Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Apr 1.
Published in final edited form as: Hematol Oncol Clin North Am. 2014 Jan 18;28(2):287–299. doi: 10.1016/j.hoc.2013.11.003

The Role of Adenosine Signaling in Sickle Cell Therapeutics

Joshua J Field 1,2, David G Nathan 3,4,5, Joel Linden 6
PMCID: PMC3997263  NIHMSID: NIHMS558359  PMID: 24589267

Abstract

Recent data suggest a role for adenosine signaling in the pathogenesis of sickle cell disease (SCD). Signaling through the adenosine A2A receptor (A2AR) has demonstrated beneficial effects in SCD. Activation of A2ARs decreases inflammation in mice and patients with SCD largely by blocking activation of invariant NKT cells. Decreased inflammation may reduce the severity of vaso-occlusive crises. In contrast, adenosine signaling through the A2B receptor (A2BR) may be detrimental for patients with SCD. Priapism and the formation of sickle erythrocytes may be a consequence of A2BR activation on corpus cavernosal cells and erythrocytes, respectively. Whether adenosine signaling predominantly occurs through A2ARs or A2BRs may depend on differing levels of adenosine and disease state (steady state versus crisis). There may be opportunities to develop novel therapeutic approaches targeting A2ARs and/or A2BRs for patients with SCD.

Keywords: sickle cell disease, adenosine, adenosine A2A receptor, adenosine A2B receptor, NKT cells

Introduction

Sickle cell disease (SCD) is characterized by rigid, sickle-shaped erythrocytes, microvascular occlusion and tissue ischemia1. Sickle erythrocytes initiate the development of vaso-occlusion that ultimately leads to tissue ischemia in a complex multi-cellular process. Tissue ischemia promotes an inflammatory response that is further amplified by ischemia re-perfusion injury (IRI) 2. Thereafter, a vicious cycle of vaso-occlusion, tissue injury and inflammation is set into motion promoting further erythrocyte sickling1,2. The consequences of vaso-occlusion are pain, end-organ damage and often premature death3,4.

Therapies to prevent or treat sickle cell vaso-occlusion are limited. Broadly, treatments either prevent the formation of sickle erythrocytes (e.g., hydroxyurea) or interrupt the cellular interactions that follow red cell sickling and lead to vaso-occlusion5. Hydroxyurea is an anti-mitotic agent that disrupts the polymerization of sickle hemoglobin and, to date, is the only FDA-approved therapy for the prevention of painful vaso-occlusive crises (pVOC)6. Patient and provider barriers have limited the widespread use of hydroxyurea in patients with SCD, affecting the impact of the drug7. Hematopoietic stem cell transplantation is a potential cure; however, it is only an option for a few patients with SCD8.

In this review, we examine the role of adenosine signaling in SCD pathogenesis. There are opportunities to modulate adenosine pathways using therapies to prevent or treat SCD complications. As evidence has emerged about the importance of adenosine in SCD, separate lines of investigation have demonstrated protective and detrimental effects of adenosine in regards to disease severity. Recent data suggest that actions of adenosine mediated through the adenosine A2A receptor (A2AR) decrease inflammation, largely by selectively inhibiting the activation of a subset of lymphocytes called invariant NKT (iNKT) cells911. In contrast, other studies have shown that adenosine signaling through the adenosine A2B receptor (A2BR) may contribute to the adverse processes of erythrocyte sickling12 and priapism1315. Although much additional work is needed to fully elucidate the roles of adenosine signaling in SCD, targeting these pathways may produce novel therapeutic approaches.

Adenosine signaling pathway

Adenosine physiology

Adenosine signaling protects tissues by promoting vaso-dilation as well as decreasing heart rate and inflammation16. During periods of cellular hypoxia or stress, adenosine is released from cells along with the adenine nucleotides, ATP, ADP and AMP, which are converted to adenosine by ecto-nucleotidases. Binding of adenosine to four receptor subtypes, A1, A2A, A2B, or A3, elicits responses that are dependent upon the receptor subtypes found in various tissues (Table 1). Adenosine receptors are 7-transmembrane, G-coupled receptors that signal through adenylyl cyclase, affecting the production of cyclic AMP (cAMP), calcium, or the conductance of ion channels. A1 and A3 receptors couple to inhibitory G receptors (Gi) and decrease adenylyl cyclase activity, whereas A2A and A2B increase adenylyl cyclase activity by coupling to stimulatory G receptors (Gs or Go)16. The affinity for adenosine also differs among the receptor subtypes, affecting the concentration of adenosine necessary for activation. A1 and A2A are high affinity receptors, activating at lower concentrations of adenosine (~0.01 μM to 1 μM). A2B is a low affinity receptor requiring 10 to 1000-fold higher levels of adenosine (~10 μM) for activation17. Downstream from adenylyl cyclase and cAMP, adenosine signaling modifies the activity of nuclear factor kappa-B (NF-κB), JAK-STAT and ERK pathways, regulating transcription and ultimately cellular functions18. Adenosine that accumulates during cellular stress is removed by uptake into cells and converted to AMP or inosine by adenosine kinase and adenosine deaminase (ADA), respectively. In patients with SCD, tissue injury may increase levels of plasma adenosine suggesting that adenosine pathways may influence SCD pathogenesis12.

