Dear reader of ADxS.org, please excuse the disruption.

ADxS.org needs about $63500 in 2024. In 2023 we received donations of about $ 32200. Unfortunately, 99.8% of our readers do not donate. If everyone who reads this request makes a small contribution, our fundraising campaign for 2024 would be over after a few days. This donation request is displayed 23,000 times a week, but only 75 people donate. If you find ADxS.org useful, please take a minute and support ADxS.org with your donation. Thank you!

Since 01.06.2021 ADxS.org is supported by the non-profit ADxS e.V..

$8975 of $63500 - as of 2024-02-29
14%
Header Image
Adenosine

Sitemap

Adenosine

Adenosine is

  • a nucleoside
  • a compound of adenine and ribose
  • a neurotransmitter
  • a component of adenosine triphosphate and adenosine diphosphate.

Adenosine is found in almost all body cells. The physiological half-life is in the range of seconds.

Extracellular adenosine levels:1

  • 20 to 300 nM under normal physiological conditions
  • Increase to low micromolar values under extreme physiological conditions, e.g:
    • intensive sport
    • low oxygen content in the air at high altitude
  • Increase to high micromolar values (30 µM) under pathological conditions, e.g:
    • Ischemia

The adenosine system is closely linked to the dopaminergic system. Adenosine receptors are located in particular in the regions of the brain that are insolated in ADHD. Adenosine receptors are closely linked to dopamine receptors and form heteromers with them.
Adenosine inhibits dopamine. Adenosine antagonists (primarily A2A antagonists) are currently being researched for the treatment of Parkinson’s disease and, in our opinion, could also be considered for the treatment of ADHD. At the same time, adenosine modulates striatal DA release by stimulating glutamate release at adenosine receptors in the striatum, which increases dopamine levels.2

1. Behavior regulation through adenosine

Adenosine:

  • Energy regulation of the cells
    • Among other things, adenosine is used for autoregulation in the event of impending energy deficiency in the cell (e.g. when the cell is overstressed or lacks oxygen): If the ATP content in a cell drops, more adenosine is produced as a hydrolysis product. Some of this adenosine is released from the cell and binds to adenosine receptors of neighboring cells, which is intended to compensate for the disturbed balance between energy consumption and energy supply.3
  • Sleep
    • has a dampening / sedative effect3
    • via A1 receptors4
    • Together with melatonin, adenosine regulates sleep depending on neuronal activity and energy metabolism.53
    • Through this mechanism, the increased adenosine level after sleep deprivation could influence the light sensitivity of the circadian clock. 6-hour sleep deprivation reduced the light response in the SCN. Caffeine almost completely restored this SCN light response,6 suggesting an interaction between adenosine and glutamate5
  • Vascular dilation7
    • increased adenosine causes vasodilation
  • Pain8
    • Inhibition of pain via A1 receptors3
    • increased pain sensation via A2A receptors3
  • Inflammation7
    • increased adenosine reduces inflammation
  • Neuromodulation in the CNS8
    • reduces the spontaneous activity of neurons in many regions of the brain
    • inhibits in the CNS:
      • Glutamate5
      • Acetylcholine3
      • Noradrenaline3
      • Dopamine3
        • especially in the mesolimbic dopamine system
      • GABA3
      • Serotonin3
  • Control of voluntary movements (via A2A receptor)9
  • Motivation (via A2A receptor)9
  • Emotion (via A2A receptor)9
  • Cognition (via A2A receptor)9
  • Arousal7
  • Learning7
  • Memory7
  • cerebral blood flow7
  • increases the cramp threshold3
  • is released from heart muscle cells during a heart attack3
    • has a cardioprotective effect via A1 and A3 receptors
    • overexpressed A1 receptors reduce the risk of heart attack
    • overexpressed A3 receptors increase cardiomyopathies.

Chronic overproduction of adenosine is pathological, e.g. in:7

  • Parkinson’s disease
    • Caffeine has a protective effect against Parkinson’s disease
  • Fibrosis
  • Hepatic steatosis
  • Colitis
  • Asthma
    • For asthma:3
      • increased release of adenosine at the bronchial vessels
      • activates bronchial A1 receptors
      • these inhibit adenylyl cyclase
      • cAMP drop has a constrictive effect in bronchial muscles
  • Diabetes
  • Cancer
  • epileptic convulsions (via A2A receptor and neurotrophins)
  • chronic pain10
    • via A2B receptors and IL-6
  • increased sensitivity10
    • via A2B receptors and IL-6

2. Formation of adenosine

Adenosine is formed in two ways:

  • intracellular adenosine synthesis11 (physiologically predominant synthesis pathway in a healthy state)1 in the striatum, among other places
    • AMP is hydrolyzed to adenosine by cytoplasmic 5′-nucleotidase (stronger)
    • S-adenosyl-homocysteine (SAH) hydrolyzed to adenosine by SAH hydrolase (lower)12
    • Release of adenosine into the extracellular space by bidirectional equilibrative nucleoside transporters (ENT)
  • extracellular adenosine synthesis8 (predominant in cellular stress such as injury, hypoxia, neurodegeneration, neuroinflammation or excitotoxicity)1
    • ATP from neurons or glial cells is dephosphorylated to ADP and AMP by the enzyme ectonucleoside triphosphate diphosphohydrolase (CD39). ADP and AM are then converted to adenosine by a specific ecto-5′-nucleotidase enzyme (CD73).1
      • ATP itself appears to be a neurotransmitter and to have an inhibitory effect in the intestine and an exitatory effect in the autonomic nervous system.1314

Adenosine is a degradation product of ATP. High ATP consumption by the cells (due to high neuronal activity) leads to high adenosine.12 This mechanism serves to regulate the energy level of the cells.
Rising adenosine levels increase sleep pressure and thus promote recovery. Sleeping at night reduces adenosine and thus lowers sleep pressure.

Neurodegenerative diseases release high concentrations of ATP from the damaged neuronal and non-neuronal brain cells, which is then enzymatically degraded to adenosine. Neurodegenerative diseases (dementia, Alzheimer’s, Parkinson’s, Huntington’s) appear to cause two-stage damage: primarily through various protein aggregates, resulting in secondary superimposed damage, particularly through P2X7 and A2AR activation. Small molecule antagonists can efficiently prevent or attenuate this damage. P2X7 and A2AR also appear to be involved in ADHD, depression and obsessive-compulsive disorder.14

3. Effect of adenosine: receptors

Adenosine receptors are G-protein-coupled and occur frequently in almost all human tissues and organs.
They are a subgroup of purinoceptors (purinergic receptors), which are divided into ATP receptors (P2 receptors) and adenosine receptors (P1 receptors).14

Affinity:1
Under normal conditions, adenosine has a higher affinity for A1 and A2A receptors. Only at higher adenosine levels are A2B and A3 receptors also addressed.

3.1. A1 receptor

  • inhibits adenylyl cyclase via Gi/o proteins8
  • encoded by the ADORA1 gene on chromosome 1 (1q32.1)

3.1.1. A1 receptors in the brain

  • Cortex8
  • Hippocampus8
  • Cerebellum8
  • Nerve endings15
  • Spinal cord15
  • Glial cells15
  • Striatum
    • at the neck of dendritic spines, where they can interact with extrasynaptic dopamine and metabotropic glutamate receptors as heteromers 11
    • presynaptically at glutamatergic axon terminals, where they modulate glutamate release11
    • presynaptically at dopaminergic synapses, where they inhibit the release of dopamine16
      • the modulation of dopamine release mediated by A1 receptors in the striatum is complex and differs in different striatal compartments

3.1.2. Effect mediated by A1 receptors

3.1.2.1. Neurophysiological effect in the brain
  • Inhibition of the release of neurotransmitters817

  • Reduction of neuronal excitability817

  • Reduction of dopamine D1 signaling18

  • Synaptic plasticity

    • Brain development19
    • Long-term potentiation (LTP, “learning”) primarily via agonists, long-term depression (LTD, “forgetting”) primarily via antagonists in17
      • Hippocampus
      • Striatum
      • Hypothalamus
      • Cerebellum
  • Increase in Homer1a expression18

  • presynaptic:18

    • Inhibition of the release of
      • Dopamine
      • Glutamate
      • Serotonin
      • Acetylcholine
  • postsynaptic:18

    • Impairment of neuronal signal transmission due to
      • Hyperpolarization of the neuron membrane
      • Reduction in the excitability of potassium channels
  • Promotion of self-adaptive changes to regulate neuronal plasticity by heteromers with A2A and D1 receptors18

  • The A1 agonist CPA increased the binding of the alpha2-adrenoceptor in the nucleus tractus solitarius. The increase in binding was around 10 times greater with SHR than with WKY.20

3.1.2.2. Behavior influenced by A1
  • Learning17
  • Memory17
  • Movement activity8
  • Discrimination8
  • Search8
  • Reward8
  • Regulation of sleep8
  • Sedation8
  • anticonvulsant8
  • anxiolytic8
    • Adenosine A1 receptors modulate the anxiolytic effect of ethanol2122
  • Sensation of pain8

Studies confirm the potential of A1 agonists as an effective strategy to combat the effects caused by psychostimulants.

3.1.2.3. Physiological effect in the body
  • reduces renal blood flow23
  • reduces glomerular filtration rate (GFR)23
  • stimulates the release of renal renin23
  • increases proximal tubular sodium reabsorption23

3.1.3. A1 receptor ligands

3.1.3.1. A1 agonists
  • 2-chloro-N6-cyclopentyladenosine (CCPA)21
  • N6-cyclopentyladenosine (CPA) is an A1 agonist2425
  • 5’-N-Ethylcarboxamidoadenosine (NECA) is an A2/A1 agonist24
  • N6-R-phenylisopropyladenosine (L-PIA) (selective)12
3.1.3.2. A1 antagonists
  • Caffeine is a non-selective adenosine receptor antagonist24
    • Adenosine A1 receptors in the brain regulate the need for sleep. Adenosine A1 receptors inhibit the enzyme adenylyl cyclase, which is required for the conversion of ATP into cAMP. This inhibition is prevented by caffeine, the cAMP level remains high. This increases alertness.26
  • Rolofylline23
  • [3H]-DPCPX (8-cyclopentyl-1,3-dipropylxanthine)2725
  • Theophylline (3-methyxanthine)
  • CPT (8-cyclopentyltheophylline)24
  • Doxofylline (7-(1,3-dioxalan-2-yl-methyl) theophylline)
    • similar efficacy to theophylline with lower adenosine receptor affinity28

One side effect of A1 antagonists is (epileptic) seizures.

Iron deficiency appears to be related to a change in the expression of adenosine receptor subtypes in the cortico-striatal glutamatergic terminals:2930

  • Downregulaton from A1R
  • Relative upregulation of A2AR

3.2. A2A receptor

Former name: RDC8
Activation of the A2A receptor activates adenylyl cyclase via Gi/o proteins.8
Encoded by the ADORA2A gene on chromosome 22 (22q11.23)

3.2.1. A2A receptors in the brain

A2A receptors are most commonly found in the striatum of the brain in both humans and rats. In other areas of the brain, they are found much less frequently.31

  • Striatum8
    • A2A is mainly found in the striatum and less in other brain regions.12 In the entire striatum:
      • Caudate nucleus31
      • dorsal striatum31
      • ventral striatum31
      • Nucleus accumbens12
    • Distribution In the striatum31
      • up to 3 % on astrocytes
      • to 90 % on neurons, of which
        • 70 % postsynaptic
          • mainly in the GABAergic medium spiny neurons of the indirect pathway (projecting to the external segment of the globus pallidus), which also express a high density of D2 receptors and enkephalin, with A2A receptors located close to the D2 receptors.
          • hardly in neurons of the direct striato-nigral pathway (which selectively express D1 receptors and the peptide dynorphin)
        • 23 % presynaptic
          • mainly at cortico-thalamic glutaminergic axon terminals that were in contact with medium spiny neurons of the direct and indirect GABAergic pathways and at cholinergic neurons that modulate acetylcholine release
          • presynaptic
        • 3 % extrasynaptic
        • In the dorsal striatum12
          • 95-96 % of A2A is co-expressed with D2
          • 3-6 % of A2A co-express D1 or substance P mRNA.
        • In the ventral striatum12
          • 89-92 % of A2A co-express with preproenkephalin-A
          • 93-95 % of A2A co-express with D2 receptors
      • in medium, but not in large neurons12
    • in particular at the perisynaptic ring of the glutamatergic synapse in enkephalin neurons, where they can interact with D2 and mGlu5 receptors as heteromers.11
    • at the neck of dendritic spines, where they can interact with extrasynaptic dopamine and metabotropic glutamate receptors as heteromers.11
    • presynaptically at glutamatergic axon terminals, where they modulate glutamate release11
  • Olfactory bulb8
  • optical cortex (low)31
  • Amygdala (low)31
  • Hippocampus (low)31
  • Substantia nigra (low)31
  • Cerebellum (low)31

