Manrique, M.M.; Sahlholm, K.; Lopez-Cano, M.; Soler, C.; Altafaj, X.; Watanabe, M.; Ferre, S.; et al. Decreased striatal adenosine A2A-dopamine D2 receptor heteromerization in schizophrenia. Neuropsychopharmacology 2021, 46, 665–672. [Google Scholar] [CrossRef]
  137. Brunstein, M.G.; Ghisolfi, E.S.; Ramos, F.L.; Lara, D.R. A clinical trial of adjuvant allopurinol therapy for moderately refractory schizophrenia. J. Clin. Psychiatry 2005, 66, 213–219. [Google Scholar] [CrossRef]
  138. Dickerson, F.B.; Stallings, C.R.; Origoni, A.E.; Sullens, A.; Khushalani, S.; Sandson, N.; Yolken, R.H. A double-blind trial of adjunctive allopurinol for schizophrenia. Schizophr. Res. 2009, 109, 66–69. [Google Scholar] [CrossRef]
  139. Akhondzadeh, S.; Shasavand, E.; Jamilian, H.; Shabestari, O.; Kamalipour, A. Dipyridamole in the treatment of schizophrenia: Adenosine-dopamine receptor interactions. J. Clin. Pharm. Ther. 2000, 25, 131–137. [Google Scholar] [CrossRef]
  140. Akhondzadeh, S.; Safarcherati, A.; Amini, H. Beneficial antipsychotic effects of allopurinol as add-on therapy for schizophrenia: A double blind, randomized and placebo controlled trial. Prog. Neuropsychopharmacol. Biol. Psychiatry 2005, 29, 253–259. [Google Scholar] [CrossRef]
  141. Wonodi, I.; Gopinath, H.V.; Liu, J.; Adami, H.; Hong, L.E.; Allen-Emerson, R.; McMahon, R.P.; Thaker, G.K. Dipyridamole monotherapy in schizophrenia: Pilot of a novel treatment approach by modulation of purinergic signaling. Psychopharmacology 2011, 218, 341–345. [Google Scholar] [CrossRef] [Green Version]
  142. Weiser, M.; Gershon, A.A.; Rubinstein, K.; Petcu, C.; Ladea, M.; Sima, D.; Podea, D.; Keefe, R.S.; Davis, J.M. A randomized controlled trial of allopurinol vs. placebo added on to antipsychotics in patients with schizophrenia or schizoaffective disorder. Schizophr. Res. 2012, 138, 35–38. [Google Scholar] [CrossRef]
  143. Lintunen, J.; Lahteenvuo, M.; Tiihonen, J.; Tanskanen, A.; Taipale, H. Adenosine modulators and calcium channel blockers as add-on treatment for schizophrenia. NPJ Schizophr. 2021, 7, 1. [Google Scholar] [CrossRef]
  144. Huang, H.; Zheng, S.; Chen, M.; Xie, L.; Li, Z.; Guo, M.; Wang, J.; Lu, M.; Zhu, X. The potential of the P2X7 receptor as a therapeutic target in a sub-chronic PCP-induced rodent model of schizophrenia. J. Chem. Neuroanat. 2021, 116, 101993. [Google Scholar] [CrossRef]
  145. Wardas, J. Potential role of adenosine A2A receptors in the treatment of schizophrenia. Front. Biosci.-Landmark 2008, 13, 4071–4096. [Google Scholar] [CrossRef] [Green Version]
  146. Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [Green Version]
  147. Lawson, D.A.; Kessenbrock, K.; Davis, R.T.; Pervolarakis, N.; Werb, Z. Tumour heterogeneity and metastasis at single-cell resolution. Nat. Cell Biol. 2018, 20, 1349–1360. [Google Scholar] [CrossRef]
  148. Plaks, V.; Kong, N.; Werb, Z. The cancer stem cell niche: How essential is the niche in regulating stemness of tumor cells? Cell Stem Cell 2015, 16, 225–238. [Google Scholar] [CrossRef] [Green Version]
  149. Takai, K.; Le, A.; Weaver, V.M.; Werb, Z. Targeting the cancer-associated fibroblasts as a treatment in triple-negative breast cancer. Oncotarget 2016, 7, 82889–82901. [Google Scholar] [CrossRef] [Green Version]