Table 1.

Adenosine receptor subtypes17,32

Adenosine receptor subtypes
Characteristics A1 A2A A2B A3
Predominant tissue/cell expression
  • Lung

  • Heart

  • Brain

  • Leukocytes

  • Lung

  • Brain

  • Vasculature

  • Leukocytes

  • Lung

  • Colon

  • Vasculature

  • Leukocytes

  • Erythrocytes

  • Penis

  • Lung

  • Liver

  • Heart

  • Brain

  • Leukocytes

Actions
  • Negative chronotropic

  • ↑inflammation

  • Vasodilation

  • ↓inflammation

  • ↑inflammation in lung and colon

  • ↓inflammation

Affinity for adenosine High High Low High
Major disease associations
  • Sepsis

  • SCD

  • Ischemia

  • Arthritis

  • Wound healing

  • SCD

  • Asthma

  • COPD

  • IBD

  • Asthma

  • Arthritis

Current therapeutic uses of adenosine and adenosine derivatives

Drugs that target adenosine receptors are part of current standard practice. Adenosine, dipyridamole and adenosine A2AR agonists (e.g., regadenoson) are used clinically to induce cardiac hyperemia during myocardial stress testing via activation of coronary artery A2ARs. Adenosine-mediated activation of A1 receptors in the heart is a treatment for tachyarrhythmias. Theophylline, either alone or in complex with ethylenediame (aminophylline), non-selectively blocks A1, A2A and A2B receptors and is used as a therapy for asthma16. A limitation of these therapies is lack of selectivity for the adenosine receptor subtypes, sometimes resulting in unwanted and potentially dangerous side effects. These include hypotension and tachycardia from A2AR activation, bradycardia or heart-block from A1 activation and bronchospasm from A2BR activation in patients with asthma. Newer generation adenosine agonists and antagonists have greater receptor subtype selectivity thereby minimizing toxicities16. There is emerging cellular, animal and human data suggesting a role for the A2A and A2B receptors in the pathogenesis of SCD (Table 2). Adenosine-based therapies are currently being examined in patients with SCD (Clinicaltrials.gov #01788631).

Table 2.

Comparison of A2AR and A2BR data in sickle cell disease

Reference Type of study Transgenic
SCD model
# SCD
Patients
Agents
evaluated
Key findings
A2AR
  VOC
  Wallace et al. Blood 201010 Investigation of transgenic SCD mice NY1DD - A2AR agonist ATL146e
  • SCD causes induction of A2AR on iNKT cells

  • A2AR agonist ATL146e reverses pulmonary dysfunction in SCD mice

  Field et al. Blood 201311 Phase 1 clinical study - 27 A2AR agonist regadenoson
  • A2AR agonist regadenoson decreases activation of iNKT cells in patients with SCD during VOC

  • A2AR agonist regadenoson is safe in patients with SCD

  Lin et al. PLosOne 201319 Investigation of transgenic SCD mice NY1DD 8 -
  • SCD causes induction of A2AR on iNKT cells

  • A2AR expression on iNKT cells is mediated through NF-κB

A2BR
  Red blood cell sickling
  Zhang et al. Nat Med 201112 Investigation of transgenic SCD mice, cultured human RBCs and SCD patient blood samples Berkley 12 PEG-ADA, theophylline, A2BR antagonist MRS1754
  • Adenosine is elevated in the plasma of mice and patients with SCD

  • A2BR on erythrocytes mediates sickling via decreasing hemoglobin oxygen binding affinity

  Priapism
  Mi et al. JCI 200815 Investigation of ADA deficient and transgenic SCD mice Berkley - PEG-ADA, theophylline, A2BR antagonist MRS1706
  • Priapism is present in ADA−/− mice and corrected by PEG-ADA

  • Interruption of A2BR activity on corpus cavernosal cells decreases priapism

  Wen et al. J Sex Med 201014 Investigation of ADA deficient and transgenic SCD mice Berkley - PEG-ADA
  • PEG-ADA therapy prevented episodes of priapism in SCD mice

  Wen et al. FASEB 201013 Investigation of ADA deficient and transgenic SCD mice Berkley - PEG-ADA, A2BR antagonist MRS1706
  • PEG-ADA therapy prevented penile fibrosis in SCD mice

  • Interruption of A2BR in corpus cavernosal fibroblast cells prevents penile fibrosis

Role of A2AR in sickle cell disease

A2AR

A2AR activation is well-known for producing vaso-dilation due to effects on vascular smooth muscle and some endothelial cells. In addition, A2AR has a central role in the regulation of inflammation and immunity17. Ubiquitously expressed on neutrophils, monocytes, macrophages, T cells, NK cells and iNKT cells, adenosine signaling through the A2AR has been shown to suppress key inflammatory and immune responses, including leukocyte activation, recruitment and cytokine production18. These immune suppressive effects of A2AR activation are mediated by cAMP and protein kinase A (PKA)18. PKA signaling can inhibit other signaling pathways that activate inflammation mediated by NF-κB or the JAK-STAT pathway and serves to decrease transcription of key inflammatory genes19. The NF-κB pathway deserves special attention as it has been used as a marker of iNKT cell activity in clinical trials of the A2AR agonist, regadenoson, in patients with SCD11.