Stimulation of the A2A receptor could be a potential therapeutic target for the treatment of drug addiction.8

3.1.2. Effect mediated by A2A receptors

3.1.2.1. Neurophysiological effect in the brain of A2A
  • Reduction of dopamine D2 signaling18
    • Conversely, inhibition of adenosine by dopamine. These interactions arise (at least in part) through allosteric receptor-receptor interactions within heteromeric A2AR/D2R complexes12
  • Increase in the release of excitatory neurotransmitters18
  • Regulation of neuroinflammation187
    • In chronic autoimmune rheumatic diseases, A2A and A3 receptors are overexpressed in lymphocytes. A2A and A3 agonists inhibited the activation of NF-κB, the release of typical proinflammatory cytokines and the concentration of metalloproteinases, which are involved in the inflammatory reactions in chronic autoimmune rheumatic diseases.32
  • Modulation of neuronal glutamate release7
    • Increase in mGLUR5 signaling18
    • Potentiation of NMDA-mediated effects7
    • Release of glutamate from glutamatergic endings7
    • Inhibition of the glutamate-1 transporter (GLT-1) in astrocytes7
  • Modulation of glial reactivity7
  • Modulation of the permeability of the blood-brain barrier7
  • Infiltration of peripheral immune cells7
  • Ischemic damage7
  • Influencing neurite branching, neurite length and axonal growth in PFC neurons33
3.1.2.2. Behavior influenced by A2A
  • Regulation of vigilance34
  • ADHD-like behavior34
  • Synaptic plasticity
    • Corticoaccumbens and hippocampus
      • reduced long-term potentiation (LTP, “learning”) due to antagonists17
    • Hippocampus
      • increased kainate and BDNF-modulated LTP in the hippocampus by A2A agonists
    • Striatum
  • Learning3517
  • Memory3517
    • Caffeine (as a non-selective adenosine antagonist) as well as selective adenosine A2A antagonists can improve memory performance in rodents and protect against memory impairment
  • Cognition17

A2A overexpression correlates with36

  • Fear
  • Depression

A2A polymorphisms correlate with:37

  • Fear
  • Panic disorders

A2A knockout mice (mice without A2A receptor):38

  • Aggressiveness strongly increased (males)
  • Increased anxiety
  • Exploratory behavior reduced
    • Caffeine further reduced this
  • Pain sensation reduced
  • Increased blood pressure
  • Increased heart rate
  • Increased platelet aggregation

3.2.2. A2A receptor ligands

3.2.2.1. A2A agonists

  • 2-p-(2-carboxyethyl)phenethylamino-5’-N-ethylcarboxamidoadenosine (CGS 21680) is an A2A agonist24
  • 5’-N-Ethylcarboxamidoadenosine (NECA) is an A2/A1 agonist24
  • CGS 216803933
    • causes a two- to threefold decrease in the affinity of D2 receptors for dopamine receptor agonists 12
    • reduced the availability of D2 receptors in the striatum in a study in rats.40
    • reduced effect at very high, saturating doses, presumably due to A2A downregulation41
  • 3,4-Methylenedioxybenzoyl-2-thienylhydrazone (LASSBio-294)42
    • lowered blood pressure and prevented heart defects in SHR after myocardial infarction

3.2.2.2. A2A antagonists

  • Istradefylline (KW 6002)1243
    • Approval in 2019 in the USA (first ever approved A2A antagonist) for the treatment of Parkinson’s disease (brand name: Nourianz®),44 but only when the drug’s overall target was restricted from Parkinson’s to Parkinson’s with off-episodes.
    • Approval refused in the EU.4546
    • The most frequently reported side effects were46
      • Dyskinesia
      • Hallucinations
      • Constipation
      • Dizziness
      • Nausea
      • Vomiting
    • Patent expires in 202444
    • Istradefylline did not reduce the availability of D2 receptors in the striatum in a study in rats.40
  • Preladenant (SCH 420814)4743
    • Development discontinued after no more effective than placebo in phase III
  • Tozadenant (RO-449351, SYN-115; 4-hydroxy-N-(4-methoxy-7-(4-morpholinyl)benzo[d]thiazol-2-yl)-4-methylpiperidine-l-carboxamide)4743
    • several phase III trials discontinued prematurely
    • Death of 5 patients in a Phase III study with 409 participants due to drug-induced agranulocytosis (formation of antibodies against neutrophil membrane glycoproteins, which leads to the destruction of neutrophils). Other approved drugs also have this side effect. Has not yet been observed with Istradefylline.44
  • Vipadenant4743
  • KW-6356 (2nd generation, selective)44
    • is to become a successor to Istradefylline
    • positive results in phase II test
  • KW-600248
  • ST 15354743
  • PBF-50943
  • ST420643
  • V8144443
  • DMPX (3,7-dimethyl-1-propargylxanthine) is a selective A2 antagonist24
  • MSX-339
  • SCH 58261 (selective)33
  • ZM241385 (4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo-[2,3-a][1,3,5]triazin-5-yl-amino]ethyl)phenol)2127
  • SCH58261 (selective)12
  • SCH-41234812
  • Cmpd-127
    • is a dual antagonist of A2AR / NR2B.
    • has a 15-fold higher affinity to the A2AR than to the A1R
    • binds NR2B with high affinity (pKi = 8.25)
    • is therefore a potential Parkinson’s drug
  • AZD4635 (HTL1071, Imaradenant)49
    • a 5,6-diaryl-1,2,4-triazine-3-amine derivative
    • is currently undergoing clinical trials for cancer immunotherapy
    • had previously been considered for the treatment of ADHD
    • Ki value: 1.7 nM for the human A2A receptor
    • more than 30 times the selectivity of other adenosine receptors

A2A antagonists have been shown to be helpful in:

  • Working memory problems505152
  • Memory disorders53
  • Reverse learning48
  • Motivation 5455
  • goal-oriented behavior56
  • Task change44
  • Fear conditioning57
  • Improvement in cognitive performance55
    • The effect of istradefylline on cognition was not assessed in its clinical trials in Parkinson’s patients
  • Mood (depression) in Parkinson’s disease (here due to Istradefylline)58
  • Anhedonia55
  • traumatic brain injuries59
    • this is also reported for ADHD stimulants
  • acute and chronic stress6061
  • Restless legs syndrome (RLS)7
    • RLS is a common comorbidity of ADHD
  • Huntington’s disease51
  • Alzheimer’s disease62
  • severe depression7
  • Schizophrenia7
  • Epilepsy7

Iron deficiency, which is also a cause of RLS, appears to be related to a change in the expression of adenosine receptor subtypes in the cortico-striatal glutamatergic terminals:2930

  • Downregulation of A1R
  • Relative upregulation of A2AR

3.3. A2B receptor

Encoded by the ADORA2B gene on chromosome 17 (17p12)

Occurrence in the brain in8

  • Astrocytes
  • Nerve cells
  • Microglia

The A2B receptor

  • stimulates adenylyl cyclase via Gi/o proteins8
  • hardly represented in the brain811
  • weak adenosine binding. Only activated when extracellular adenosine levels are very high (micromolar), e.g. after tissue damage (e.g. inflammation, hypoxia, ischemia, brain injury)12

Ligands:

  • 5’-N-Ethylcarboxamidoadenosine (NECA) is an A2/A1 agonist24

3.4. A3 receptor

Occurs in the brain only in small quantities11 in:8
Encoded by the ADORA3 gene on chromosome 1 (1p13.2)

  • Cortex
  • Thalamus
  • Hypothalamus
  • Hippocampus
  • Motor nerve endings
  • Retinal ganglion cells
  • Pial and intracerebral arteries
  • Glia

The A3 receptor

  • inhibits adenylyl cyclase via Gi/o proteins8
  • hardly represented in the brain
  • influences synaptic plasticity in the hippocampus17
  • In chronic autoimmune rheumatic diseases, A2A and A3 receptors are overexpressed in lymphocytes. A2A and A3 agonists inhibited the activation of NF-κB, the release of typical proinflammatory cytokines and the concentration of metalloproteinases, which are involved in the inflammatory reactions in chronic autoimmune rheumatic diseases.32
  • weak adenosine binding. Only activated when extracellular adenosine levels are very high (micromolar), e.g. after tissue damage (e.g. inflammation, hypoxia, ischemia, brain injury)12

Agonists:

Cl-IBMECA is a selective A3 agonist17

  • increased theta burst-induced LTP and attenuated LTD

Antagonists

MRS-1191 is a selective A3R antagonist17

  • reduced theta burst-induced LTP and attenuated LTD

3.5. Receptor heteromers

Adenosine receptors also occur as monomers, heteromers and omnimers:8

  • with each other (e.g. A1 / A2A)1
    • low adenosine preferentially stimulates A1 –> inhibition of glutamatergic transmission
    • high adenosine stimulates A2A –> blockade of A1-mediated effects –> potentiation of glutamate release
  • with other receptors (e.g. A2A / D2 heteroidmer)
    • A2A / D2 heterodimers appear to be partly responsible for the psychomotor and reinforcing effects of psychostimulants such as cocaine and amphetamine8

In the striatum31

  • A2A / A2A (on the cell surface)
  • A2A / A1
  • A2A / A2B
  • A2A / A3
  • A2A / CB1
  • A2A / D2
    • especially in GABAergic medium spiny neurons (MSN) of the indirect pathway12
      • striatopallidal: motor control
      • Nucleus accumbens: reward-related behavior
    • in astrocytes12
    • in Glia12
    • in cholinergic interneurons12
    • The mutual inhibition of adenosine and dopamine in mammals is mediated by A2A / and D2 receptors, at least parts of which are mediated by A2A / D2 heteromeret.
    • A2A agonists inhibit and A2A antagonists enhance D2-mediated locomotor activation (e.g. in striatopallidal GABAergic medium spiny neurons (MSN) of the indirect pathway) and goal-directed behavior.
    • A2A / D2 co-aggregate, co-internalize and co-desensitize, i.e. mechanisms such as downregulation do not affect one part of the heteromer alone, but the entirety12
  • A2A / D3
  • A2A / mGlu5
  • A2A / FGFR1 (fibroblast growth factor receptor)
  • A2A / Sigma1 receptors
  • A1 / A2A (in glutamate axon terminals presynaptic)
  • A2A-D2 heterodimers in equilibrium with trimeric A1-A2A-D2 heteroreceptor complexes
  • A2A/D2 receptor heterodimers appear to form heterotetramers. It follows that, at high concentrations, A2A antagonists act in the same way as A2A agonists, namely by reducing D2 receptor-mediated activity in neurons.63

In the hippocampus
found in moderate to high density in the dorsal hippocampus, mainly in the pyramidal cell layer31

  • A1 / A2A
  • A2A / A2B
  • A2A / A3

3.6. Agonists and antagonists

3.6.1. Agonists

  • Adenosine (A1 and A2A antagonist; used as A1 antagonist in aroxysmal supraventricular tachycardia (PSVT); used as A2A antagonist in myocardial perfusion)1
  • 2-chloroadenosine (CAD) is a non-specific adenosine receptor agonist64

3.6.2. Antagonists

  • Xanthine
    • Caffeine (A2A and A1 antagonist)
      • Recommended daily dose max 5.7 mg/kg65
    • Theophylline (A1 and A2 antagonist; only minor A3 antagonist)
    • Paraxanthin (caffeine metabolite)66
    • 1-methylxanthine (caffeine and theophylline metabolite)66
    • Theobromine (weak antagonist)66
  • Istradefylline (A2A antagonist; used for Parkinson’s disease)1 (possibly also for depression)67
  • Regadenoson1
  • Doxofylline128
  • Bamifylline1

Relative potency of methylxanthines:3

Effect Caffeine Theophylline Theobromine
CNS stimulation +++ +++ -
Heart + +++ ++
Bronchodilation and vasodilation + +++ ++
Skeletal muscle stimulation +++ ++ +
Diuresis + +++ ++
3.6.2.1. Caffeine

Due to the special importance of caffeine, we dedicate a separate section to it.

3.6.2.1.1. General information on caffeine

1.6 billion cups of coffee are consumed worldwide every day.68 Coffee was first mentioned in a medical text in 1025.