  150. Di Virgilio, F.; Adinolfi, E. Extracellular purines, purinergic receptors and tumor growth. Oncogene 2017, 36, 293–303. [Google Scholar] [CrossRef] [Green Version]
  151. Allard, B.; Longhi, M.S.; Robson, S.C.; Stagg, J. The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets. Immunol. Rev. 2017, 276, 121–144. [Google Scholar] [CrossRef] [Green Version]
  152. Ohta, A.; Sitkovsky, M. Role of G-protein-coupled adenosine receptors in downregulation of inflammation and protection from tissue damage. Nature 2001, 414, 916–920. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Vecchio, E.A.; Tan, C.Y.; Gregory, K.J.; Christopoulos, A.; White, P.J.; May, L.T. Ligand-Independent Adenosine A2B Receptor Constitutive Activity as a Promoter of Prostate Cancer Cell Proliferation. J. Pharmacol. Exp. Ther. 2016, 357, 36–44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Vannoni, D.; Bernini, A.; Carlucci, F.; Civitelli, S.; Di Pietro, M.C.; Leoncini, R.; Rosi, F.; Tabucchi, A.; Tanzini, G.; Marinello, E. Enzyme activities controlling adenosine levels in normal and neoplastic tissues. Med. Oncol. 2004, 21, 187–195. [Google Scholar] [CrossRef] [PubMed]
  155. Mahajan, M.; Tiwari, N.; Sharma, R.; Kaur, S.; Singh, N. Oxidative stress and its relationship with adenosine deaminase activity in various stages of breast cancer. Indian J. Clin. Biochem. 2013, 28, 51–54. [Google Scholar] [CrossRef] [Green Version]
  156. Ghaderi, B.; Amini, S.; Maroofi, F.; Jalali, C.; Javanmardi, M.; Roshani, D.; Abdi, M. Adenosine Deaminase Activity in Chronic Lymphocytic Leukemia and Healthy Subjects. Iran. J. Cancer Prev. 2016, 9, e5069. [Google Scholar] [CrossRef] [Green Version]
  157. Biri, H.; Ozturk, H.S.; Kacmaz, M.; Karaca, K.; Tokucoglu, H.; Durak, I. Activities of DNA turnover and free radical metabolizing enzymes in cancerous human prostate tissue. Cancer Investig. 1999, 17, 314–319. [Google Scholar] [CrossRef]
  158. Namiot, Z.; Stasiewicz, J.; Namiot, A.; Kemona, A.; Kralisz, M.; Gorski, J. Adenosine deaminase activity in patients with the intestinal type of gastric carcinoma. Cancer Lett. 1996, 109, 199–202. [Google Scholar] [CrossRef]
  159. Murray, J.L.; Perez-Soler, R.; Bywaters, D.; Hersh, E.M. Decreased adenosine deaminase (ADA) and 5’nucleotidase (5NT) activity in peripheral blood T cells in Hodgkin disease. Am. J. Hematol. 1986, 21, 57–66. [Google Scholar] [CrossRef]
  160. Turcotte, M.; Spring, K.; Pommey, S.; Chouinard, G.; Cousineau, I.; George, J.; Chen, G.M.; Gendoo, D.M.; Haibe-Kains, B.; Karn, T.; et al. CD73 is associated with poor prognosis in high-grade serous ovarian cancer. Cancer Res. 2015, 75, 4494–4503. [Google Scholar] [CrossRef] [Green Version]
  161. Cai, X.Y.; Wang, X.F.; Li, J.; Dong, J.N.; Liu, J.Q.; Li, N.P.; Yun, B.; Xia, R.L. Overexpression of CD39 and high tumoral CD39(+)/CD8(+) ratio are associated with adverse prognosis in resectable gastric cancer. Int. J. Clin. Exp. Pathol. 2015, 8, 14757–14764. [Google Scholar]
  162. Inoue, Y.; Yoshimura, K.; Kurabe, N.; Kahyo, T.; Kawase, A.; Tanahashi, M.; Ogawa, H.; Inui, N.; Funai, K.; Shinmura, K.; et al. Prognostic impact of CD73 and A2A adenosine receptor expression in non-small-cell lung cancer. Oncotarget 2017, 8, 8738–8751. [Google Scholar] [CrossRef] [Green Version]