NF-κB is a critically important transcription factor that generally enhances inflammation20. Comprised of a dimer of transcription factors from the RelA family of proteins (p50 or p52 and p65), NF-κB resides in the cytoplasm of cells bound to the inhibitory protein IκB. Upon activation of the NF-κB by numerous inflammatory mediators including tumor necrosis factor-α or interleukin-1, IκB is phosphorylated by IκB kinase, ubiquinated and degraded, thus liberating NF-κB to translocate into the nucleus and promote the transcription of pro-inflammatory genes. When NF-κB is released from IκB, the 65 kDa subunit (p65) can be phosphorylated on several sites including Ser536. The phosphorylation of p65 (phospho-p65) serves as a marker of NF-κB activity used in flow cytometry assays11. In the case of A2AR activation, in vitro data suggest that agonists of the A2AR reduce IκB degradation, decreasing the ability of NF-κB to promote a pro-inflammatory cellular response21. NF-κB has also been shown to mediate the up-regulation of A2AR in iNKT cells following activation19.

A2AR agonist decreases inflammation following ischemia-reperfusion injury by interfering with iNKT cell activation

Murine models of liver and kidney transplant demonstrated that activation of A2ARs by adenosine analogues administered during or after IRI markedly inhibit inflammation and secondary injury22. An investigation of the cell type primarily responsible for the protective effect of A2AR activation implicated the iNKT cell as the primary target22. Although iNKT cells normally constitute < 1% of the lymphocyte population, iNKT cells can rapidly release large amounts of pro-inflammatory cytokines giving them a critical role in inflammation, despite representing a only small proportion of lymphocytes23. Similar to B and T cells that produce adaptive immune responses, iNKT cell activation requires the engagement of an antigen presented on an antigen presenting cell24. Unlike B and T cells, which undergo genetic recombination to generate diverse receptors that recognize various peptides, iNKT cells express a semi-invariant T cell receptor that non-specifically binds to lipid antigens presented on CD1d, a MHC class I-like molecule. Different lipids (glycolipids, phospholipids) have been shown to activate iNKT cells24,25. The activation of iNKT cells is enhanced by cytokines produced by antigen presenting cells in response to Toll-like-receptor activation26. Thus, the activation of iNKT cells is facilitated by innate immune responses stimulated by pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs). Upon CD1d-restricted activation, iNKT cells rapidly make mRNAs and release large quantities of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2) and interleukin-4 (IL-4)27. IFN-γ stimulates the production in many cells of interferon-inducible CXCR3 chemokines, CXCL9, CXCL10 and CXCL1128. IL-2 is known to induce CXCR3 receptors on lymphocytes28. Thus, through rapid activation and generation of copious amounts of cytokines and chemokines, iNKT cells stimulate a pro-inflammatory cascade that may promote and sustain vaso-occlusion9,10. Activation of A2ARs, abundantly expressed on activated iNKT cells, reduces this inflammatory response and are critical to modulating the immune functions of iNKT cells10,11.

A2AR agonists decrease iNKT cell activation and reduce inflammation in SCD mice

In a series of experiments in an NY1DD mouse model of SCD, Linden and colleagues generated several lines of evidence implicating iNKT cells as critical to the process of sickle cell vaso-occlusion9,10. Lung inflammation and injury were reduced when: 1) iNKT cells were antibody depleted or genetically knocked out, 2) activation or chemotaxis was inhibited and 3) upon administration of A2AR agonists9. NY1DD mice treated with a continuous subcutaneous infusion of the A2AR agonist ATL146e demonstrated a maximal improvement in lung function, histology and inflammatory cell infiltrate in 3 days at an infusion rate of 10 ng/kg/minute. The improvement was sustained up to the end of infusion at 7 days10. The infused dose of ATL146e only achieved plasma concentrations about 1 nM, and the mice did not demonstrate cardiovascular toxicities10. The absence of toxicity is in accord with prior studies demonstrating that the anti-inflammatory effects of A2AR agonists occur at 10–100 fold lower concentrations compared to the cardiovascular effects22. Blockade or depletion of iNKT cells mitigated the beneficial effects of the A2AR agonist, providing evidence that the anti-inflammatory actions of A2AR activation are mediated largely through iNKT cells9,10.

In the plasma of adult patients with SCD, circulating iNKT cells were also more likely to be activated and expanded compared to healthy controls9. There is selective expansion of iNKT cells among lymphocytes, from < 1% in control blood to an average of about 5% in the blood of SCD patients whose iNKT cells were also more like to express the activation markers CD69, intracellular IFN-γ, and CXCR39.