Caffeine is an A2A and A1 antagonist.
The highest blood caffeine level occurs approx. 30 to 40 minutes after consumption. The half-life is approx. 3 to 6 hours and is longer in pregnant women and shorter in smokers69

The values per portion (cup, jar, can, 50 g bar)69

  • ground coffee 105 mg
  • Energy drinks 80 mg
  • Instant coffee 54 mg
  • Tea (bags, loose leaves, instant tea, green tea) 40 mg
  • Cola 16 to 30 mg
  • Chocolate 8 to 27 mg

Caffeine from coffee and black tea is released differently.
Caffeine from roasted coffee is bound to a chlorogenic acid-potassium complex and releases caffeine immediately after contact with stomach acid. Coffee caffeine therefore works quickly.
Caffeine from tea is bound to polyphenols. The caffeine is only produced through fermentation and is released in the intestine, so it has a later and longer-lasting effect.70
Tea is therefore preferable to coffee when it comes to treating ADHD.
Green tea contains just as much caffeine as black tea. However, the same source states that caffeine is only released during fermentation. Green tea is unfermented, black tea is fermented. Young tea leaves (pekoe) contain particularly high levels of caffeine.71

3.6.2.1.2. Long-term caffeine consumption and habituation

Chronic (long-term) caffeine consumption causes adaptations in the adenosine system that counteract the effects of isolated caffeine intake.69

Chronic caffeine administration in SHR (a genetic animal model of ADHD-HI) of 2 mg/kg for 21 days (which should be sufficient for receptor adaptation regulation) induced72

  • normalized dopaminergic function (reduced in SHR per se)
  • improved memory and attention deficits (which are typical in SHR)
  • upregulation of A2A in frontocortical nerve endings

Caffeine further improved in vitro in the striatum of SHR the

  • GABA release (reduced with SHR per se)
  • GABA reuptake via GAT1 transporter (reduced in SHR per se)

whereas this was not the case with Wistar rats (which are not an ADHD animal model).73

This result could be an indication of a positive effect of caffeine on ADHD.
In contrast, other studies suggest that the effects of caffeine on alertness and cognitive performance do not represent a net benefit for functioning, but merely a reversal of withdrawal effects. Acute caffeine withdrawal (e.g. overnight) worsens alertness, cognitive performance and mood; caffeine consumption restores these to normal levels but does not improve them beyond that 7469

One study found that increased caffeine consumption in students correlated with increased anxiety and depression symptoms and poorer academic performance.75 It remains to be seen whether these symptoms are causally caused by caffeine or whether caffeine is used as (insufficient) self-medication due to a dopamine deficit.

Humans and laboratory animals develop tolerance to some, but not all, of the effects of caffeine767778
Note: The following doses are 10 times the maximum recommended dose for humans of 5.7 mg/kg/day.

  • A1
    • chronic doses above 50 mg / kg / day: upregulation of A1 receptors in the cerebral cortex,79 and also in the hippocampus (CA3), without changes in gene transcription, apparently due to a blockade of downregulation caused by adenosine in the absence of caffeine.80 Other studies found no increase in A1 receptors in the hippocamus, cerebral cortex or cerebellum.81
      • The inhibition of lipolysis in fat cells by adenosine remained unchanged
    • chronic doses below 50 mg / kg / day:
      • Number of receptors unchanged8283
      • Tolerance development, possibly by means of altered gene transcription84
  • A2A
    • Receptor number not or hardly changed by chronic high doses8584
      • A1 can be easily downregulated in vitro, whereas A2A cannot
    • basal adenylyl cyclase activity and cyclase activities stimulated by adenosine agonists, GTP gamma S or forskolin are reduced in cells desensitized by chronic caffeine administration, but also in ways other than changes in receptor number85
  • increased functional sensitivity to adenosine8687888990

In one study, subjects were given 300 mg of caffeine or placebo daily. Mood and subjective effects only differed within the first 4 days. A high dose of caffeine (300 mg twice a day) was able to improve the effects of caffeine

  • Tension / anxiety
  • jittery / nervous / shaky
  • active / excited / energetic

only in the placebo group. The caffeine group had developed complete tolerance.

3.6.2.1.2.1. Tolerance development

Tolerance to caffeine developed within 1 to 3 days and ended 3 to 4 days after cessation of chronic caffeine administration91
Tolerance development in rats from a dose of 6 mg/kg/day86

Repeated daily caffeine intake can reduce the physiological effects of a single dose of caffeine within a few days. A single dose of caffeine causes86

  • increased water excretion
  • Salivation
  • increased metabolic rate (oxygen consumption)
  • increased blood pressure
  • increased noradrenaline and adrenaline plasma levels
  • increased renin plasma level activity
  • Sleep disorders
    • After 7 days of caffeine consumption, total sleep time, sleep efficiency and frequency of awakenings no longer differed from the initial value.

Little is known about the mechanisms of caffeine tolerance formation. Therefore, the question of whether long-term high caffeine consumption has advantages or disadvantages in relation to ADHD will need to be discussed in more detail. A concern seems to be that chronic caffeine consumption increases sensitivity to adenosine and thus counteracts the desired effect of reducing adenosine in order to reduce the inhibition of dopamine by adenosine.87888990 In contrast, such receptor adaptation does not occur with D-amphetamine.92
It remains to be seen whether moderate or daily alternating caffeine consumption can achieve a reduction in adenosine without sensitization to adenosine.

3.6.2.1.2.2. Withdrawal

Symptoms of withdrawal from chronic high caffeine intake are described:86

  • reduced locomotor skills9291
    • single caffeine administration increased locomotion, chronic caffeine administration did not, while D-amphetamine increased locomotion with both single and chronic administration92
    • locomotion normalized within 4 days after the end of chronic caffeine administration. Then even a single dose of caffeine had the same effect as before chronic caffeine administration92
    • Rats showed no withdrawal effects with regard to locomotor activity after withdrawal from 19 mg/kg/day or 36 mg/kg/day. Only at 67 mg/kg/day did locomotion halve on the first day of withdrawal.91 ) Withdrawal from 190 mg/kg/day of caffeine over 7 weeks showed a reduction in locomotion to one fifth on the first day92
  • reduced operant behavior939495
    • Caffeine was more effective than thephylline than theobromine in this respect93
  • reduced amplification threshold for electrical brain stimulation94
  • relatively longer sleep phases I and II96
  • Avoidance of previously preferred flavors if they are now presented without caffeine97
    • A positive assessment of the taste of caffeinated foods is directly caused by the effect of the caffeine itself. A test with two novel-tasting fruit juices, which were initially judged as equally positive, with which either a capsule containing caffeine or a placebo was taken, showed a clearly positive assessment of the taste by the test subjects who received caffeine - but mainly by those test subjects who were used to caffeine and were currently in “withdrawal”.98
  • Headache frequent withdrawal effect
    • Caffeine causes vasoconstriction. After chronic caffeine consumption, vasodilation occurs on cessation, leading to increased cerebral blood flow, which appears to be a common cause of headaches.99

The withdrawal period in rats was approx. 10 days,94 whereby individual behaviors only returned to normal after 30 days.96
A study in rats with chronic caffeine consumption of 30 mg/kg/day (corresponding to 4-5 cups of coffee/day in humans) over 12 weeks found a decrease in A1 receptors in the brain by around a third on the first day of withdrawal, which had returned to normal after 27 hours on average. In contrast to the other brain regions, the nucleus accumbens and hypothalamus showed no change in A1 receptors.100

In humans, after 3 x 250 mg/day of caffeine over 7 days, the blood plasma was free of caffeine after 60 hours87
The withdrawal period in humans appears to be around 14 days.86

3.6.2.2. Theophylline

Theophylline is an A1 and A2 antagonist; weaker A3 antagonist. The affinity to human adenosine receptors is (from strong to weak):101102

  • A2A: 2-10 µM
  • A1: 10-30 µM
  • A2B: 10-30 µM
  • A3 receptor: 20-100 µM

Theophylline also has the following effects3

  • an inhibition of phosphodiesterases
    • Phosphodiesterases break down the regulatory substance cyclic adenosine monophosphate (cAMP)
    • cAMP acts as a second information carrier (“second messenger”) and has a number of functions:
      • Hormone stimulation
      • Mediation of reactions to neurotransmitters
      • Triggering chemotactic behavior
      • act on effector cells that are involved in the pathogenesis of type I allergies
  • the mobilization of intracellular Ca2+ depots by means of ryanodine receptors.

Theophylline occurs naturally in
Guarana - up to 0.25 %
Tea leaves - approx. 0.03 % dry weight103 to 0.1 %70
black tea 0.02 to 0.04 %104
Mate leaves - unclear:
0 to 0.004 % 104
approx. 0.05 to 0.1 %70
Cocoa - traces103 up to approx. 0.05 %
Coffee bean - Traces103
Kola nut - Traces

3.6.2.2.1. Theophylline for asthma and COPD
  • Use for asthma and COPD)1
  • only available on prescription in D, A, CH
  • Theophylline is able to reduce corticosteroid resistance via genetic pathways. This is the pathway by which theophylline is helpful in COPD and asthma.10528
    • this apparently already at low levels of 5mg/L101
  • weak, non-selective inhibitor of PDE28
    • PDE degrade the cyclic nucleotides in the cell
      • leads to an increase in intracellular
        • cyclic 3’5’-adenosine monophosphate (AMP)
        • cyclic 3’,5’-guanosine monophosphate (GMP)
    • PDE inhibition at therapeutically relevant doses only low (5 - 10 % in human lung extracts)106
    • PDE inhibition stronger in asthma107
  • prevents the bronchoconstrictor effect of adenosine at therapeutic doses108
  • IL10 increasing
    • IL-10 is reduced in asthma and COPD109
    • unclear whether this is also relevant for therapeutic doses
  • Inhibits NF-κB110
    • prevents translocation of NF-kB into the cell nucleus
    • possible pathway to prevent expression of inflammatory genes in asthma and COPD
  • Inhibits phosphoinositie-3-kinases directly, but weakly
    • pI3K (p110)-δ subtype (IC50 75 μM) still strongest
      • involved in reactions to oxidative stress
    • the ability of theophylline to reverse corticosteroid resistance appears to be based on this mechanism111
      • important for its clinical effect in severe asthma and COPD
  • Plasma concentrations above 20 mg/L often show side effects28
    • Headache
      • through inhibition of PDE4
    • Nausea and vomiting
      • through inhibition of PDE4 in the vomiting center
    • Stimulation
      • could be the desired effect for ADHD
      • via A1 antagonism
    • Discomfort in the abdomen
    • Restlessness
    • increased gastric acid secretion
      • via A1 antagonism
    • gastroesophageal reflux
    • Diuresis
      • via A1 antagonism
    • increased inflammation
      • via A2A antagonism101
  • at even higher levels
    • Cramps
    • Cardiac arrhythmia
      • through inhibition of PDE3
      • via A1 antagonism
  • Deaths after additional intravenous administration of aminophylline, e.g. in the emergency room for severe asthma.112

Asthma treatment28

  • Theophylline is recommended as an additional bronchodilator if high doses of inhaled corticosteroids are not sufficient
  • higher side effects than long-acting inhaled β2 agonists, which are also more effective
3.6.2.2.2. Theophylline for ADHD

Theophylline was shown to be equivalent to methylphenidate in the treatment of ADHD in children in parent and teacher ratings in a small double-blind randomized study over 6 weeks. A dose of 3 mg/kg/day in children up to 11 years and 4 mg/kg/day in children 12 years and older produced equivalent results to 1 mg/kg/day of methylphenidate. The side effects of theophylline were lower than those of MPH (headache and dropout rate).113 The fact that the results were obtained over a period of 6 weeks indicates that theophylline (unlike caffeine) does not show any habituation effects.

In a double-blind crossover study of 14 (asymptomatic) children with asthma, theophylline showed an improvement in behavior in the second week of treatment after parent rating. Cognitive improvements were not observed. 114

A meta-study found an effect on ADHD symptoms in 10 studies on theophylline in asthma sufferers (without ADHD). Hyperactivity was mentioned most frequently, as were distractibility, inattention, irritability and sleep problems.115 As the studies involved children without ADHD, the causation of ADHD symptoms is an indication that theophylline affects these symptoms. Given the inverted-U effect116117 of neurotransmitters and dopamine in particular (too little dopamine causes similar symptoms to too much dopamine), the causation of ADHD symptoms in non-ADHD sufferers could be explained by the fact that an increase in dopamine levels caused too much dopamine in them. Since ADHD is characterized by a dopamine deficiency, use in ADHD sufferers could compensate for this deficit.

Due to its degradation mechanism, theophylline increases the plasma level and bioavailability of melatonin.118 Melatonin in turn deactivates the HPA axis, although this has probably only been studied at extremely high doses in rats.

4. Adenosine and neurotransmitters

4.1. Adenosine and dopamine

A1 receptors form heteromers with D1 receptors.
A2A receptors form heteromers with D2 receptors.8

4.1.1. Adenosine inhibits dopamine at A1 receptors

Adenosine inhibits dopaminergic neurotransmission via adenosine A1 and A2 receptors. Since A1 receptors primarily form heteromers with D1 receptors, which are located outside the striatum, adenosine inhibits D1 receptors in particular. Since A2A receptors primarily form heteromers with D2 receptors within the striatum, adenosine inhibits dopaminergic transmission in the striatum, the reward system, at the latter.8

Adenosine A1 receptor antagonists have the effect of increasing dopamine levels.