  163. Leclerc, B.G.; Charlebois, R.; Chouinard, G.; Allard, B.; Pommey, S.; Saad, F.; Stagg, J. CD73 Expression Is an Independent Prognostic Factor in Prostate Cancer. Clin. Cancer Res. 2016, 22, 158–166. [Google Scholar] [CrossRef] [Green Version]
  164. Tondell, A.; Wahl, S.G.F.; Sponaas, A.M.; Sorhaug, S.; Borset, M.; Haug, M. Ectonucleotidase CD39 and Checkpoint Signalling Receptor Programmed Death 1 are Highly Elevated in Intratumoral Immune Cells in Non-small-cell Lung Cancer. Transl. Oncol. 2020, 13, 17–24. [Google Scholar] [CrossRef]
  165. Nagate, Y.; Ezoe, S.; Fujita, J.; Okuzakis, D.; Motooka, D.; Ishibashi, T.; Ichii, M.; Tanimura, A.; Kurashige, M.; Morii, E.; et al. Ectonucleotidase CD39 is highly expressed on ATLL cells and is responsible for their immunosuppressive function. Leukemia 2020, 35, 107–118. [Google Scholar] [CrossRef]
  166. Mandapathil, M.; Boduc, M.; Roessler, M.; Guldner, C.; Walliczek-Dworschak, U.; Mandic, R. Ectonucleotidase CD39 expression in regional metastases in head and neck cancer. Acta Otolaryngol. 2018, 138, 428–432. [Google Scholar] [CrossRef]
  167. Li, J.; Wang, L.; Chen, X.; Li, L.; Li, Y.; Ping, Y.; Huang, L.; Yue, D.; Zhang, Z.; Wang, F.; et al. CD39/CD73 upregulation on myeloid-derived suppressor cells via TGF-beta-mTOR-HIF-1 signaling in patients with non-small cell lung cancer. Oncoimmunology 2017, 6, e1320011. [Google Scholar] [CrossRef] [Green Version]
  168. Zhang, B.; Cheng, B.; Li, F.S.; Ding, J.H.; Feng, Y.Y.; Zhuo, G.Z.; Wei, H.F.; Zhao, K. High expression of CD39/ENTPD1 in malignant epithelial cells of human rectal adenocarcinoma. Tumour Biol. 2015, 36, 9411–9419. [Google Scholar] [CrossRef]
  169. Quezada, C.; Garrido, W.; Oyarzun, C.; Fernandez, K.; Segura, R.; Melo, R.; Casanello, P.; Sobrevia, L.; San Martin, R. 5′-ectonucleotidase mediates multiple-drug resistance in glioblastoma multiforme cells. J. Cell. Physiol. 2013, 228, 602–608. [Google Scholar] [CrossRef]
  170. Loi, S.; Pommey, S.; Haibe-Kains, B.; Beavis, P.A.; Darcy, P.K.; Smyth, M.J.; Stagg, J. CD73 promotes anthracycline resistance and poor prognosis in triple negative breast cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 11091–11096. [Google Scholar] [CrossRef] [Green Version]
  171. Cappellari, A.R.; Rockenbach, L.; Dietrich, F.; Clarimundo, V.; Glaser, T.; Braganhol, E.; Abujamra, A.L.; Roesler, R.; Ulrich, H.; Battastini, A.M. Characterization of ectonucleotidases in human medulloblastoma cell lines: Ecto-5’NT/CD73 in metastasis as potential prognostic factor. PLoS ONE 2012, 7, e47468. [Google Scholar] [CrossRef]
  172. Hilchey, S.P.; Kobie, J.J.; Cochran, M.R.; Secor-Socha, S.; Wang, J.C.; Hyrien, O.; Burack, W.R.; Mosmann, T.R.; Quataert, S.A.; Bernstein, S.H. Human follicular lymphoma CD39+-infiltrating T cells contribute to adenosine-mediated T cell hyporesponsiveness. J. Immunol. 2009, 183, 6157–6166. [Google Scholar] [CrossRef] [Green Version]
  173. Azambuja, J.H.; Schuh, R.S.; Michels, L.R.; Iser, I.C.; Beckenkamp, L.R.; Roliano, G.G.; Lenz, G.S.; Scholl, J.N.; Sevigny, J.; Wink, M.R.; et al. Blockade of CD73 delays glioblastoma growth by modulating the immune environment. Cancer Immunol. Immunother. 2020, 69, 1801–1812. [Google Scholar] [CrossRef]