Phase 1 study of the A2AR agonist regadenoson in patients with SCD: study design and rationale

Based on the promising data from mice and patients with SCD suggesting that A2AR agonists may interrupt activation of iNKT cells and potentially decrease sickle cell complications, a phase 1 trial was conducted of the A2AR agonist regadenoson29. FDA-approved for inducing cardiac hyperemia during myocardial imaging, regadenoson is a selective A2AR agonist with 10-fold greater affinity for A2A versus A1 and few if any effects on A2B or A3 30,31. When used for myocardial imaging regadenoson is administered as a 400 μg bolus over 10 seconds30. Bolus injection induces vaso-dilation and hyperemia in a time frame appropriate for capturing images before blood flow reverts to normal31. If the goal of administering regadenoson is to dampen the severity of a pVOC over several days, a continuous infusion would be necessary given its terminal half-life of 2 hours. When designing the study, three relatively low doses of regadenoson were selected based on data extrapolated from animal models29. All of these doses produced the desired anti-inflammatory effects while avoiding cardiovascular toxicities29. Using a traditional 3+3 study design, the dose levels were examined during a 12 hour infusion of regadenoson while patients with SCD were at steady state29. Once the highest dose of infusional regadenoson (1.44 mcg/kg/hour) was found to be safe, SCD subjects were examined during a 24 or 48 hour infusion at steady state and then during a pVOC29.

To evaluate the effects of regadenoson on iNKT cell activation, various activation markers were examined. Phosphorylation of the p65 subunit of NF-κB (phospho-p65 NF-κB) was identified as the most promising marker of iNKT cell activation because, as opposed to cell surface markers or cytokines, changes in phosphorylation are pre-transcriptional and thus occur quickly.

Phase 1 study of the A2AR agonist regadenoson in patients with SCD: study results

Twenty-seven adult patients with SCD were administered regadenoson, 21 at steady state and 6 during pVOC11. Circulating iNKT cells from adults with SCD during pVOC showed increased phospho-p65 NF-κB activation compared to steady state or healthy controls. When adults with SCD were administered a 24 hour infusion of the A2AR agonist regadenoson during pVOC, the percentage of iNKT cells expressing increased phospho-p65 NF-κB decreased to levels similar to steady state patients and healthy controls. The effects of regadenoson were achieved at plasma concentrations that peaked at 2 ng/ml and were devoid of effects on heart rate or blood pressure. A large, randomized-controlled, phase 2 trial is in progress to evaluate the clinical efficacy of regadenoson during pVOC (Clinicaltrials.gov #01788631).

Role of A2BR in sickle cell disease

A2BR

The A2BR is a lower affinity receptor that has well-described pro-inflammatory roles in the pathogenesis of asthma, chronic obstructive pulmonary disease and inflammatory bowel disease32. Higher levels of adenosine are necessary to activate the A2BR (10–100 fold > A2AR) and therefore signaling through A2BR occurs selectively in stressed cells when adenosine is generated by ischemia, injury, or inflammation17. Although the distribution of the A2BR is widespread on cells and tissues including smooth muscle cells, endothelial cells and macrophages, pathogenic inflammation is most notably promoted by the actions of the A2BR on mast cells and intestinal epithelial cells32. Recent data suggest that activation of A2BR on mast cells results in the production of cytokines with important roles in asthma pathogenesis, such as IL-4 and IL-13, and stimulates B cells to produce IgE33. The role of A2BR in asthma is also supported by data demonstrating increased inflammation in asthma patients after inhalation of adenosine34,35. To this end, the non-selective adenosine receptor antagonist, theophylline, has a long-standing role in the management of asthma in part working through blockade of the A2BR36; although, its lack of selectivity is associated with side effects. A2BR expression on intestinal epithelial cells is also important in disease pathogenesis promoting IL-6 production and resulting in intestinal inflammation, potentially contributing to the process of inflammatory bowel disease37. More recently, A2BR activation in the corpus cavernosum of the penis and erythrocytes has been shown to promote priapism1315 and red cell sickling12, respectively, in a murine model of SCD.

Adenosine signaling through A2BR is implicated in priapism and penile fibrosis

Studies in non-SCD mouse models demonstrating that intracavernosal injections of adenosine provoked priapism provided preliminary evidence for a role for adenosine signaling in the pathogenesis of priapism38,39. More recent work has found that the actions of adenosine are mediated through A2BR signaling on corpus cavernosum smooth muscle cells within the penis40. In a series of experiments, investigators determined that ADA-deficient mice had higher intra-penile adenosine levels leading to cavernosal smooth muscle relaxation and priapism15. Administration of ADA or the A2BR antagonists, theophylline or MRS1706, antagonized the effects of excess adenosine and reversed priapism14,15. ADA−/−/A2BR−/− mice were also protected from priapic episodes15. Many of the same findings were recapitulated in a transgenic SCD mouse model. SCD mice had higher plasma levels of adenosine than controls and similarly their episodes of priapism were counteracted by decreasing adenosine levels with ADA or administration of an A2BR antagonist15. In SCD mice, there is also evidence that adenosine signaling through the A2BR contributes to penile fibrosis, a consequence of priapism episodes that contributes to erectile dysfunction13. Taken together, these data suggest that interruption of adenosine signaling through A2BR may have a role in the treatment of priapism and prevention of penile fibrosis in SCD.