4.1.2. Adenosine and adenosine antagonists inhibit D2 receptors in A2A-D2 heteromers

A2A-D2 heteromers occur primarily in the striatum, especially in the GABA-ergic striatopallidal neurons. Here, A2A activation increases GABA release and counteracts the effects induced by D2 receptors.8

In the striatum8

  • antagonistic relationship between A2A and D2
    • predominant (due to the higher distribution of adenylyl cyclase type V)
  • upon upregulation of “Activator of G-protein signaling 3” (AGS3):
    • increasingly synergistic interaction
    • e.g. chronic exposure to addictive substances

Spine neurons in the striatum are predominantly controlled by dopamine, glutamate, acetylcholine and adenosine. Adenosine is released inside and outside the synapse, from where it addresses extrasynaptic and intrasynaptic adenosine receptors in and near glutamatergic synapses.11

At least in A2A / D2 heteromers, A2A ligands (agonists as well as antagonists alone, but not when agonists and antagonists occur simultaneously) cause a reduced affinity and signaling effect of D2 agonists.6312 If, on the other hand, a D2 agonist binds to a D2 heteromer, the binding of A2A agonists is suppressed12
The A2A / D2 interaction may influence the intracellular formation of cyclic AMP not only at the membrane level, but also at the second messenger level. This could even be the decisive effect.12

Details on the interaction of A2A and D2 receptors

A2A / D2 heteromers cause antagonistic interactions between A2A and D2 at the adenylyl cyclase level
A2A and D2 receptors can be connected in two opposite ways: at the membrane level and intracellularly8

  • Membrane level
    • A2A activation has a balancing effect on D2 stimulation:41
      • Activation has a balancing effect compared to D2 stimulation119
        • reduced D2 dopamine affinity
        • increased tonic dopamine level in the nucleus accumbens
        • increased extracellular GABA level
          • in the nucleus accumbens
            • but not by dopamine antagonist
          • in the ipsilateral ventral pallidum
            • also through
              • Dopamine antagonist
              • joint administration of A2A agonist and D2 antagonist in such low doses that they were ineffective on their own
        • reduced reward and addictive behavior with cocaine120
      • functional effects induced by D2 stimulation are attenuated
    • Animal models with excessive A2A expression in the brain show reduced D2 numbers in the striatum.
      • A2A activation reduces behavioral responses to psychostimulants
  • synergistic interaction between A2A and D2 at the adenylyl cyclase level in the striatum, with overexpression of the “Activator of G-protein signaling 3” (AGS3)
    • occurs with upregulation of AGS3, e.g. with ethanol consumption or withdrawal from cocaine, ethanol, morphine
    • AGS3 activity
      • stabilizes / inhibits the GDP-bound form of Gi
      • simultaneously increases the βγ-dependent effect of the Gs/olf protein
        –> strong increase in cAMP-PKA signaling

A2A receptor agonists mediate neuroprotection by increasing NF-κB8

4.2. Adenosine inhibits noradrenaline

The adenosine A1 and A2A antagonist caffeine promotes noradrenaline.121 in the nucleus coeruleus.122

4.3. Adenosine and glutamate, acetylcholine, serotonin, histamine

Adenosine correlates with increased glutamatergic neurotransmission. Stimulation of glutamate NMDA receptors releases adenosine at the postsynapse of striatal neurons. Presynaptically, increased glutamate input (presumably through increased release of synaptic ATP) causes a rapid increase in adenosine at the glutamatergic synapse11

A1 and A2A receptors in cholinergic nerve terminals appear to modulate striatal acetylcholine release.123
A1 and A2A receptors modulate the release of serotonin. It is possible that A1 / A2A receptor heteromers exist that control both acetylcholine and serotonin release.123 A2A antagonists cause serotonin release in the tractus solitarius.124
Adenosine is able to modulate the ascending histaminergic excitation system via A2A receptors in the hypothalamus.123

5. Degradation of adenosine

5.1. ENT transporter

Under resting conditions, adenosine is approximately equally abundant intracellularly and extracellularly.
In pathophysiological conditions (inflammation, ischemia and hypoxia) characterized by high adenosine concentrations, reuptake by ENTs is the main mechanism of extracellular adenosine degradation.
There are two adenosine transporters:7

  • ENT1
  • ENT2

Adenosine reuptake inhibitors:

  • [3H]nitrobenzylthioinosine ([3H]NBMPR)125
  • Cannabinoids inhibit the reuptake of126
    • Adenosine (stronger)
    • Dopamine (weaker)
      in the striatum. This applies to a large number of endogenous and exogenous cannabinoid ligands. The maximum strength of reuptake inhibition often corresponded to that of the dopamine reuptake inhibitor GBR12783 and the equilibrative nucleoside reuptake inhibitor dipyridamole. The inhibition was apparently not through the cannabinoid-1 receptor.

5.2. Adenosine deaminase (ADA)

Conversion of adenosine:8

  • Adenosine kinase (AK) to AMP (phosphorylation)
    • AK has a higher affinity for adenosine than ADA.
    • predominant intracellular degradation pathway in a healthy state
  • Adenosine deaminase (ADA) to inosine (deamination)
    • predominant intracellular degradation pathway in the pathological state
    • also extracellular

6. Regulation of adenosine

Adenosine is increased by:7

  • Hypoxia (lack of oxygen)
  • Ischemia
  • Tissue damage

Adenosine, dopamine and endocannabinoids modulate the release of each other in the dorsolateral striatum and thus control synaptic plasticity.
At a second level of interaction, they regulate each other’s action via the formation of receptor heteromers.

7. Adenosine and ADHD

So far, there is little information on the connection between adenosine and ADHD.
It should be emphasized that adenosine inhibits dopamine and adenosine antagonists promote dopamine. Adenosine and dopamine receptors are closely linked, especially in the areas of the brain that are particularly involved in ADHD. If we look at the areas of action of A2A antagonists, we find a considerable range of typical ADHD symptoms and ADHD comorbidities.
Cannabinoids, which also act as ADHD medications, inhibit the reuptake of adenosine and dopamine in the striatum, thus increasing adenosine and dopamine.

7.1. Methylphenidate and adenosine

Several studies indicate that MPH - at least at extreme doses - also appears to act via A1 receptors.127128

MPH appears to reduce ATP. ATP is the precursor of adenosine in extracellular adenosine synthesis. In mice, chronic MPH administration resulted in a reduction of ATP in the hippocampus by approximately 12 %.129 Since adenosine inhibits dopamine, the reduction in ATP could contribute to the increase in dopamine.

7.2. Caffeine and ADHD

Caffeine is a strong adenosine A1 and A2A receptor antagonist.
In contrast, the other effects (A2B antagonist, A3 antagonist, GABAA antagonist, calcium mobilization and phosphodiesterase inhibition) appear to be negligible. In addition, caffeine increases noradrenaline turnover in the nucleus coeruleus.122 There are indications that caffeine also exerts its dopamine-related effects independently of adenosine receptors.130

Adolescents with ADHD consume caffeine twice as often as those without the disorder131
Caffeine consumption also correlates positively with the severity of ADHD symptoms,132133 which indicates possible “self-medication”.122

Caffeine improves:

  • Attention134
  • Learning ability134
  • Memory1346117
  • Odor discrimination134
  • Reactions to chronic stress61
  • cognitive performance135 in an auditory oddball test136
  • Mood135

Caffeine does not cause any change in:134

  • Blood pressure
  • Body weight

The effect of caffeine is unclear:134

  • Hyperactivity
    • Increase with naive caffeine input84
    • no increase in hyperactivity after habituation to caffeine (60 mg/kg/day - far above the recommended dosage for humans of 5.7 mg/kg/day), not even by increasing the dose84
  • Impulsiveness

PFC neurons of the SHR show fewer neurite branches, a shorter maximum neurite length and a lower axonal growth than PFC neurons of the WKY.
Caffeine restored neurite branching and elongation in SHR neurons via both PKA and PI3K signaling. The A2A agonist CGS 21680 improved neurite branching via PKA signaling. The selective A2A antagonist SCH 58261 restored axonal growth of SHR neurons via PI3K signaling (not PKA signaling)33

Unfortunately, caffeine induces strong tolerance and increased sensitivity of adenosine receptors, so it is doubtful that caffeine has any benefit for ADHD beyond alternating use of small doses (1-2 cups of coffee every 2 days).

7.3. Adenosine antagonist theophylline possibly equivalent to MPH for ADHD

A smaller study found an equivalent effect of theophylline compared to MPH in children with ADHD. As this study ran for 6 weeks, this could indicate that theophylline has a lower tolerance than caffeine,

7.4. ADHD drug viloxazine increases theophylline

Viloxazine significantly increases the plasma level of the adenosine A1 and A2 antagonist theophylline.137138
It is quite conceivable that part of the effect of viloxazine in ADHD could be due to the dopamine-increasing effect of the adenosine antagonist theophylline.

More about viloxazine at =&gt Viloxazine for ADHD

7.5. A2A receptor gene and ADHD

A study found a possible link between the polymorphism SNP rs35320474 of the ADORA2A gene (A2A receptor gene) and ADHD34
A combination of certain A2A and D2 receptor genes appears to increase the risk of anxiety disorders in children with ADHD.139

7.6. Adenosine system altered in SHR

The Spontaneously Hypertensive Rat (SHR) is a genetic animal model for ADHD-HI with hyperactivity.
An altered adenosine system was detected in the SHR. Find out more at =&gt Adenosine system altered in SHR In the article =&gt ADHD in an animal model

The amount of A2A receptors in frontocortical axon terminals is increased in SHR72

Adenosine antagonists improve various ADHD symptoms in SHR

  • Caffeine (non-selective A1 and A2A antagonist)
    • Object recognition140
    • social recognition141
    • spatial learning142
    • no influence on high blood pressure142
  • DPCPX (8-cyclopenthyl-1,3-dipropylxanthine, A1 antagonist)
    • Object recognition140
    • no influence on high blood pressure142
  • ZM241385 (4-(2-[7-amino-2-[2-furyl][1,2,4]triazolo-[2,3-a][1,3,5]triazin-5-yl-amino]ethyl) phenol, A2A-Antagonist)
    • Object recognition140
    • social recognition141
    • no influence on high blood pressure142

Chronic caffeine input72

  • normalized the dopaminergic function
  • improved memory and attention deficits
  • induced upregulation of A2AARs in frontocortical nerve endings

Chronic administration of caffeine or MPH before puberty improved object recognition in adult SHR, while the same treatment worsened it in adult Wistar rats143

There is evidence of an interaction between the cannabinoid and adenosine systems in relation to impulsive behavior in SHR:144

  • WIN55212-2 (cannabinoid receptor agonist) increased impulsive behavior
  • acute pre-treatment with caffeine canceled this out
  • chronic caffeine intake increased impulsivity

7.7. Some comorbidities associated with ADHD show elevated adenosine levels

Asthma, inflammatory disorders (such as neurodermatitis) and diabetes often occur comorbidly with ADHD. These 3 disorders are often associated with highly elevated adenosine levels7
A combination of these comorbidities with ADHD therefore increasingly indicates an elevated adenosine level. While we know that dopamine deficiency can be a consequence of elevated adenosine in ADHD and that ADHD can therefore be a consequence of elevated adenosine (although there are many other possible causes), we do not know the causality in asthma, neurodermatitis and diabetes.

7.8. Some ADHD risk factors show increased adenosine levels

  • Pre-eclampsia (gestational gestosis, high blood pressure during pregnancy) increases the risk of ADHD by 30 to 188%. Pre-eclampsia is associated with changes in the adenosine system including adenosine transporters and adenosine receptors. SHR are born in a pre-eclampsia-like situation due to adult maternal hypertension. Caffeine (an adenosine antagonist) in 7-day-old SHR prevented the negative consequences of preeclampsia (hyperactivity, worsened social interaction, worsened contextual fear conditioning), while it enhanced these symptoms in Wistar rats145
  • High levels of the (weak) adenosine antagonist theobromine correlated negatively with pre-eclampsia in humans.146
  • Hypoxia (lack of oxygen) increases adenosine.
    • Adenosine antagonists can prevent or remedy the negative consequences of hypoxia (see above).
    • Methylphenidate can also eliminate the ADHD symptoms triggered by hypoxia (here: addictive behavior).147

7.9. ADHD symptoms that are promoted by adenosine

7.9.1. Hypermotor skills

striatopallidal: motor control

  • especially in GABAergic medium spiny neurons (MSN) of the indirect pathway12148
  • mainly via A2A, hardly ever via A112148

7.9.2. Motivation problems

Adenosine inhibits dopaminergic neurotransmission and thus the reward system in the striatum via

primarily in the nucleus accumbens,

  • here mainly in GABAergic medium spiny neurons (MSN) of the indirect pathway12

8. Outlook - Adenosine (A2A) antagonists as ADHD medication?

There is some evidence that adenosine antagonists can have positive effects on ADHD symptoms. They are therefore being considered as ADHD medications.149 In addition to the empirical evidence of increased caffeine consumption by ADHD sufferers reported above, this is based on neurophysiological findings.