  174. Kashyap, A.S.; Thelemann, T.; Klar, R.; Kallert, S.M.; Festag, J.; Buchi, M.; Hinterwimmer, L.; Schell, M.; Michel, S.; Jaschinski, F.; et al. Antisense oligonucleotide targeting CD39 improves anti-tumor T cell immunity. J. Immunother. Cancer 2019, 7, 67. [Google Scholar] [CrossRef] [Green Version]
  175. Wang, R.; Zhang, Y.; Lin, X.; Gao, Y.; Zhu, Y. Prognositic value of CD73-adenosinergic pathway in solid tumor: A meta-analysis and systematic review. Oncotarget 2017, 8, 57327–57336. [Google Scholar] [CrossRef]
  176. Waickman, A.T.; Alme, A.; Senaldi, L.; Zarek, P.E.; Horton, M.; Powell, J.D. Enhancement of tumor immunotherapy by deletion of the A2A adenosine receptor. Cancer Immunol. Immunother. 2012, 61, 917–926. [Google Scholar] [CrossRef] [Green Version]
  177. Koroskenyi, K.; Kiss, B.; Szondy, Z. Adenosine A2A receptor signaling attenuates LPS-induced pro-inflammatory cytokine formation of mouse macrophages by inducing the expression of DUSP1. Biochim. Biophys. Acta 2016, 1863 Pt A, 1461–1471. [Google Scholar] [CrossRef]
  178. Cekic, C.; Linden, J. Adenosine A2A receptors intrinsically regulate CD8+ T cells in the tumor microenvironment. Cancer Res. 2014, 74, 7239–7249. [Google Scholar] [CrossRef] [Green Version]
  179. Hasko, G.; Pacher, P.; Deitch, E.A.; Vizi, E.S. Shaping of monocyte and macrophage function by adenosine receptors. Pharmacol. Ther. 2007, 113, 264–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Wehbi, V.L.; Tasken, K. Molecular Mechanisms for cAMP-Mediated Immunoregulation in T cells—Role of Anchored Protein Kinase A Signaling Units. Front. Immunol. 2016, 7, 222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Desrosiers, M.D.; Cembrola, K.M.; Fakir, M.J.; Stephens, L.A.; Jama, F.M.; Shameli, A.; Mehal, W.Z.; Santamaria, P.; Shi, Y. Adenosine deamination sustains dendritic cell activation in inflammation. J. Immunol. 2007, 179, 1884–1892. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Panther, E.; Idzko, M.; Herouy, Y.; Rheinen, H.; Gebicke-Haerter, P.J.; Mrowietz, U.; Dichmann, S.; Norgauer, J. Expression and function of adenosine receptors in human dendritic cells. FASEB J. 2001, 15, 1963–1970. [Google Scholar] [CrossRef]
  183. Mirza, A.; Basso, A.; Black, S.; Malkowski, M.; Kwee, L.; Pachter, J.A.; Lachowicz, J.E.; Wang, Y.; Liu, S. RNA interference targeting of A1 receptor-overexpressing breast carcinoma cells leads to diminished rates of cell proliferation and induction of apoptosis. Cancer Biol. Ther. 2005, 4, 1355–1360. [Google Scholar] [CrossRef] [Green Version]
  184. Lin, Z.; Yin, P.; Reierstad, S.; O’Halloran, M.; Coon, V.J.; Pearson, E.K.; Mutlu, G.M.; Bulun, S.E. Adenosine A1 receptor, a target and regulator of estrogen receptoralpha action, mediates the proliferative effects of estradiol in breast cancer. Oncogene 2010, 29, 1114–1122. [Google Scholar] [CrossRef] [Green Version]
  185. Merighi, S.; Benini, A.; Mirandola, P.; Gessi, S.; Varani, K.; Leung, E.; Maclennan, S.; Borea, P.A. A3 adenosine receptor activation inhibits cell proliferation via phosphatidylinositol 3-kinase/Akt-dependent inhibition of the extracellular signal-regulated kinase 1/2 phosphorylation in A375 human melanoma cells. J. Biol. Chem. 2005, 280, 19516–19526. [Google Scholar] [CrossRef]