Sickle erythrocyte formation promoted through A2BR

Recently, another detrimental effect of A2BR activation was described when Zhang and colleagues reported a novel mechanism of erythrocyte sickling in transgenic SCD mice caused by high plasma adenosine and activation of A2BRs expressed on red cells12. These investigators first discovered that in transgenic SCD mice a reduction in circulating adenosine levels following pegylated (PEG)-ADA administration was associated with a decreased number of sickle erythrocytes. To identify the adenosine receptor subtype responsible for promoting sickle cell formation, red cells from mice genetically deficient in one of the four receptor subtypes were activated with an adenosine analogue. The deleterious effects of adenosine were found to be mediated through the A2BR due to an increase in intra-erythrocyte 2,3-DPG levels and a decreased hemoglobin-oxygen affinity. The resulting increased formation of deoxy-hemoglobin provides an explanation for the increased erythrocyte sickling. Translating these findings to humans, levels of plasma adenosine were found to be higher in patients with SCD versus healthy controls as were intra-erythrocyte 2,3-DPG levels (the latter more likely due to the effects of anemia). In vitro treatment of erythrocytes from patients with SCD with PEG-ADA or A2BR antagonists reduced red cell sickling, consistent with the findings from the murine model.

Can adenosine have both protective and deleterious roles in SCD?

Recent data describing the role of adenosine signaling in SCD suggest discrepant effects on morbidity when the actions of adenosine are medicated through the A2AR versus A2BR41. On one hand, data show that activation of A2AR decreases inflammation, largely through the inhibition of iNKT cell activation, potentially dampening the severity of pVOC10,11. On the other hand, a separate line of research suggests that adenosine signaling through A2BR promotes priapism1315 and erythrocyte sickling12. Although further studies are needed to fully understand the seemingly conflicting roles of adenosine signaling in SCD, differences in adenosine receptor expression and affinity along with a better understanding of in vivo levels of adenosine in patients with SCD may help to reconcile the confusion.

Effects of adenosine levels and receptor density on A2AR versus A2BR signaling in SCD

Under steady-state conditions and potentially during pVOC, levels of adenosine may be sufficient to signal through the A2AR, yet not high enough to trigger A2BR signaling. In comparison to A2BR, affinity of adenosine for A2AR is 10 to 1000 times greater than A2BR17. Moreover, the effects of adenosine are further potentiated in cells or tissue densely expressing adenosine receptors, as is the case for A2AR expression on activated iNKT cells. Compared to CD4+ T cells, iNKT cells express 10 times more A2AR and receptor expression increases further (100-fold) during pVOC10,19. Thus, iNKT cells are exquisitely sensitive to inhibition through adenosine signaling due to dense expression of the high affinity receptor, A2AR. Based on receptor affinity and density, a model for adenosine signaling emerges whereby differing levels of adenosine in a localized area may produce actions through A2AR and/or A2BR. Potentially, under conditions where levels of adenosine in SCD are not extremely high, the effects of adenosine are mediated through the A2AR and only during the more extreme conditions of pVOC would the deleterious effects of the A2BR activation be observed (Figure 1).

Figure 1. The proposed roles of the adenosine A2A receptor (A2AR) and the adenosine A2B receptor (A2BR) in the pathogenesis of sickle cell disease (SCD).

Figure 1

Sickle cell vaso-occlusion leads to tissue injury and liberation of adenosine. Left side of schema: expression of A2AR are increased on iNKT cells and activation of A2AR on iNKT cells leads to a reduction in pro-inflammatory mediators (IFN-γ, IL-4) and dampening in the severity of vaso-occlusion. A2AR agonists (e.g., regadenoson) may promote the anti-inflammatory effects of the A2AR. Right side of schema: Activation of A2BR on erythrocytes results in increased levels of 2,3 DPG decreasing hemoglobin affinity for oxygen and contributing to red cell sickling. On corpus cavernosal cells of the penis, activation of A2BR promotes priapism. Antagonists to A2BR may decrease the formation of sickle erythrocytes and prevent priapic episodes. Potentially, dual therapy with A2AR agonist/A2BR antagonist would be an ideal for patients with SCD: anti-inflammatory, anti-sickling, anti-priapism.

Adenosine measurements have limitations

Levels of adenosine in patients with SCD have been shown to be higher than healthy controls and, likely, levels will rise further during pVOC12. Adenosine measurements in the extracellular space reflect the sum of adenosine’s formation, transport and degradation42. During periods of microvascular occlusion and tissue ischemia, extracellular concentrations of adenosine may increase and activate adenosine signaling pathways to counteract further tissue damage by increasing blood flow and reducing heart rate and inflammation. Unfortunately, rapid cellular uptake and degradation cause adenosine to have a half life of approximately 5 seconds creating challenges to obtaining accurate measurements42. Further compounding errors in measurement is the compartmentalization of adenosine between the intravascular and interstitial spaces and the restricted, tissue-specific rise that may occur in adenosine levels following ischemia and injury42. Thus, measurement of adenosine levels in blood from the right arm may not accurately reflect the physiologic effects of adenosine in a patient who is experiencing ongoing vaso-occlusion in the vasculature of the left leg. A final consideration in the interpretation of adenosine levels in the preceding studies are the differences in adenosine biology between people and mice. Adenosine’s half-life is longer in mice than in people, and therefore blood levels are also higher in mice41. Under these conditions, adenosine signaling through A2BR may be more pronounced in mouse models than in patients. A better understanding of adenosine levels in patients with SCD may clarify the role of A2AR and A2BR signaling under varied conditions and the contribution of this signaling to SCD morbidities.