A2A / D2 heteromers are involved in reward mechanisms. They are found in particular in GABAergic neurons of the ventral striatopallidal area, which are responsible for reward and motivational effects8

A2A antagonists can have a similar effect to psychostimulants at low doses, as long as they are not given together with A2A agonists.150 151 The fact that they could potentially act as a drug at very high doses corresponds to the stimulants methylphenidate and amphetamine drugs used as ADHD medications: here too, the dose makes the poison.
A blockade of A2A receptors led (here: in cocaine-dependent subjects) to an increase in dopamine in the striatum, which triggered a strong stimulation of the PFC.152 This corresponds to the desired effect pathways of ADHD drugs at medical doses.

At the same time, A2A antagonists offer the potential of addiction therapy or withdrawal support and the treatment of children with consequences of fetal drug intoxication.153 Systemic administration of A2A antagonists reduced addictive behavior in rats with respect to heroin and THC, but not with respect to cocaine.154155

So far, adenosine antagonists have only been researched from the perspective of Parkinson’s treatment. It is to be hoped that research will also focus on their use in relation to ADHD.

Istradefylline, the first A2A antagonist for the treatment of Parkinson’s disease, was approved in the USA in 2019 (brand name: Nourianz®).44 The EMA has so far refused approval for Europe, citing contradictory study results.


  1. Borea, Gessi, Merighi, Vincenzi, Varani (2018): Pharmacology of Adenosine Receptors: The State of the Art. Physiol Rev. 2018 Jul 1;98(3):1591-1625. doi: 10.1152/physrev.00049.2017. PMID: 29848236. REVIEW

  2. Quiroz, Orrú, Rea, Ciudad-Roberts, Yepes, Britt, Ferré (2016): Local Control of Extracellular Dopamine Levels in the Medial Nucleus Accumbens by a Glutamatergic Projection from the Infralimbic Cortex. J Neurosci. 2016 Jan 20;36(3):851-9. doi: 10.1523/JNEUROSCI.2850-15.2016.

  3. Hänsel, Sticher (2010): Pharmakognosie - Phytopharmazie, 9. Auflage, S. 1349

  4. Wenzel (2012): Koffeinhaltige Lebensmittel; Eine Zusammenstellung zur Geschichte, Verwendung und Wirkung von koffeinhaltigen Lebensmitteln; Hochschule Weihenstephan

  5. Borbély, Daan, Wirz-Justice, Deboer (2016): The two-process model of sleep regulation: a reappraisal. J Sleep Res. 2016 Apr;25(2):131-43. doi: 10.1111/jsr.12371. PMID: 26762182.

  6. van Diepen, Lucassen, Yasenkov, Groenen, Ijzerman, Meijer, Deboer (2014): Caffeine increases light responsiveness of the mouse circadian pacemaker. Eur J Neurosci. 2014 Nov;40(10):3504-11. doi: 10.1111/ejn.12715. PMID: 25196050.

  7. Borea, Gessi, Merighi, Vincenzi, Varani (2017): Pathological overproduction: the bad side of adenosine. Br J Pharmacol. 2017 Jul;174(13):1945-1960. doi: 10.1111/bph.13763. PMID: 28252203; PMCID: PMC6398520. REVIEW

  8. Ballesteros-Yáñez, Castillo, Merighi, Gessi (2018): The Role of Adenosine Receptors in Psychostimulant Addiction. Front Pharmacol. 2018 Jan 10;8:985. doi: 10.3389/fphar.2017.00985. PMID: 29375384; PMCID: PMC5767594. REVIEW

  9. Chen, Lee, Chern (2014): Adenosine receptor neurobiology: overview. Int Rev Neurobiol. 2014;119:1-49. doi: 10.1016/B978-0-12-801022-8.00001-5. PMID: 25175959. REVIEW

  10. Hu, Adebiyi, Luo, Sun, Le, Zhang, Wu, Zhao, Karmouty-Quintana, Liu, Huang, Wen, Zaika, Mamenko, Pochynyuk, Kellems, Eltzschig, Blackburn, Walters, Huang, Hu, Xia (2016): Sustained Elevated Adenosine via ADORA2B Promotes Chronic Pain through Neuro-immune Interaction. Cell Rep. 2016 Jun 28;16(1):106-119. doi: 10.1016/j.celrep.2016.05.080. PMID: 27320922; PMCID: PMC5662192.

  11. Ferré, Agnati, Ciruela, Lluis, Woods, Fuxe, Franco (2007): Neurotransmitter receptor heteromers and their integrative role in ‘local modules’: the striatal spine module. Brain Res Rev. 2007 Aug;55(1):55-67. doi: 10.1016/j.brainresrev.2007.01.007. PMID: 17408563; PMCID: PMC2039920.

  12. Prasad, de Vries, Elsinga, Dierckx, van Waarde (2021): Allosteric Interactions between Adenosine A2A and Dopamine D2 Receptors in Heteromeric Complexes: Biochemical and Pharmacological Characteristics, and Opportunities for PET Imaging. Int J Mol Sci. 2021 Feb 9;22(4):1719. doi: 10.3390/ijms22041719. PMID: 33572077; PMCID: PMC7915359. REVIEW

  13. Kennedy (2021): ATP as a cotransmitter in sympathetic and parasympathetic nerves - another Burnstock legacy. Auton Neurosci. 2021 Nov;235:102860. doi: 10.1016/j.autneu.2021.102860. PMID: 34340045.

  14. Illes P, Ulrich H, Chen JF, Tang Y (2023): Purinergic receptors in cognitive disturbances. Neurobiol Dis. 2023 Jul 13;185:106229. doi: 10.1016/j.nbd.2023.106229. PMID: 37453562. REVIEW

  15. Chen, Eltzschig, Fredholm (2013): Adenosine receptors as drug targets–what are the challenges? Nat Rev Drug Discov. 2013 Apr;12(4):265-86. doi: 10.1038/nrd3955. PMID: 23535933; PMCID: PMC3930074.

  16. Borycz, Pereira, Melani, Rodrigues, Köfalvi, Panlilio, Pedata, Goldberg, Cunha, Ferré (2007): Differential glutamate-dependent and glutamate-independent adenosine A1 receptor-mediated modulation of dopamine release in different striatal compartments. J Neurochem. 2007 Apr;101(2):355-63. doi: 10.1111/j.1471-4159.2006.04386.x. PMID: 17254024.

  17. Chen (2014): Adenosine receptor control of cognition in normal and disease. Int Rev Neurobiol. 2014;119:257-307. doi: 10.1016/B978-0-12-801022-8.00012-X. PMID: 25175970. REVIEW

  18. Pasquini, Contri, Merighi, Gessi, Borea, Varani, Vincenzi (2022): Adenosine Receptors in Neuropsychiatric Disorders: Fine Regulators of Neurotransmission and Potential Therapeutic Targets. Int J Mol Sci. 2022 Jan 22;23(3):1219. doi: 10.3390/ijms23031219. PMID: 35163142; PMCID: PMC8835915. REVIEW

  19. Kashfi, Ghaedi, Baharvand, Nasr-Esfahani, Javan (2017): A1 Adenosine Receptor Activation Modulates Central Nervous System Development and Repair. Mol Neurobiol. 2017 Dec;54(10):8128-8139. doi: 10.1007/s12035-016-0292-6. PMID: 27889899. REVIEW

  20. Carrettiero, Almeida, Fior-Chadi (2008): Adenosine modulates alpha2-adrenergic receptors within specific subnuclei of the nucleus tractus solitarius in normotensive and spontaneously hypertensive rats. Hypertens Res. 2008 Dec;31(12):2177-86. doi: 10.1291/hypres.31.2177. PMID: 19139607.

  21. Prediger, Batista, Takahashi (2004): Adenosine A1 receptors modulate the anxiolytic-like effect of ethanol in the elevated plus-maze in mice. Eur J Pharmacol. 2004 Sep 19;499(1-2):147-54. doi: 10.1016/j.ejphar.2004.07.106. PMID: 15363961.

  22. Prediger, da Silva, Batista, Bittencourt, Takahashi (2006): Activation of adenosine A1 receptors reduces anxiety-like behavior during acute ethanol withdrawal (hangover) in mice. Neuropsychopharmacology. 2006 Oct;31(10):2210-20. doi: 10.1038/sj.npp.1301001. PMID: 16407902.

  23. Massie, O’Connor, Metra, Ponikowski, Teerlink, Cotter, Weatherley, Cleland, Givertz, Voors, DeLucca, Mansoor, Salerno, Bloomfield, Dittrich (2010): PROTECT Investigators and Committees. Rolofylline, an adenosine A1-receptor antagonist, in acute heart failure. N Engl J Med. 2010 Oct 7;363(15):1419-28. doi: 10.1056/NEJMoa0912613. PMID: 20925544.

  24. Poleszak, Malec (2002): Cocaine-induced hyperactivity is more influenced by adenosine receptor agonists than amphetamine-induced hyperactivity. Pol J Pharmacol. 2002 Jul-Aug;54(4):359-66. PMID: 12523489.

  25. Bruns, Katims, Annau, Snyder, Daly (1983): Adenosine receptor interactions and anxiolytics. Neuropharmacology. 1983 Dec;22(12B):1523-9. doi: 10.1016/0028-3908(83)90121-1. PMID: 6199685. REVIEW

  26. Wenzel (2012): Koffein-haltige Lebensmittel; Eine Zusammenstellung zur Geschichte, Verwendung und Wirkung von coffeinhaltigen Lebensmitteln; Hochschule Weihenstephan

  27. Sun, Bachhawat, Chu, Wood, Ceska, Sands, Mercier, Lebon, Kobilka, Kobilka (2017): Crystal structure of the adenosine A2A receptor bound to an antagonist reveals a potential allosteric pocket. Proc Natl Acad Sci U S A. 2017 Feb 21;114(8):2066-2071. doi: 10.1073/pnas.1621423114. PMID: 28167788; PMCID: PMC5338372.

  28. Barnes (2010): Theophylline. Pharmaceuticals (Basel). 2010 Mar 18;3(3):725-747. doi: 10.3390/ph3030725. PMID: 27713276; PMCID: PMC4033977. REVIEW

  29. Quiroz C, Gulyani S, Ruiqian W, Bonaventura J, Cutler R, Pearson V, Allen RP, Earley CJ, Mattson MP, Ferré S (2016): Adenosine receptors as markers of brain iron deficiency: Implications for Restless Legs Syndrome. Neuropharmacology. 2016 Dec;111:160-168. doi: 10.1016/j.neuropharm.2016.09.002. PMID: 27600688; PMCID: PMC5056844.

  30. Rodrigues MS, Ferreira SG, Quiroz C, Earley CJ, García-Borreguero D, Cunha RA, Ciruela F, Köfalvi A, Ferré S (2022): Brain Iron Deficiency Changes the Stoichiometry of Adenosine Receptor Subtypes in Cortico-Striatal Terminals: Implications for Restless Legs Syndrome. Molecules. 2022 Feb 23;27(5):1489. doi: 10.3390/molecules27051489. PMID: 35268590; PMCID: PMC8911604.

  31. Wydra, Gawliński, Gawlińska, Frankowska, Borroto-Escuela, Fuxe, Filip (2020): Adenosine A2AReceptors in Substance Use Disorders: A Focus on Cocaine. Cells. 2020 Jun 1;9(6):1372. doi: 10.3390/cells9061372. PMID: 32492952; PMCID: PMC7348840.

  32. Ravani, Vincenzi, Bortoluzzi, Padovan, Pasquini, Gessi, Merighi, Borea, Govoni, Varani (2017): Role and Function of A2A and A₃ Adenosine Receptors in Patients with Ankylosing Spondylitis, Psoriatic Arthritis and Rheumatoid Arthritis. Int J Mol Sci. 2017 Mar 24;18(4):697. doi: 10.3390/ijms18040697. PMID: 28338619; PMCID: PMC5412283.

  33. Alves, Almeida, Marques, Faé, Machado, Oliveira, Portela, Porciúncula (2020): Caffeine and adenosine A2A receptors rescue neuronal development in vitro of frontal cortical neurons in a rat model of attention deficit and hyperactivity disorder. Neuropharmacology. 2020 Apr;166:107782. doi: 10.1016/j.neuropharm.2019.107782. PMID: 31756336.

  34. Molero, Gumpert, Serlachius, Lichtenstein, Walum, Johansson, Anckarsäter, Westberg, Eriksson, Halldner (2013): A study of the possible association between adenosine A2A receptor gene polymorphisms and attention-deficit hyperactivity disorder traits. Genes Brain Behav. 2013 Apr;12(3):305-10. doi: 10.1111/gbb.12015. PMID: 23332182.

  35. Takahashi, Pamplona, Prediger (2008): Adenosine receptor antagonists for cognitive dysfunction: a review of animal studies. Front Biosci. 2008 Jan 1;13:2614-32. doi: 10.2741/2870. PMID: 17981738. REVIEW

  36. Coelho, Alves, Canas, Valadas, Shmidt, Batalha, Ferreira, Ribeiro, Bader, Cunha, do Couto, Lopes (2014): Overexpression of Adenosine A2A Receptors in Rats: Effects on Depression, Locomotion, and Anxiety. Front Psychiatry. 2014 Jun 13;5:67. doi: 10.3389/fpsyt.2014.00067. PMID: 24982640; PMCID: PMC4055866.