  186. Merighi, S.; Mirandola, P.; Milani, D.; Varani, K.; Gessi, S.; Klotz, K.N.; Leung, E.; Baraldi, P.G.; Borea, P.A. Adenosine receptors as mediators of both cell proliferation and cell death of cultured human melanoma cells. J. Investig. Dermatol. 2002, 119, 923–933. [Google Scholar] [CrossRef] [Green Version]
  187. Kasama, H.; Sakamoto, Y.; Kasamatsu, A.; Okamoto, A.; Koyama, T.; Minakawa, Y.; Ogawara, K.; Yokoe, H.; Shiiba, M.; Tanzawa, H.; et al. Adenosine A2b receptor promotes progression of human oral cancer. BMC Cancer 2015, 15, 563. [Google Scholar] [CrossRef] [Green Version]
  188. Zhou, Y.; Chu, X.; Deng, F.; Tong, L.; Tong, G.; Yi, Y.; Liu, J.; Tang, J.; Tang, Y.; Xia, Y.; et al. The adenosine A2b receptor promotes tumor progression of bladder urothelial carcinoma by enhancing MAPK signaling pathway. Oncotarget 2017, 8, 48755–48768. [Google Scholar] [CrossRef] [Green Version]
  189. Gessi, S.; Merighi, S.; Varani, K.; Cattabriga, E.; Benini, A.; Mirandola, P.; Leung, E.; Mac Lennan, S.; Feo, C.; Baraldi, S.; et al. Adenosine receptors in colon carcinoma tissues and colon tumoral cell lines: Focus on the A(3) adenosine subtype. J. Cell. Physiol. 2007, 211, 826–836. [Google Scholar] [CrossRef]
  190. Koszalka, P.; Golunska, M.; Urban, A.; Stasilojc, G.; Stanislawowski, M.; Majewski, M.; Skladanowski, A.C.; Bigda, J. Specific Activation of A3, A2A and A1 Adenosine Receptors in CD73-Knockout Mice Affects B16F10 Melanoma Growth, Neovascularization, Angiogenesis and Macrophage Infiltration. PLoS ONE 2016, 11, e0151420. [Google Scholar] [CrossRef] [Green Version]
  191. Allen-Gipson, D.S.; Wong, J.; Spurzem, J.R.; Sisson, J.H.; Wyatt, T.A. Adenosine A2A receptors promote adenosine-stimulated wound healing in bronchial epithelial cells. Am. J. Physiol. Lung Cell Mol. Physiol. 2006, 290, L849–L855. [Google Scholar] [CrossRef]
  192. Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef] [PubMed]
  193. Kan, L.K.; Williams, D.; Drummond, K.; O’Brien, T.; Monif, M. The role of microglia and P2X7 receptors in gliomas. J. Neuroimmunol. 2019, 332, 138–146. [Google Scholar] [CrossRef] [PubMed]
  194. Adinolfi, E.; Capece, M.; Franceschini, A.; Falzoni, S.; Giuliani, A.L.; Rotondo, A.; Sarti, A.C.; Bonora, M.; Syberg, S.; Corigliano, D.; et al. Accelerated tumor progression in mice lacking the ATP receptor P2X7. Cancer Res. 2015, 75, 635–644. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. De Marchi, E.; Orioli, E.; Pegoraro, A.; Sangaletti, S.; Portararo, P.; Curti, A.; Colombo, M.P.; Di Virgilio, F.; Adinolfi, E. The P2X7 receptor modulates immune cells infiltration, ectonucleotidases expression and extracellular ATP levels in the tumor microenvironment. Oncogene 2019, 38, 3636–3650. [Google Scholar] [CrossRef]
  196. Takai, E.; Tsukimoto, M.; Harada, H.; Sawada, K.; Moriyama, Y.; Kojima, S. Autocrine regulation of TGF-beta1-induced cell migration by exocytosis of ATP and activation of P2 receptors in human lung cancer cells. J. Cell Sci. 2012, 125, 5051–5060. [Google Scholar] [CrossRef] [Green Version]
  197. Leone, R.D.; Emens, L.A. Targeting adenosine for cancer immunotherapy. J. Immunother. Cancer 2018, 6, 57. [Google Scholar] [CrossRef] [Green Version]
  198. Levy, R. SD-101 and BMS-986178 in Treating Patients with Advanced or Metastatic Solid Malignancies. 2020. Available online: https://clinicaltrials.gov/ct2/show/NCT03831295 (accessed on 9 August 2022).