Limitations of adenosine therapeutics in SCD

The main challenges for using adenosine analogues to treat and prevent vaso-occlusion are the need for a continuous intravenous infusion and unwanted side effects. The short half-life of regadenoson necessitates a continuous infusion, which requires stable intravenous access, and limits the role of regadenoson to the treatment of acute crises. Regadenoson has multi-phasic pharmacokinetics, but the terminal half-life is still only 2 hours30. Current conceptual models of SCD describe ongoing vaso-occlusion with inflammation and end organ damage that is punctuated by severe pVOC that result in a hospitalization. Treatments aimed to shorten the duration of hospitalizations, such as regadenoson, have value as there is a higher risk of death during these acute events4, however, they will not affect the continuing daily damage that culminates in organ dysfunction. Preventing a major crisis is a better strategy to decease morbidity and mortality than treating one. To this end, a humanized monoclonal antibody that targets iNKT cells independently of the adenosine signaling pathways is currently under investigation in a phase 1 study (Clinicaltrials.gov #01783691). This investigational agent could potentially deplete iNKT cells on a longer-term basis than the effects of regadenoson and prevent crises. Another challenge to the use of adenosine signaling in the treatment of SCD is unwanted side effects due to a lack of adenosine receptor selectivity. Adenosine and dipyridamole cause activation of all adenosine receptor subtypes and are associated with severe side effects such as heart-block and hypotension. The A2AR agonist regadenoson has been administered to patients with SCD without toxicity due to a high degree of selectivity for the A2AR, along with the fact that lower concentrations of the drug are still able to achieve anti-inflammatory effects11.

Future directions: combined A2AR and A2BR therapies for SCD?

Modulating the adenosine signaling pathway holds promise as a treatment for patients with SCD. Independent lines of investigation have provided evidence that the A2AR and A2BR are respectively protective and deleterious in the pathogenesis of vaso-occlusion. The combination of an A2AR agonist with an A2BR antagonist may be an ideal treatment in SCD. Thus far, the A2AR agonist regadenoson is the only adenosine-based therapeutic that has been studied in patients with SCD11. Another approach that has been suggested is PEG-ADA infusions to lower circulating adenosine levels and minimize signaling through the A2BR. ADA is an FDA-approved therapy for patients with ADA deficiency43. The possible shortcoming of this approach is that positive effects from adenosine signaling through the A2AR might be negated with ADA therapy. A more novel and potentially effective approach to prevent and treat vaso-occlusion is dual therapy with A2BR antagonists to prevent erythrocyte sickling and A2AR agonists to decrease inflammation and dampen the severity of pVOC (Figure 1)44. Highly selective A2BR antagonists are currently in development for maintenance treatment of asthma16,44. Clearly, rigorously-designed clinical trials demonstrating the clinical efficacy of A2AR agonists and/or A2BR antagonists need to be conducted prior to considering combination therapy, however, innovative approaches are needed in the treatment of SCD and multi-modal therapies may be necessary to demonstrate clinical benefit.

Key points.

  • Activation of adenosine A2A receptor (A2AR) on invariant NKT cells decreases inflammation in a transgenic mouse model of sickle cell disease (SCD). The effects of regadenoson, an A2AR agonist, are currently being examined in patients with SCD.

  • The adenosine A2B receptor (A2BR) on red blood cells and corpus cavernosal cells of the penis has been implicated in the formation of sickle erythrocytes and priapism, respectively.

  • These two independent lines of research examining the roles of A2AR and A2BR signaling in SCD may provide opportunities for new therapies.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosure: JJF and DGN were consultants for NKT Therapeutics