  37. Hohoff, Mullings, Heatherley, Freitag, Neumann, Domschke, Krakowitzky, Rothermundt, Keck, Erhardt, Unschuld, Jacob, Fritze, Bandelow, Maier, Holsboer, Rogers, Deckert (2010): Adenosine A(2A) receptor gene: evidence for association of risk variants with panic disorder and anxious personality. J Psychiatr Res. 2010 Oct;44(14):930-7. doi: 10.1016/j.jpsychires.2010.02.006. PMID: 20334879.

  38. Ledent, Vaugeois, Schiffmann, Pedrazzini, El Yacoubi, Vanderhaeghen, Costentin, Heath, Vassart, Parmentier (1997): Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2a receptor. Nature. 1997 Aug 14;388(6643):674-8. doi: 10.1038/41771. PMID: 9262401.

  39. Filip, Frankowska, Zaniewska, Przegaliński, Muller, Agnati, Franco, Roberts, Fuxe (2006): Involvement of adenosine A2A and dopamine receptors in the locomotor and sensitizing effects of cocaine. Brain Res. 2006 Mar 10;1077(1):67-80. doi: 10.1016/j.brainres.2006.01.038. PMID: 16516871.

  40. Prasad, de Vries, Sijbesma, Garcia-Varela, Vazquez-Matias, Moraga-Amaro, Willemsen, Dierckx, van Waarde (2022): Impact of an Adenosine A2A Receptor Agonist and Antagonist on Binding of the Dopamine D2 Receptor Ligand [11C]raclopride in the Rodent Striatum. Mol Pharm. 2022 Jul 18. doi: 10.1021/acs.molpharmaceut.2c00450. PMID: 35849844.

  41. Ferre, von Euler, Johansson, Fredholm, Fuxe (1991): Stimulation of high-affinity adenosine A2 receptors decreases the affinity of dopamine D2 receptors in rat striatal membranes. Proc Natl Acad Sci U S A. 1991 Aug 15;88(16):7238-41. doi: 10.1073/pnas.88.16.7238. PMID: 1678519; PMCID: PMC52269.

  42. da Silva, Gabriel-Costa, Sudo, Wang, Groban, Ferraz, Nascimento, Fraga, Barreiro, Zapata-Sudo (2017): Adenosine A2A receptor agonist prevents cardiac remodeling and dysfunction in spontaneously hypertensive male rats after myocardial infarction. Drug design, development and therapy, 11, 553–562. https://doi.org/10.2147/DDDT.S113289

  43. Pinna (2014): Adenosine A2A receptor antagonists in Parkinson’s disease: progress in clinical trials from the newly approved istradefylline to drugs in early development and those already discontinued. CNS Drugs. 2014 May;28(5):455-74. doi: 10.1007/s40263-014-0161-7. PMID: 24687255.

  44. Chen, Cunha (2020): The belated US FDA approval of the adenosine A2A receptor antagonist istradefylline for treatment of Parkinson’s disease. Purinergic Signal. 2020 Jun;16(2):167-174. doi: 10.1007/s11302-020-09694-2. PMID: 32236790; PMCID: PMC7367999.

  45. EMA: Nouryant

  46. EMA (2021): Assessment report Nouryant

  47. Jenner (2014): An overview of adenosine A2A receptor antagonists in Parkinson’s disease. Int Rev Neurobiol. 2014;119:71-86. doi: 10.1016/B978-0-12-801022-8.00003-9. PMID: 25175961.

  48. O’Neill, Brown (2007): The effect of striatal dopamine depletion and the adenosine A2A antagonist KW-6002 on reversal learning in rats. Neurobiol Learn Mem. 2007 Jul;88(1):75-81. doi: 10.1016/j.nlm.2007.03.003. PMID: 17467309.

  49. Merighi S, Borea PA, Varani K, Vincenzi F, Jacobson KA, Gessi S. A2A Adenosine Receptor Antagonists in Neurodegenerative Diseases. Curr Med Chem. 2022;29(24):4138-4151. doi: 10.2174/0929867328666211129122550. PMID: 34844537; PMCID: PMC9148371.

  50. Ko, Camus, Li, Yang, McGuire, Pioli, Bezard (2016): An evaluation of istradefylline treatment on Parkinsonian motor and cognitive deficits in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated macaque models. Neuropharmacology. 2016 Nov;110(Pt A):48-58. doi: 10.1016/j.neuropharm.2016.07.012. PMID: 27424102.

  51. Li, Silva, Real, Wang, Rial, Li, Payen, Zhou, Muller, Tomé, Cunha, Chen (2015): Inactivation of adenosine A2A receptors reverses working memory deficits at early stages of Huntington’s disease models. Neurobiol Dis. 2015 Jul;79:70-80. doi: 10.1016/j.nbd.2015.03.030. PMID: 25892655.

  52. Zhou, Zhu, Shu, Du, Song, Wang, Zheng, Cai, Chen, He (2009): Preferential enhancement of working memory in mice lacking adenosine A(2A) receptors. Brain Res. 2009 Dec 15;1303:74-83. doi: 10.1016/j.brainres.2009.09.082. PMID: 19785999.

  53. Canas, Porciúncula, Cunha, Silva, Machado, Oliveira, Oliveira, Cunha (2009): Adenosine A2A receptor blockade prevents synaptotoxicity and memory dysfunction caused by beta-amyloid peptides via p38 mitogen-activated protein kinase pathway. J Neurosci. 2009 Nov 25;29(47):14741-51. doi: 10.1523/JNEUROSCI.3728-09.2009. PMID: 19940169; PMCID: PMC6665997.

  54. Mingote, Font, Farrar, Vontell, Worden, Stopper, Port, Sink, Bunce, Chrobak, Salamone (2008): Nucleus accumbens adenosine A2A receptors regulate exertion of effort by acting on the ventral striatopallidal pathway. J Neurosci. 2008 Sep 3;28(36):9037-46. doi: 10.1523/JNEUROSCI.1525-08.2008. PMID: 18768698; PMCID: PMC2806668.

  55. Cho, Choi, Kim, Kim (2018): Association of coffee consumption and non-motor symptoms in drug-naïve, early-stage Parkinson’s disease. Parkinsonism Relat Disord. 2018 May;50:42-47. doi: 10.1016/j.parkreldis.2018.02.016. PMID: 29449185.

  56. Li, He, Chen, Pu, Chen, Li, Li, Li, Huang, Li, Chen (2016): Optogenetic Activation of Adenosine A2A Receptor Signaling in the Dorsomedial Striatopallidal Neurons Suppresses Goal-Directed Behavior. Neuropsychopharmacology. 2016 Mar;41(4):1003-13. doi: 10.1038/npp.2015.227. PMID: 26216520; PMCID: PMC4748425.

  57. Wei, Augusto, Gomes, Singer, Wang, Boison, Cunha, Yee, Chen (2013): Regulation of fear responses by striatal and extrastriatal adenosine A2A receptors in forebrain. Biol Psychiatry. 2014 Jun 1;75(11):855-63. doi: 10.1016/j.biopsych.2013.05.003. PMID: 23820821; PMCID: PMC4058554.

  58. Nagayama, Kano, Murakami, Ono, Hamada, Toda, Sengoku, Shimo, Hattori (2019): Effect of istradefylline on mood disorders in Parkinson’s disease. J Neurol Sci. 2019 Jan 15;396:78-83. doi: 10.1016/j.jns.2018.11.005. PMID: 30423541.

  59. Ning, Yang, Chen, Xiong, Zhang, Li, Zhao, Chen, Liu, Peng, Wang, Chen, Zhou (2013): Adenosine A2A receptor deficiency alleviates blast-induced cognitive dysfunction. J Cereb Blood Flow Metab. 2013 Nov;33(11):1789-98. doi: 10.1038/jcbfm.2013.127. PMID: 23921902; PMCID: PMC3824177.

  60. Batalha, Pego, Fontinha, Costenla, Valadas, Baqi, Radjainia, Müller, Sebastião, Lopes (2013): Adenosine A(2A) receptor blockade reverts hippocampal stress-induced deficits and restores corticosterone circadian oscillation. Mol Psychiatry. 2013 Mar;18(3):320-31. doi: 10.1038/mp.2012.8. PMID: 22371048.

  61. Kaster, Machado, Silva, Nunes, Ardais, Santana, Baqi, Müller, Rodrigues, Porciúncula, Chen, Tomé, Agostinho, Canas, Cunha (2015): Caffeine acts through neuronal adenosine A2A receptors to prevent mood and memory dysfunction triggered by chronic stress. Proc Natl Acad Sci U S A. 2015 Jun 23;112(25):7833-8. doi: 10.1073/pnas.1423088112. PMID: 26056314; PMCID: PMC4485143.

  62. Dall’Igna, Fett, Gomes, Souza, Cunha, Lara (2006): Caffeine and adenosine A(2a) receptor antagonists prevent beta-amyloid (25-35)-induced cognitive deficits in mice. Exp Neurol. 2007 Jan;203(1):241-5. doi: 10.1016/j.expneurol.2006.08.008. PMID: 17007839.

  63. Bonaventura, Navarro, Casadó-Anguera, Azdad, Rea, Moreno, Brugarolas, Mallol, Canela, Lluís, Cortés, Volkow, Schiffmann, Ferré, Casadó (2015): Allosteric interactions between agonists and antagonists within the adenosine A2A receptor-dopamine D2 receptor heterotetramer. Proc Natl Acad Sci U S A. 2015 Jul 7;112(27):E3609-18. doi: 10.1073/pnas.1507704112. PMID: 26100888; PMCID: PMC4500251.

  64. Fahim, Mustafa (2001): Evidence for the presence of A(1) adenosine receptors in the aorta of spontaneously hypertensive rats. Br J Pharmacol. 2001 Dec;134(8):1760-6. doi: 10.1038/sj.bjp.0704433. PMID: 11739253; PMCID: PMC1572910.

  65. Bundesredierung: Koffein: Die Dosis macht’s

  66. Müller, Jacobson (2011): Xanthines as adenosine receptor antagonists. Handb Exp Pharmacol. 2011;(200):151-99. doi: 10.1007/978-3-642-13443-2_6. PMID: 20859796; PMCID: PMC3882893.

  67. Yamada, Kobayashi, Mori, Jenner, Kanda (2013): Antidepressant-like activity of the adenosine A(2A) receptor antagonist, istradefylline (KW-6002), in the forced swim test and the tail suspension test in rodents. Pharmacol Biochem Behav. 2013 Dec;114-115:23-30. doi: 10.1016/j.pbb.2013.10.022. PMID: 24201052.

  68. Cappelletti, Piacentino, Sani, Aromatario (2015): Caffeine: cognitive and physical performance enhancer or psychoactive drug? Curr Neuropharmacol. 2015 Jan;13(1):71-88. doi: 10.2174/1570159X13666141210215655. Erratum in: Curr Neuropharmacol. 2015;13(4):554. Daria, Piacentino [corrected to Piacentino, Daria]. PMID: 26074744; PMCID: PMC4462044. REVIEW

  69. Rogers, Smith (2011): Caffeine, mood and cognition. In: Benton (Hrsg.): Lifetime Nutritional Influences on Cognition, Behaviour and Psychiatric Illness. Woodhead Publishing Ltd; 2011. Seiten 251–271.

  70. Matissek (2016): Alkaloidhaltige Lebensmittel. In: Matissek, Baltes: Lebensmittelchemie, 8. Aufl. S. 557

  71. Matissek (2016): Alkaloidhaltige Lebensmittel. In: Matissek, Baltes: Lebensmittelchemie, 8. Aufl. S. 564

  72. Pandolfo, Machado, Köfalvi, Takahashi, Cunha (2013): Caffeine regulates frontocorticostriatal dopamine transporter density and improves attention and cognitive deficits in an animal model of attention deficit hyperactivity disorder. Eur Neuropsychopharmacol. 2013 Apr;23(4):317-28. doi: 10.1016/j.euroneuro.2012.04.011. PMID: 22561003.

  73. Kubrusly, da Rosa Valli, Ferreira, de Moura, Borges-Martins, Martins, Ferreira, Sathler, de Melo Reis, Ferreira, Manhães, Dos Santos Pereira (2021): Caffeine Improves GABA Transport in the Striatum of Spontaneously Hypertensive Rats (SHR). Neurotox Res. 2021 Dec;39(6):1946-1958. doi: 10.1007/s12640-021-00423-0.PMID: 34637050.

  74. Rogers, Richardson, Dernoncourt (1995): Caffeine use: is there a net benefit for mood and psychomotor performance? Neuropsychobiology. 1995;31(4):195-9. doi: 10.1159/000119192. PMID: 7659200.

  75. Gilliland, Andress (2006): Ad lib caffeine consumption, symptoms of caffeinism, and academic performance. Am J Psychiatry. 1981 Apr;138(4):512-4. doi: 10.1176/ajp.138.4.512. PMID: 7212112.