  199. Terp, M.G.; Olesen, K.A.; Arnspang, E.C.; Lund, R.R.; Lagerholm, B.C.; Ditzel, H.J.; Leth-Larsen, R. Anti-human CD73 monoclonal antibody inhibits metastasis formation in human breast cancer by inducing clustering and internalization of CD73 expressed on the surface of cancer cells. J. Immunol. 2013, 191, 4165–4173. [Google Scholar] [CrossRef] [Green Version]
  200. Yegutkin, G.G.; Marttila-Ichihara, F.; Karikoski, M.; Niemela, J.; Laurila, J.P.; Elima, K.; Jalkanen, S.; Salmi, M. Altered purinergic signaling in CD73-deficient mice inhibits tumor progression. Eur. J. Immunol. 2011, 41, 1231–1241. [Google Scholar] [CrossRef]
  201. Stagg, J.; Divisekera, U.; Duret, H.; Sparwasser, T.; Teng, M.W.; Darcy, P.K.; Smyth, M.J. CD73-deficient mice have increased antitumor immunity and are resistant to experimental metastasis. Cancer Res. 2011, 71, 2892–2900. [Google Scholar] [CrossRef] [Green Version]
  202. Wang, L.; Zhou, X.; Zhou, T.; Ma, D.; Chen, S.; Zhi, X.; Yin, L.; Shao, Z.; Ou, Z.; Zhou, P. Ecto-5’-nucleotidase promotes invasion, migration and adhesion of human breast cancer cells. J. Cancer Res. Clin. Oncol. 2008, 134, 365–372. [Google Scholar] [CrossRef]
  203. Desmet, C.J.; Gallenne, T.; Prieur, A.; Reyal, F.; Visser, N.L.; Wittner, B.S.; Smit, M.A.; Geiger, T.R.; Laoukili, J.; Iskit, S.; et al. Identification of a pharmacologically tractable Fra-1/ADORA2B axis promoting breast cancer metastasis. Proc. Natl. Acad. Sci. USA 2013, 110, 5139–5144. [Google Scholar] [CrossRef] [Green Version]
  204. Zuber, V.; Jonsson, E.G.; Frei, O.; Witoelar, A.; Thompson, W.K.; Schork, A.J.; Bettella, F.; Wang, Y.; Djurovic, S.; Smeland, O.B.; et al. Identification of shared genetic variants between schizophrenia and lung cancer. Sci. Rep. 2018, 8, 674. [Google Scholar] [CrossRef] [Green Version]
  205. Sargazi, S.; Nia, M.H.; Mirinejad, S.; Moudi, M.; Shahroudi, M.J.; Saravani, R.; Valian-Borojeni, S. Association of a Novel KIF26B Gene Polymorphism with Susceptibility to Schizophrenia and Breast Cancer: A Case-Control Study. Iran. J. Public Health 2021, 50, 397–406. [Google Scholar] [CrossRef]
  206. Ibanez, K.; Boullosa, C.; Tabares-Seisdedos, R.; Baudot, A.; Valencia, A. Molecular evidence for the inverse comorbidity between central nervous system disorders and cancers detected by transcriptomic meta-analyses. PLoS Genet. 2014, 10, e1004173. [Google Scholar] [CrossRef]
Figure 1. Perturbations of the adenosine system in schizophrenia and cancer. Overview of the population and biochemical evidence for perturbed adenosine system function in schizophrenia and cancer. Reduced cancer rates in schizophrenia patients despite elevated cancer risk implies some underlying biochemical protective mechanism. Reduced extracellular adenosine availability, and hypouricemia observed in these patients may explain the reduced cancer rates. Generated in Biorender.