References

  • 1.Hebbel RP, Osarogiagbon R, Kaul D. The endothelial biology of sickle cell disease: inflammation and a chronic vasculopathy. Microcirculation. 2004;11:129–151. [PubMed] [Google Scholar]
  • 2.Kaul DK, Hebbel RP. Hypoxia/reoxygenation causes inflammatory response in transgenic sickle mice but not in normal mice. J Clin Invest. 2000;106:411–420. doi: 10.1172/JCI9225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Platt OS, Thorington BD, Brambilla DJ, et al. Pain in sickle cell disease. Rates and risk factors. N Engl J Med. 1991;325:11–16. doi: 10.1056/NEJM199107043250103. [DOI] [PubMed] [Google Scholar]
  • 4.Platt OS, Brambilla DJ, Rosse WF, et al. Mortality in sickle cell disease. Life expectancy and risk factors for early death. N Engl J Med. 1994;330:1639–1644. doi: 10.1056/NEJM199406093302303. [DOI] [PubMed] [Google Scholar]
  • 5.Vichinsky E. Emerging 'A' therapies in hemoglobinopathies: agonists, antagonists, antioxidants, and arginine. Hematology Am Soc Hematol Educ Program. 2012:271–275. doi: 10.1182/asheducation-2012.1.271. [DOI] [PubMed] [Google Scholar]
  • 6.Ware RE, Aygun B. Advances in the use of hydroxyurea. Hematology Am Soc Hematol Educ Program. 2009:62–69. doi: 10.1182/asheducation-2009.1.62. [DOI] [PubMed] [Google Scholar]
  • 7.Lanzkron S, Haywood C, Jr, Hassell KL, Rand C. Provider barriers to hydroxyurea use in adults with sickle cell disease: a survey of the Sickle Cell Disease Adult Provider Network. J Natl Med Assoc. 2008;100:968–973. [PubMed] [Google Scholar]
  • 8.Dew A, Collins D, Artz A, et al. Paucity of HLA-identical unrelated donors for African-Americans with hematologic malignancies: the need for new donor options. Biol Blood Marrow Transplant. 2008;14:938–941. doi: 10.1016/j.bbmt.2008.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Wallace KL, Marshall MA, Ramos SI, et al. NKT cells mediate pulmonary inflammation and dysfunction in murine sickle cell disease through production of IFN-gamma and CXCR3 chemokines. Blood. 2009;114:667–676. doi: 10.1182/blood-2009-02-205492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wallace KL, Linden J. Adenosine A2A receptors induced on iNKT and NK cells reduce pulmonary inflammation and injury in mice with sickle cell disease. Blood. 2010;116:5010–5020. doi: 10.1182/blood-2010-06-290643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Field JJ, Lin G, Okam MM, et al. Sickle cell vaso-occlusion causes activation of iNKT cells that is decreased by the adenosine A2A receptor agonist regadenoson. Blood. 2013;121:3329–3334. doi: 10.1182/blood-2012-11-465963. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zhang Y, Dai Y, Wen J, et al. Detrimental effects of adenosine signaling in sickle cell disease. Nat Med. 2011;17:79–86. doi: 10.1038/nm.2280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Wen J, Jiang X, Dai Y, et al. Increased adenosine contributes to penile fibrosis, a dangerous feature of priapism, via A2B adenosine receptor signaling. FASEB J. 2010;24:740–749. doi: 10.1096/fj.09-144147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wen J, Jiang X, Dai Y, et al. Adenosine deaminase enzyme therapy prevents and reverses the heightened cavernosal relaxation in priapism. J Sex Med. 2010;7:3011–3022. doi: 10.1111/j.1743-6109.2009.01552.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Mi T, Abbasi S, Zhang H, et al. Excess adenosine in murine penile erectile tissues contributes to priapism via A2B adenosine receptor signaling. J Clin Invest. 2008;118:1491–1501. doi: 10.1172/JCI33467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Jacobson KA, Gao ZG. Adenosine receptors as therapeutic targets. Nat Rev Drug Discov. 2006;5:247–264. doi: 10.1038/nrd1983. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hasko G, Linden J, Cronstein B, Pacher P. Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov. 2008;7:759–770. doi: 10.1038/nrd2638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Milne GR, Palmer TM. Anti-inflammatory and immunosuppressive effects of the A2A adenosine receptor. ScientificWorldJournal. 2011;11:320–339. doi: 10.1100/tsw.2011.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Lin G, Field JJ, Yu JC, Ken R, Neuberg D, Nathan DG, Linden J. NF-kB is activated in CD4+ iNKT cells by sickle cell disease and mediates rapid induction of adenosine A2A receptors. PLoS One. 2013 doi: 10.1371/journal.pone.0074664. accepted. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Karin M, Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF-κB activity. Annu Rev Immunol. 2000;18:621–663. doi: 10.1146/annurev.immunol.18.1.621. [DOI] [PubMed] [Google Scholar]
  • 21.Lukashev D, Ohta A, Apasov S, Chen JF, Sitkovsky M. Cutting edge: Physiologic attenuation of proinflammatory transcription by the Gs protein-coupled A2A adenosine receptor in vivo. J Immunol. 2004;173:21–24. doi: 10.4049/jimmunol.173.1.21. [DOI] [PubMed] [Google Scholar]
  • 22.Lappas CM, Day YJ, Marshall MA, Engelhard VH, Linden J. Adenosine A2A receptor activation reduces hepatic ischemia reperfusion injury by inhibiting CD1d-dependent NKT cell activation. J Exp Med. 2006;203:2639–2648. doi: 10.1084/jem.20061097. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Matsuda JL, Mallevaey T, Scott-Browne J, Gapin L. CD1d-restricted iNKT cells, the 'Swiss-Army knife' of the immune system. Curr Opin Immunol. 2008;20:358–368. doi: 10.1016/j.coi.2008.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Van Kaer L, Parekh VV, Wu L. Invariant natural killer T cells: bridging innate and adaptive immunity. Cell Tissue Res. 2011;343:43–55. doi: 10.1007/s00441-010-1023-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Gumperz JE, Roy C, Makowska A, et al. Murine CD1d-restricted T cell recognition of cellular lipids. Immunity. 2000;12:211–221. doi: 10.1016/s1074-7613(00)80174-0. [DOI] [PubMed] [Google Scholar]
  • 26.Salio M, Speak AO, Shepherd D, et al. Modulation of human natural killer T cell ligands on TLR-mediated antigen-presenting cell activation. Proc Natl Acad Sci USA. 2007;104:20490–20495. doi: 10.1073/pnas.0710145104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kronenberg M, Gapin L. The unconventional lifestyle of NKT cells. Nat Rev Immunol. 2002;2:557–568. doi: 10.1038/nri854. [DOI] [PubMed] [Google Scholar]
  • 28.Singh UP, Venkataraman C, Singh R, Lillard JW., Jr CXCR3 axis: role in inflammatory bowel disease and its therapeutic implication. Endocr Metab Immune Disord Drug Targets. 2007;7:111–123. doi: 10.2174/187153007780832109. [DOI] [PubMed] [Google Scholar]
  • 29.Field JJ, Nathan DG, Linden J. Targeting iNKT cells for the treatment of sickle cell disease. Clin Immunol. 2011;140:177–183. doi: 10.1016/j.clim.2011.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Astellas Pharma. Lexiscan [package insert] Northbrook, IL: Astellas Pharma; 2009. [Google Scholar]
  • 31.Buhr C, Gossl M, Erbel R, Eggebrecht H. Regadenoson in the detection of coronary artery disease. Vasc Health Risk Manag. 2008;4:337–340. doi: 10.2147/vhrm.s1798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hasko GC, B Nemeth ZH, Vizi ES, Pacher P. A2B adenosine receptors in immunity and inflammation. Trends Immunology. 2009;30:263–270. doi: 10.1016/j.it.2009.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ryzhov S, Goldstein AE, Matafonov A, Zeng D, Biaggioni I, Feoktistov I. Adenosine-activated mast cells induce IgE synthesis by B lymphocytes: an A2B-mediated process involving Th2 cytokines IL-4 and IL-13 with implications for asthma. J Immunol. 2004;172:7726–7733. doi: 10.4049/jimmunol.172.12.7726. [DOI] [PubMed] [Google Scholar]
  • 34.Cushley MJ, Tattersfield AE, Holgate ST. Inhaled adenosine and guanosine on airway resistance in normal and asthmatic subjects. Br J Clin Pharmacol. 1983;15:161–165. doi: 10.1111/j.1365-2125.1983.tb01481.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Holgate ST. The Quintiles Prize Lecture 2004. The identification of the adenosine A2B receptor as a novel therapeutic target in asthma. Br J Pharmacol. 2005;145:1009–1015. doi: 10.1038/sj.bjp.0706272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Wilson CN. Adenosine receptors and asthma in humans. Br J Pharmacol. 2008;155:475–486. doi: 10.1038/bjp.2008.361. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kolachala VL, Bajaj R, Chalasani M, Sitaraman SV. Purinergic receptors in gastrointestinal inflammation. Am J Physiol Gastrointest Liver Physiol. 2008;294:G401–G410. doi: 10.1152/ajpgi.00454.2007. [DOI] [PubMed] [Google Scholar]
  • 38.Lin CS, Lin G, Lue TF. Cyclic nucleotide signaling in cavernous smooth muscle. J Sex Med. 2005;2:478–491. doi: 10.1111/j.1743-6109.2005.00080.x. [DOI] [PubMed] [Google Scholar]
  • 39.Noto T, Inoue H, Mochida H, Kikkawa K. Role of adenosine and P2 receptors in the penile tumescence in anesthetized dogs. Eur J Pharmacol. 2001;425:51–55. doi: 10.1016/s0014-2999(01)01167-0. [DOI] [PubMed] [Google Scholar]
  • 40.Faria M, Magalhaes-Cardoso T, Lafuente-de-Carvalho JM, Correia-de-Sa P. Corpus cavernosum from men with vasculogenic impotence is partially resistant to adenosine relaxation due to endothelial A2B receptor dysfunction. J Pharmacol Exp Ther. 2006;319:405–413. doi: 10.1124/jpet.106.107821. [DOI] [PubMed] [Google Scholar]
  • 41.Gladwin MT. Adenosine receptor crossroads in sickle cell disease. Nat Med. 2011;17:38–40. doi: 10.1038/nm0111-38. [DOI] [PubMed] [Google Scholar]
  • 42.Ramakers BP, Pickkers P, Deussen A, et al. Measurement of the endogenous adenosine concentration in humans in vivo: methodological considerations. Curr Drug Metab. 2008;9:679–685. doi: 10.2174/138920008786049249. [DOI] [PubMed] [Google Scholar]
  • 43.Gaspar HB, Aiuti A, Porta F, Candotti F, Hershfield MS, Notarangelo LD. How I treat ADA deficiency. Blood. 2009;114:3524–3532. doi: 10.1182/blood-2009-06-189209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Haas MJ. Two Edges of Sickle Cell Disease. Science-Business Exchange. 2011 Jan 20; Web 22 May 2013. [Google Scholar]

RESOURCES