  76. Fredholm, Bättig, Holmén, Nehlig, Zvartau (1999): Actions of caffeine in the brain with special reference to factors that contribute to its widespread use. Pharmacol Rev. 1999 Mar;51(1):83-133. PMID: 10049999.

  77. Robertson, Wade, Workman, Woosley, Oates (1981): Tolerance to the humoral and hemodynamic effects of caffeine in man. J Clin Invest. 1981 Apr;67(4):1111-7. doi: 10.1172/jci110124. PMID: 7009653; PMCID: PMC370671.

  78. Holtzman, Finn (1988): Tolerance to behavioral effects of caffeine in rats. Pharmacol Biochem Behav. 1988 Feb;29(2):411-8. doi: 10.1016/0091-3057(88)90179-7. PMID: 3362935.

  79. [Fredholm (1982): Adenosine actions and adenosine receptors after 1 week treatment with caffeine. Acta Physiol Scand. 1982 Jun;115(2):283-6. doi: 10.1111/j.1748-1716.1982.tb07078.x. PMID: 6291335.)](https://pubmed.ncbi.nlm.nih.gov/6291335/

  80. Johansson, Ahlberg, van der Ploeg, Brené, Lindefors, Persson, Fredholm (1993): Effect of long term caffeine treatment on A1 and A2 adenosine receptor binding and on mRNA levels in rat brain. Naunyn Schmiedebergs Arch Pharmacol. 1993 Apr;347(4):407-14. doi: 10.1007/BF00165391. PMID: 8510768.

  81. Johansson, Georgiev, Lindström, Fredholm (1997): A1 and A2A adenosine receptors and A1 mRNA in mouse brain: effect of long-term caffeine treatment. Brain Res. 1997 Jul 11;762(1-2):153-64. doi: 10.1016/s0006-8993(97)00378-8. PMID: 9262169.

  82. [Bona, Adén, Fredholm, Hagberg (1995): The effect of long term caffeine treatment on hypoxic-ischemic brain damage in the neonate. Pediatr Res. 1995 Sep;38(3):312-8. doi: 10.1203/00006450-199509000-00007. PMID: 7494652.)](https://pubmed.ncbi.nlm.nih.gov/7494652/

  83. Johansson, Georgiev, Kuosmanen, Fredholm (1996): Long-term treatment with some methylxanthines decreases the susceptibility to bicuculline- and pentylenetetrazol-induced seizures in mice. Relationship to c-fos expression and receptor binding. Eur J Neurosci. 1996 Dec;8(12):2447-58. doi: 10.1111/j.1460-9568.1996.tb01539.x. PMID: 8996794.

  84. Holtzman, Mante, Minneman (1991): Role of adenosine receptors in caffeine tolerance. J Pharmacol Exp Ther. 1991 Jan;256(1):62-8. PMID: 1846425.

  85. Chern, Lai, Fong, Liang (1993): Multiple mechanisms for desensitization of A2a adenosine receptor-mediated cAMP elevation in rat pheochromocytoma PC12 cells. Mol Pharmacol. 1993 Nov;44(5):950-8. PMID: 8246918.

  86. Griffiths, Mumford (1995): Caffeine Reinforcement, Discrimination, Tolerance and Physical Dependence in Laboratory Animals and Humans. In: Schuster, Kihar (Hrsg.): Pharmacological Aspects of Drug Dependence. Toward an Integrated Neurobehavioral Approach. Springer. S. 315 - 369

  87. Biaggioni, Paul, Puckett, Arzubiaga (1991): Caffeine and theophylline as adenosine receptor antagonists in humans. J Pharmacol Exp Ther. 1991 Aug;258(2):588-93. PMID: 1865359.

  88. Ahlijanian, Takemori (1986): Cross-tolerance studies between caffeine and (-)-N6-(phenylisopropyl)-adenosine (PIA) in mice. Life Sci. 1986 Feb 17;38(7):577-88. doi: 10.1016/0024-3205(86)90051-2. PMID: 3003486.

  89. Green, Stiles (1986): Chronic caffeine ingestion sensitizes the A1 adenosine receptor-adenylate cyclase system in rat cerebral cortex. J Clin Invest. 1986 Jan;77(1):222-7. doi: 10.1172/JCI112280. PMID: 3003150; PMCID: PMC423330.

  90. von Borstel, Wurtman, Conlay (1983): Chronic caffeine consumption potentiates the hypotensive action of circulating adenosine. Life Sci. 1983 Mar 7;32(10):1151-8. doi: 10.1016/0024-3205(83)90121-2. PMID: 6827895.)

  91. Finn, Holtzman (1986): Tolerance to caffeine-induced stimulation of locomotor activity in rats. J Pharmacol Exp Ther. 1986 Aug;238(2):542-6. PMID: 3735131.

  92. Holtzman (1983): Complete, reversible, drug-specific tolerance to stimulation of locomotor activity by caffeine. Life Sci. 1983 Aug 22;33(8):779-87. doi: 10.1016/0024-3205(83)90784-1. PMID: 6888193.

  93. Carney (1982): Effects of caffeine, theophylline and theobromine on scheduled controlled responding in rats. Br J Pharmacol. 1982 Mar;75(3):451-4. doi: 10.1111/j.1476-5381.1982.tb09161.x. PMID: 7066599; PMCID: PMC2071561.

  94. Mumford, Neill, Holtzman (1988): Caffeine elevates reinforcement threshold for electrical brain stimulation: tolerance and withdrawal changes. Brain Res. 1988 Aug 30;459(1):163-7. doi: 10.1016/0006-8993(88)90298-3. PMID: 3167574.

  95. Carroll, Hagen, Asencio, Brauer (1988): Behavioral dependence on caffeine and phencyclidine in rhesus monkeys: interactive effects. Pharmacol Biochem Behav. 1988 Dec;31(4):927-32. doi: 10.1016/0091-3057(88)90406-6. PMID: 3252284.

  96. Sinton, Petitjean (1989): The influence of chronic caffeine administration on sleep parameters in the cat. Pharmacol Biochem Behav. 1989 Feb;32(2):459-62. doi: 10.1016/0091-3057(89)90179-2. PMID: 2727004.

  97. Vitiello, Woods (1977): Evidence for withdrawal from caffeine by rats. Pharmacol Biochem Behav. 1977 May;6(5):553-5. doi: 10.1016/0091-3057(77)90116-2. PMID: 561407.

  98. Brunstrom (2004): Does dietary learning occur outside awareness? Conscious Cogn. 2004 Sep;13(3):453-70. doi: 10.1016/j.concog.2004.05.004. PMID: 15336241.

  99. Couturier, Laman, van Duijn, van Duijn (1997): Influence of caffeine and caffeine withdrawal on headache and cerebral blood flow velocities. Cephalalgia. 1997 May;17(3):188-90. doi: 10.1046/j.1468-2982.1997.1703188.x. PMID: 9170342.

  100. Nabbi-Schroeter, Elmenhorst, Oskamp, Laskowski, Bauer, Kroll (2018): Effects of Long-Term Caffeine Consumption on the Adenosine A1 Receptor in the Rat Brain: an In Vivo PET Study with [18F]CPFPX. Mol Imaging Biol. 2018 Apr;20(2):284-291. doi: 10.1007/s11307-017-1116-4. PMID: 28895043.

  101. Matera, Page, Cazzola (2017): Doxofylline is not just another theophylline! Int J Chron Obstruct Pulmon Dis. 2017 Dec 5;12:3487-3493. doi: 10.2147/COPD.S150887. PMID: 29255355; PMCID: PMC5723117.

  102. Spina, Page (2017): Xanthines and Phosphodiesterase Inhibitors. Handb Exp Pharmacol. 2017;237:63-91. doi: 10.1007/164_2016_71. PMID: 27844172.

  103. Jalal, Collin (1976): ESTIMATION OF CAFFEINE, THEOPHYLLINE AND THEOBROMINE IN PLANT MATERIAL

  104. Hänsel, Sticher (2010): Pharmakognosie - Phytopharmazie, 9. Auflage, S. 1359

  105. Barnes PJ. Theophylline. Am J Respir Crit Care Med. 2013 Oct 15;188(8):901-6. doi: 10.1164/rccm.201302-0388PP. PMID: 23672674.

  106. Polson, Krzanowski, Goldman, Szentivanyi (1978): Inhibition of human pulmonary phosphodiesterase activity by therapeutic levels of theophylline. Clin Exp Pharmacol Physiol. 1978 Sep-Oct;5(5):535-9. doi: 10.1111/j.1440-1681.1978.tb00707.x. PMID: 215363.

  107. Estenne, Yernault, De Troyer (1980): Effects of parenteral aminophylline on lung mechanics in normal human. Am Rev Respir Dis. 1980 Jun;121(6):967-71. doi: 10.1164/arrd.1980.121.6.967. PMID: 7416595.

  108. Cushley, Tattersfield, Holgate (1984): Adenosine-induced bronchoconstriction in asthma. Antagonism by inhaled theophylline. Am Rev Respir Dis. 1984 Mar;129(3):380-4. doi: 10.1164/arrd.1984.129.3.380. PMID: 6703496.

  109. Takanashi, Hasegawa, Kanehira, Yamamoto, Fujimoto, Satoh, Okamura (1999): Interleukin-10 level in sputum is reduced in bronchial asthma, COPD and in smokers. Eur Respir J. 1999 Aug;14(2):309-14. doi: 10.1034/j.1399-3003.1999.14b12.x. PMID: 10515406.

  110. Tomita, Chikumi, Tokuyasu, Yajima, Hitsuda, Matsumoto, Sasaki (1999): Functional assay of NF-kappaB translocation into nuclei by laser scanning cytometry: inhibitory effect by dexamethasone or theophylline. Naunyn Schmiedebergs Arch Pharmacol. 1999 Apr;359(4):249-55. doi: 10.1007/pl00005349. PMID: 10344522.

  111. To, Ito, Kizawa, Failla, Ito, Kusama, Elliott, Hogg, Adcock, Barnes (2010): Targeting phosphoinositide-3-kinase-delta with theophylline reverses corticosteroid insensitivity in chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 2010 Oct 1;182(7):897-904. doi: 10.1164/rccm.200906-0937OC. PMID: 20224070; PMCID: PMC2970861.

  112. Eason, Markowe (1989): Aminophylline toxicity–how many hospital asthma deaths does it cause? Respir Med. 1989 May;83(3):219-26. doi: 10.1016/s0954-6111(89)80035-6. PMID: 2595040.

  113. Mohammadi, Kashani, Akhondzadeh, Izadian, Ohadinia (2004): Efficacy of theophylline compared to methylphenidate for the treatment of attention-deficit hyperactivity disorder in children and adolescents: a pilot double-blind randomized trial. J Clin Pharm Ther. 2004 Apr;29(2):139-44. doi: 10.1111/j.1365-2710.2004.00545.x. PMID: 15068402. n = 32

  114. Stein, Lerner (1993): Behavioral and cognitive effect of theophylline: a dose-response study. Ann Allergy. 1993 Feb;70(2):135-40. PMID: 8430921.

  115. Creer, Gustafson (1989): Psychological problems associated with drug therapy in childhood asthma. J Pediatr. 1989 Nov;115(5 Pt 2):850-5. doi: 10.1016/s0022-3476(89)80122-2. PMID: 2681640.

  116. Cools R, D’Esposito M (2011): Inverted-U-shaped dopamine actions on human working memory and cognitive control. Biol Psychiatry. 2011 Jun 15;69(12):e113-25. doi: 10.1016/j.biopsych.2011.03.028. PMID: 21531388; PMCID: PMC3111448. REVIEW

  117. Arnsten, Pliszka (2011): Catecholamine influences on prefrontal cortical function: relevance to treatment of attention deficit/hyperactivity disorder and related disorders. Pharmacol Biochem Behav. 2011 Aug;99(2):211-6. doi: 10.1016/j.pbb.2011.01.020. PMID: 21295057; PMCID: PMC3129015. REVIEW

  118. Kennaway (2015): Potential safety issues in the use of the hormone melatonin in paediatrics. J Paediatr Child Health. 2015 Jun;51(6):584-9. doi: 10.1111/jpc.12840. PMID: 25643981. REVIEW

  119. [Ferre, von Euler, Johansson, Fredholm, Fuxe (1Ferré, O’Connor, Snaprud, Ungerstedt, Fuxe (1994): Antagonistic interaction between adenosine A2A receptors and dopamine D2 receptors in the ventral striopallidal system. Implications for the treatment of schizophrenia. Neuroscience. 1994 Dec;63(3):765-73. doi: 10.1016/0306-4522(94)90521-5. PMID: 7898676.

  120. Pintsuk, Borroto-Escuela, Pomierny, Wydra, Zaniewska, Filip, Fuxe (2016): Cocaine self-administration differentially affects allosteric A2A-D2 receptor-receptor interactions in the striatum. Relevance for cocaine use disorder. Pharmacol Biochem Behav. 2016 May;144:85-91. doi: 10.1016/j.pbb.2016.03.004. PMID: 26987369.