Figure 1. Perturbations of the adenosine system in schizophrenia and cancer. Overview of the population and biochemical evidence for perturbed adenosine system function in schizophrenia and cancer. Reduced cancer rates in schizophrenia patients despite elevated cancer risk implies some underlying biochemical protective mechanism. Reduced extracellular adenosine availability, and hypouricemia observed in these patients may explain the reduced cancer rates. Generated in Biorender.
Ijms 23 11835 g001
Figure 2. ATP and adenosine metabolism and signaling in cancer and schizophrenia. Perturbations in extracellular adenosine generating pathways are implicated in schizophrenia (green arrows) and cancer (red arrows). Differential purinergic (P1 and P2 receptors) receptor expression is reported in schizophrenia and cancer. ADA adenosine deaminase, ADK adenosine kinase, ADO adenosine, ATP adenosine triphosphate, ADP adenosine diphosphate AMP adenosine monophosphate, ENTPD ectonucleoside triphosphate diphosphohydrolases, NT5E ecto-5′nucleotidase. Generated in Biorender.
Figure 2. ATP and adenosine metabolism and signaling in cancer and schizophrenia. Perturbations in extracellular adenosine generating pathways are implicated in schizophrenia (green arrows) and cancer (red arrows). Differential purinergic (P1 and P2 receptors) receptor expression is reported in schizophrenia and cancer. ADA adenosine deaminase, ADK adenosine kinase, ADO adenosine, ATP adenosine triphosphate, ADP adenosine diphosphate AMP adenosine monophosphate, ENTPD ectonucleoside triphosphate diphosphohydrolases, NT5E ecto-5′nucleotidase. Generated in Biorender.
Ijms 23 11835 g002
Table 1. Review of Meta-analyses examining cancer rates in schizophrenia patients.
Table 1. Review of Meta-analyses examining cancer rates in schizophrenia patients.
SourceSexParticipants (N)CancersMajor Findings
[5]M/F480,356 SCZ patientsAll sites
  • Decreased overall cancer incidence (M/F)
  • No change in females alone
  • Sex and cancer type were confounding factors
[4]M/F279,938 patients across all 21
analyses
All sites
  • Colon, skin, and prostate cancer incidence decreased
  • Pooled overall cancer rates were not significantly changed prior to adjusting for smoking
  • Lung cancer rates were decreased following adjustment
  • Breast cancer incidence was increased in female SCZ patients
  • SCZ relatives’ cancer risk was decreased
[26]M/F31 studies with a median of 33,372 psychotic patientsAll sites
  • Cancer and sex specific incidence rates were reported
  • Risk ratios increased for oesophageal, breast, testicular, cervical and endometrial cancers
  • Risk ratios decreased for prostate, colon, skin, and thyroid cancers
  • Authors note better study design, controlling for confounders and appropriate comparison groups are needed
[27]M218,076 men across 13 studiesProstate
  • Decreased risk of prostate cancer across 13 cohort studies in SCZ men (SIR = 0.61).
[11]M/F496,265 SCZ patients across 12 studiesLung
  • No changes observed in lung cancer incidence
  • Authors note only one study accounted for smoking and seven studies examined lung cancer incidence only after SCZ diagnosis while the rest did not
[10]M/F312,834 SCZ patients across seven studiesLiver
  • Significantly lower liver cancer incidence was observed in SCZ males (SIR—0.71) but not in females (SIR—0.83).
  • SCZ patients have ~20% decreased risk of liver cancer
  • Authors note confounders were not adjusted for
[23]F466,244 patients across 15 studiesBreast
  • Breast cancer incidence was increased in SCZ patients.
  • Authors suggest morbidity (T2D, hyperprolactinemia, etc.) in female SCZ patients may contribute to higher breast cancer rates
M-Male, F-Female, SCZ-Schizophrenia, T2D-Type II Diabetes, SIR-Standardized Incidence Ratio.
Table 2. Nomenclature of ENTPD family enzymes.
Table 2. Nomenclature of ENTPD family enzymes.