  121. Nehlig, Daval, Debry (1992): Caffeine and the central nervous system: mechanisms of action, biochemical, metabolic and psychostimulant effects. Brain Res Brain Res Rev. 1992 May-Aug;17(2):139-70. doi: 10.1016/0165-0173(92)90012-b. PMID: 1356551.

  122. Ioannidis, Chamberlain, Müller (2014): Ostracising caffeine from the pharmacological arsenal for attention-deficit hyperactivity disorder – was this a correct decision? A literature review. Journal of Psychopharmacology, 28(9), 830–836. doi:10.1177/0269881114541014. REVIEW

  123. Ferré (2008): An update on the mechanisms of the psychostimulant effects of caffeine. J Neurochem. 2008 May;105(4):1067-79. doi: 10.1111/j.1471-4159.2007.05196.x. PMID: 18088379.

  124. Barraco, Helfman, Anderson (1996): Augmented release of serotonin by adenosine A2a receptor activation and desensitization by CGS 21680 in the rat nucleus tractus solitarius. Brain Res. 1996 Sep 16;733(2):155-61. doi: 10.1016/0006-8993(96)00279-x. PMID: 8891297.

  125. Castillo-Meléndez, Jarrott, Lawrence (1996): Markers of adenosine removal in normotensive and hypertensive rat nervous tissue. Hypertension. 1996 Dec;28(6):1026-33. doi: 10.1161/01.hyp.28.6.1026. PMID: 8952592.

  126. Pandolfo, Silveirinha, dos Santos-Rodrigues, Venance, Ledent, Takahashi, Cunha, Köfalvi (2011): Cannabinoids inhibit the synaptic uptake of adenosine and dopamine in the rat and mouse striatum. Eur J Pharmacol. 2011 Mar 25;655(1-3):38-45. doi: 10.1016/j.ejphar.2011.01.013. PMID: 21266173.

  127. Mioranzza, Botton, Costa, Espinosa, Kazlauckas, Ardais, Souza, Porciúncula (2010): Adenosine A1 receptors are modified by acute treatment with methylphenidate in adult mice. Brain Res. 2010 Oct 21;1357:62-9. doi: 10.1016/j.brainres.2010.08.004. PMID: 20699089.

  128. Mioranzza, Costa, Botton, Ardais, Matte, Espinosa, Souza, Porciúncula (2011): Blockade of adenosine A(1) receptors prevents methylphenidate-induced impairment of object recognition task in adult mice. Prog Neuropsychopharmacol Biol Psychiatry. 2011 Jan 15;35(1):169-76. doi: 10.1016/j.pnpbp.2010.10.022. PMID: 21044657.

  129. Schmitz, Pierozan, Rodrigues, Biasibetti, Grings, Zanotto, Coelho, Vargas, Leipnitz, Wyse (2017): Methylphenidate Decreases ATP Levels and Impairs Glutamate Uptake and Na+,K+-ATPase Activity in Juvenile Rat Hippocampus. Mol Neurobiol. 2017 Dec;54(10):7796-7807. doi: 10.1007/s12035-016-0289-1. PMID: 27844288.

  130. Sturgess, Ting-A-Kee, Podbielski, Sellings, Chen, van der Kooy (2010): Adenosine A1 and A2A receptors are not upstream of caffeine’s dopamine D2 receptor-dependent aversive effects and dopamine-independent rewarding effects. Eur J Neurosci. 2010 Jul;32(1):143-54. doi: 10.1111/j.1460-9568.2010.07247.x. Epub 2010 Jun 22. PMID: 20576036; PMCID: PMC2994015.

  131. Walker, Abraham, Tercyak (2010): Adolescent caffeineuse, ADHD, and cigarette smoking. Child Health Care 39: 73–90., n = 448

  132. Dosh, Helmbrecht, Anestis, Guenthner, Kelly, Martin (2010): A comparison of the associations of caffeine and cigarette use with depressive and ADHD symptoms in a sample of young adult smokers. J Addict Med. 2010 Mar;4(1):52-4. doi: 10.1097/ADM.0b013e3181b508ec. PMID: 21359163; PMCID: PMC3043357.

  133. Martin, Cook, Woodring, Burkhardt, Guenthner, Omar, Kelly (2008): Caffeine use: association with nicotine use, aggression, and other psychopathology in psychiatric and pediatric outpatient adolescents. ScientificWorldJournal. 2008 May 22;8:512-6. doi: 10.1100/tsw.2008.82. PMID: 18516472; PMCID: PMC3176831.

  134. Vázquez, Martin de la Torre, López Palomé, Redolar-Ripoll (2022): Effects of Caffeine Consumption on Attention Deficit Hyperactivity Disorder (ADHD) Treatment: A Systematic Review of Animal Studies. Nutrients. 2022 Feb 10;14(4):739. doi: 10.3390/nu14040739. PMID: 35215389; PMCID: PMC8875377. REVIEW

  135. Alasmari (2020): Caffeine induces neurobehavioral effects through modulating neurotransmitters. Saudi Pharm J. 2020 Apr;28(4):445-451. doi: 10.1016/j.jsps.2020.02.005. PMID: 32273803; PMCID: PMC7132598., REVIEW

  136. Diukova, Ware, Smith, Evans, Murphy, Rogers, Wise (2012): Separating neural and vascular effects of caffeine using simultaneous EEG-FMRI: differential effects of caffeine on cognitive and sensorimotor brain responses. Neuroimage. 2012 Aug 1;62(1):239-49. doi: 10.1016/j.neuroimage.2012.04.041. PMID: 22561357; PMCID: PMC3778750.

  137. Perault, Griesemann, Bouquet, Lavoisy, Vandel (1989): A study of the interaction of viloxazine with theophylline. Ther Drug Monit. 1989 Sep;11(5):520-2. PMID: 2815226.

  138. Laaban, Dupeyron, Lafay, Sofeir, Rochemaure, Fabiani (1986): Theophylline intoxication following viloxazine induced decrease in clearance. Eur J Clin Pharmacol. 1986;30(3):351-3. doi: 10.1007/BF00541543. PMID: 3732375.

  139. Fraporti, Contini, Tovo-Rodrigues, Recamonde-Mendoza, Rovaris, Rohde, Hutz, Salatino-Oliveira, Genro (2019): Synergistic effects between ADORA2A and DRD2 genes on anxiety disorders in children with ADHD. Prog Neuropsychopharmacol Biol Psychiatry. 2019 Jul 13;93:214-220. doi: 10.1016/j.pnpbp.2019.03.021. PMID: 30946941.

  140. Pires, Pamplona, Pandolfo, Fernandes, Prediger, Takahashi (2009): Adenosine receptor antagonists improve short-term object-recognition ability of spontaneously hypertensive rats: a rodent model of attention-deficit hyperactivity disorder. Behav Pharmacol. 2009 Mar;20(2):134-45. doi: 10.1097/FBP.0b013e32832a80bf. PMID: 19307960.

  141. Prediger, Fernandes, Takahashi (2005): Blockade of adenosine A2A receptors reverses short-term social memory impairments in spontaneously hypertensive rats. Behav Brain Res. 2005 Apr 30;159(2):197-205. doi: 10.1016/j.bbr.2004.10.017. PMID: 15817183.

  142. Prediger, Pamplona, Fernandes, Takahashi (2005): Caffeine improves spatial learning deficits in an animal model of attention deficit hyperactivity disorder (ADHD) – the spontaneously hypertensive rat (SHR). Int J Neuropsychopharmacol. 2005 Dec;8(4):583-94. doi: 10.1017/S1461145705005341. PMID: 15877934.

  143. Pires, Pamplona, Pandolfo, Prediger, Takahashi (2010): Chronic caffeine treatment during prepubertal period confers long-term cognitive benefits in adult spontaneously hypertensive rats (SHR), an animal model of attention deficit hyperactivity disorder (ADHD). Behav Brain Res. 2010 Dec 20;215(1):39-44. doi: 10.1016/j.bbr.2010.06.022. PMID: 20600342.

  144. Leffa, Ferreira, Machado, Souza, Rosa, de Carvalho, Kincheski, Takahashi, Porciúncula, Souza, Cunha, Pandolfo (2019): Caffeine and cannabinoid receptors modulate impulsive behavior in an animal model of attentional deficit and hyperactivity disorder. Eur J Neurosci. 2019 Jun;49(12):1673-1683. doi: 10.1111/ejn.14348. PMID: 30667546.

  145. Ramos, de Mattos Hungria, Camerini, Suiama, Calzavara (2020): Potential beneficial effects of caffeine administration in the neonatal period of an animal model of schizophrenia. Behav Brain Res. 2020 Aug 5;391:112674. doi: 10.1016/j.bbr.2020.112674. PMID: 32417274.

  146. Triche, Grosso, Belanger, Darefsky, Benowitz, Bracken (2008): Chocolate consumption in pregnancy and reduced likelihood of preeclampsia. Epidemiology. 2008 May;19(3):459-64. doi: 10.1097/EDE.0b013e31816a1d17. PMID: 18379424; PMCID: PMC2782959.

  147. Miguel, Bronauth, Deniz, Confortim, de Oliveira, Dalle Molle, Silveira, Pereira (2022): Neonatal hypoxia-ischemia induces dysregulated feeding patterns and ethanol consumption that are alleviated by methylphenidate administration in rats. Exp Neurol. 2022 Apr 7;353:114071. doi: 10.1016/j.expneurol.2022.114071. PMID: 35398338.

  148. Smith, Browne, Jayaraman, Bleickardt, Hodge, Lis, Yao, Rittle, Innocent, Mullins, Boykow, Reynolds, Hill, Parker, Hodgson (2014): Effects of the selective adenosine A2A receptor antagonist, SCH 412348, on the parkinsonian phenotype of MitoPark mice. Eur J Pharmacol. 2014 Apr 5;728:31-8. doi: 10.1016/j.ejphar.2014.01.052. PMID: 24486705.

  149. Basu, Barawkar, Thorat, Shejul, Patel, Naykodi, Jain, Salve, Prasad, Chaudhary, Ghosh, Bhat, Quraishi, Patil, Ansari, Menon, Unadkat, Thakare, Seervi, Meru, De, Bhamidipati, Rouduri, Palle, Chug, Mookhtiar (2017): Design, Synthesis of Novel, Potent, Selective, Orally Bioavailable Adenosine A2A Receptor Antagonists and Their Biological Evaluation. J Med Chem. 2017 Jan 26;60(2):681-694. doi: 10.1021/acs.jmedchem.6b01584. PMID: 28055204.

  150. Weerts, Griffiths (2003): The adenosine receptor antagonist CGS15943 reinstates cocaine-seeking behavior and maintains self-administration in baboons. Psychopharmacology (Berl). 2003 Jul;168(1-2):155-163. doi: 10.1007/s00213-003-1410-5. PMID: 12669180.

  151. O’Neill, Hobson, Levis, Bachtell (2014): Persistent reduction of cocaine seeking by pharmacological manipulation of adenosine A1 and A 2A receptors during extinction training in rats. Psychopharmacology (Berl). 2014 Aug;231(16):3179-88. doi: 10.1007/s00213-014-3489-2. PMID: 24562064; PMCID: PMC4111968.

  152. Moeller, Steinberg, Lane, Kjome, Ma, Ferre, Schmitz, Green, Bandak, Renshaw, Kramer, Narayana (2012): Increased Orbitofrontal Brain Activation after Administration of a Selective Adenosine A(2A) Antagonist in Cocaine Dependent Subjects. Front Psychiatry. 2012 May 28;3:44. doi: 10.3389/fpsyt.2012.00044. PMID: 22654774; PMCID: PMC3361057.

  153. Kubrusly, Bhide (2010): Cocaine exposure modulates dopamine and adenosine signaling in the fetal brain. Neuropharmacology. 2010 Feb;58(2):436-43. doi: 10.1016/j.neuropharm.2009.09.007. PMID: 19765599; PMCID: PMC2813374.

  154. Yao, McFarland, Fan, Jiang, Ueda, Diamond (2006): Adenosine A2a blockade prevents synergy between mu-opiate and cannabinoid CB1 receptors and eliminates heroin-seeking behavior in addicted rats. Proc Natl Acad Sci U S A. 2006 May 16;103(20):7877-82. doi: 10.1073/pnas.0602661103. PMID: 16684876; PMCID: PMC1458620.

  155. Justinová, Ferré, Redhi, Mascia, Stroik, Quarta, Yasar, Müller, Franco, Goldberg (2011): Reinforcing and neurochemical effects of cannabinoid CB1 receptor agonists, but not cocaine, are altered by an adenosine A2A receptor antagonist. Addict Biol. 2011 Jul;16(3):405-15. doi: 10.1111/j.1369-1600.2010.00258.x. PMID: 21054689; PMCID: PMC3115444.