Gene NameProtein NameAdditional Names
ENTPD1NTPDase1CD39, ATPase, ecto-apyrase
ENTPD2NTPDase2CD39L1, ecto-ATPase
ENTPD3NTPDase3CD39L3
ENTPD8NTPDase8liver canalicular ecto-ATPase
NT5EEcto-5′nucleotidase5′NT, CD73, NT5E
ENTPD/NTPDase: Ectonucleoside triphosphate diphosphohydrolase, CD39: Cluster of Differentiation 39, adapted from [54,55].
Table 3. Drugs targeting the adenosine system in clinical trials for cancer and schizophrenia.
Table 3. Drugs targeting the adenosine system in clinical trials for cancer and schizophrenia.
TargetDrug NamePhaseIndicationCase NumberFormulasCombination Therapy
NT5EBMS-986179I, IIAdvanced solid tumorsNCT02754141Monocolonal AbNivolumab, rHuPH20
CPI-006I, IbAdvanced solid tumorsNCT03454451Monocolonal AbCiforadenant, Pembrolizumab
MEDI-9447 (OLECLUMAB)I, IIAdvanced solid tumorsNCT03611556Monocolonal Abdurvalumab, gemcitabine, nab-paclitaxel, oxaliplatin, leucovorin, 5-FU
I, IIAdvanced solid tumorsNCT03381274 Osimertinib, AZD4635
I, IIAdvanced solid tumorsNCT03616886 Durvalumab, Carboplatin, Paclitaxel
IIAdvanced solid tumorsNCT03267589 Durbalumab, Tremelilumab, MEDI0562
IIBreast cancerNCT03875573 Durvalumab, Oleclumab
I, IITNBCNCT03742102 Durvalumab, Capivasertib, Oleclumab, Paclitaxel, Trastuzumab deruxtecan
IINCSLCNCT03334617 Durvalumab, AZD9150, AZD6738, Vistusertib, Olaparib, Oleclumab, Trastuzumab deruxtecan, cediranib
A2ARAZD-4635IIProstate, mCRPCNCT04089553C15H11ClFN5Oleclumab, Durvalumab
NCSLCNCT03381274 Osimertinib, MEDI9447
CPI-444I, IINSCLCNCT03337698C20H21N7O3Atezolizumab, Cobimetinib, RO6958688, Pemetrexed, Carboplatin, Linagliptin, Tocilizumab, Ipatasertib, Idasanutlin
NIR-178I, IISolid tumorsNCT03207867C10H8BrN7PDR001
I, IINCSLCNCT02403193
CaffeineN/ASchizophreniaNCT02832401C8H10N4O2N/A
PentoxifyllineISchizophreniaNCT04094207C13H18N4O3N/A
A3RCF-102CompleteHepatocellular CarcinomaNCT00790218C18H18ClIN6O4Cl-IB-MECA
IINASHNCT02927314 Placebo
I, IIHepatitis CNCT00790673 Placebo
IIHepatocellular CarcinomaNCT02128958 Placebo
mCRPC Metastatic Castration-Resistant Prostate Cancer, NASH Non-alcoholic steatohepatitis NCSLC Non-Small Cell Lung Cancer, TNBC Triple-Negative Breast Cancer.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Hamoud, A.-R.; Bach, K.; Kakrecha, O.; Henkel, N.; Wu, X.; McCullumsmith, R.E.; O’Donovan, S.M. Adenosine, Schizophrenia and Cancer: Does the Purinergic System Offer a Pathway to Treatment? Int. J. Mol. Sci. 2022, 23, 11835. https://doi.org/10.3390/ijms231911835

AMA Style

Hamoud A-R, Bach K, Kakrecha O, Henkel N, Wu X, McCullumsmith RE, O’Donovan SM. Adenosine, Schizophrenia and Cancer: Does the Purinergic System Offer a Pathway to Treatment? International Journal of Molecular Sciences. 2022; 23(19):11835. https://doi.org/10.3390/ijms231911835

Chicago/Turabian Style

Hamoud, Abdul-Rizaq, Karen Bach, Ojal Kakrecha, Nicholas Henkel, Xiaojun Wu, Robert E. McCullumsmith, and Sinead M. O’Donovan. 2022. "Adenosine, Schizophrenia and Cancer: Does the Purinergic System Offer a Pathway to Treatment?" International Journal of Molecular Sciences 23, no. 19: 11835. https://doi.org/10.3390/ijms231911835

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop