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
Amphetamine medication for ADHD

Sitemap

Amphetamine medication for ADHD

In the USA, amphetamine drugs are available as:1

  • Mixture of D- and L-amphetamine isomers (racemic mixture)
  • Mixed sulfates and saccharinates of D-L-amphetamine isomers (Adderall)
  • Pure D-amphetamine sulphate
    • Dexamfetamine hemisulfate (Attentin)
  • D-amphetamine as lisdexamfetamine in lysine-bound form (Elvanse, Tyvense)
  • Racemic methamphetamine sulfate (Desoxyn, USA)

In Germany, amphetamine medication had to be prepared from raw substances by pharmacists for a long time.2 Since 2011, a D-amphetamine has been available in Germany as a finished drug and approved for the treatment of ADHD (Attentin), and in 2013 a D-amphetamine prodrug was approved for the treatment of children. Lisdexamfetamine contains D-amp in lysine-bound form (Elvanse). Since May 2019, Elvanse Adult has been approved for the treatment of ADHD in adults (30, 50, 70 mg). In 2023, 20, 40, 60 mg were also approved for adults, although it is not yet clear when these will also be available.

In Austria, Elvanse can be prescribed if other medications are ineffective or show side effects. The doctor must justify this to the insurance company.

Amphetamine medication works slightly better in adults than methylphenidate3 and has slightly fewer side effects.
According to the current European consensus, amphetamine medication is the first choice ADHD medication for adults (before methylphenidate) and the second choice for children and adolescents (after methylphenidate).45 While the current text of the 2017 S3 guideline still states that lisdexamfetamine can only be used in accordance with approval after prior treatment with MPH6, the 2019 S3 guideline is quoted as saying that the first option recommended for adults with ADHD is treatment with psychostimulants, which includes the active substances methylphenidate and lisdexamfetamine, which are approved for adults.78

Due to the responder/non-responder profile, which differs from MPH, amphetamine medications are particularly suitable for ADHD sufferers who do not respond to MPH, clearly before the use of non-stimulants (e.g. noradrenergic medications or tricyclic antidepressants).9 A summary of several studies reports a 69% response rate to amphetamine medication and a 59% response rate to methylphenidate. 87 % of those affected responded to one of the two types of active substance.10

Amphetamine medications are also suitable - unlike MPH - for the co-treatment of comorbid dysphoria or depression.1112

According to a Cochrane study, all amphetamine medications work equally well in adults, regardless of the specific form of medication.13 This distinguishes amphetamine medication from methylphenidate, where even a switch to another methylphenidate preparation shows considerable individual differences.

In studies on the effects of amphetamine, it must always be borne in mind that these

  • AMP is usually used in significantly higher doses than for ADHD medication
  • use unretarded / not prolonged-acting AMP via prodrug
  • frequently inject AMP, which again results in much faster metabolization
  • these 3 factors multiply in their effect

There is no doubt that AMP in drug form has a different effect than AMP in drug form.

1. Active ingredients of amphetamine drugs

AMP has a chiral center with two enantiomers:14

  • Levo-AMP (l-AMP)
    • Noradrenaline release as strong as or stronger than d-AMP
  • Dextro-AMP (d-AMP)
    • higher dopamine release than l-AMP

As a result, the amphetamine mixed salt preparations available in the USA, which consist of equal parts racemic d,l-AMP sulfate, d,l-AMP aspartate monohydrate and two enantiomerically pure d-AMP salts (d-AMP sulfate and d-AMP saccharate), resulting in a ratio of 3:1 between d-AMP and l-AMP isomers and salts, a relatively greater release of noradrenaline than pure d-AMP, but still a greater release of dopamine than noradrenaline in absolute terms.

The following are relevant for ADHD treatment:

1.1. Dextroamphetamine (D-amphetamine)

Dextroamphetamine is also known as dexamphetamine or dextroamphetamine sulphate.
Dextroamphetamine is the dextrorotatory (D-)enantiomer of amphetamine, as opposed to the levorotatory levoamphetamine (see below).

D-amphetamine medications have a 3 to 4 times stronger effect on the central nervous system than racemic amphetamine medications, with a simultaneously lower sympathomimetic effect in the periphery, which is why D-amphetamine medications are preferred in the treatment of ADHD.15
D-amphetamine is only more potent than L-amphetamine in relation to the dopamine transporters, while the effect on noradrenaline transporters is roughly the same.16

This opens up the possibility of emphasizing dopaminergic medication (dexamphetamine) or balancing dopaminergic and noradrenergic (levoamphetamine).

D-amphetamine is more activating than MPH and is therefore preferably recommended for ADHD-I.17
It is also often more effective than MPH for parallel dysthymia / dysphoria / depression due to the noticeable serotonergic effect18.

1.1.1. D-amphetamine without lysine binding

Trade name: Attentin (Germany since the end of 2011), Dexamine (Switzerland: as magistral formulation), Dexedrine

Duration of action approx. 6 hours, so that it is usually necessary to take it twice a day.
Increased potential for abuse as no lysine binding.

Medice (2017): Attentin® - Guide for prescribing physicians

1.1.2. D-amphetamine from lisdexamfetamine (with lysine binding)

Lisdexamfetamine (LDX) is a prodrug of D-amphetamine that is bound to L-lysine to form a substance that is ineffective in itself. Lisdexamfetamine is therefore an active substance that is first converted in the body into the actual active substance, in this case D-amphetamine. This means that there is a very low risk of abuse.19

Lisdexamfetamine (LDX) bound to lysine is rapidly absorbed from the small intestine into the bloodstream. This occurs by active transport, presumably by the peptide transporter 1 [PEPT1]. Enzymatic hydrolysis of the peptide bond to release d-amphetamine into the blood occurs in the lysate and in the cytosolic extract of human erythrocytes, but not in the membrane fraction. This conversion is strongly inhibited by a protease inhibitor cocktail, bestatin and ethylenediaminetetraacetic acid, suggesting an aminopeptidase as the cause of hydrolytic cleavage of the LDX peptide bond. Aminopeptidase B does not appear to be the cause20

Due to the necessary and slow conversion step from LDX to d-AMP, the effect occurs approx. 1 hour later than when taking d-AMP sulphate. Unlike LDX, the pharmacologically active d-AMP crosses the blood-brain barrier and enters the CNS, where it exerts its effect.14

As the effect is very even throughout the day, the unpleasant rebound effects known from MPH (short-term increased restlessness at the end of the day) are eliminated.
The effect corresponds to D-amphetamine. A conversion table from dexamphetamine to Elvanse can be found at ADHSpedia.21

Trade names:

  • Elvanse (EU, since the end of 2013, for children, 20, 30, 40, 50, 60, 70 mg)22
  • Elvanse Adult (EU, since 01.05.19, for adults, 30, 50, 70 mg)22. Since 2023, 20, 40 and 60 mg have also been approved in Germany.
  • Vyvanse (USA) is available in doses from 10 mg to 70 mg23
  • Tyvense

Elvanse and Elvanse Adult are the same medicine and can therefore be used interchangeably (with the consent of the health insurance company).

Elvanse has only been classified as a BtM in Germany since 2013.
Austria appears to be the only country in which Elvanse is not classified as a narcotic drug, even as of 2023.24

1.2. Levoamphetamine (L-amphetamine)

Levoamphetamine (L-amphetamine) is the purely levorotatory isomer of amphetamine.

L-amphetamine is less potent than D-amphetamine in terms of dopamine transporters, while the effect on noradrenaline transporters is roughly the same.16 This makes it slightly more noradrenergic than D-amphetamine, but still predominantly dopaminergic.25
L-amphetamine increases blood pressure and pulse rate more than D-amphetamine26

We are not aware of any ready-to-use L-amphetamine medication approved in Europe. It would have to be produced on individual prescription in pharmacies.

1.3. Mixed amphetamine salts / amphetamine derivatives

  • Aderall (USA): 75 % dextroamphetamine and 25 % levoamphetamine
  • Evekeo (USA): 50 % dextroamphetamine and 50 % levoamphetamine

Mixed amphetamine salts are a combination of different stimulants:27

D-amphetamine saccharate
D-amphetamine sulfate
D,L-amphetamine sulfate
D,L-amphetamine aspartate monohydrate

While D,L-amphetamine sulphate mixtures are the most commonly used ADHD medication in the USA, D,L-amphetemine mixtures are only available in a few pharmacies in Germany, which produce them themselves. The production is associated with a waiting time of several weeks. The cost of 180 capsules of 5 mg amphetamine sulphate each was quoted as €200.

1.4. Methamphetamine

  • Desoxyn, USA

(1.5. Fenetyllin)

  • Captagon (in Germany until 2003; in Belgium until 2010); no longer available today

2. Amphetamine drugs work differently and in different parts of the brain than methylphenidate

Amphetamine drugs have a more complex mechanism of action than methylphenidate.
The description of the effects of amphetamine drugs is contradictory.

It is sometimes argued that amphetamine drugs merely inhibit dopamine reuptake and release dopamine and noradrenaline. More well-founded accounts from the USA (where amphetamine drugs are prescribed more frequently than in Europe and where there is therefore a more intensive debate about them) cite a reuptake inhibition of dopamine and noradrenaline transporters as the effect and no release of dopamine, noradrenaline or serotonin.

In the USA, 52.9% of adolescents with ADHD receive MPH and 39.3% amphetamine medication as their first prescribed medication. Over the course of treatment, MPH is the primary prescribed medication for around 40% and AMP is the primary prescribed medication for 33%.28

In principle, amphetamine drugs are said to have an intraneuronal effect, while methylphenidate and atomoxetine have an extraneuronal effect.29 As amphetamine drugs also address at least the dopamine transporter and the D2 autoreceptor, this is unlikely to be tenable.
AMP acts primarily in the striatum and further in the cortex and ventral tegmentum.30

The first computer models now exist that can seriously simulate the effect of ADHD drugs. A computer model for the simulation of type 1 diabetes has already been approved by the FDA as a replacement for preclinical animal studies31
A model comparing MPH and AMP in children and adults with ADHD takes into account the effect on 99 proteins involved in ADHD32

2.1. Dopamine with amphetamine medication

The dopamine increase caused by D-amphetamine in the PFC is much more pronounced and also much more dose-dependent than with MPH, and is therefore easier to control.29
AMP causes:

  • extracellular dopamine levels increased 6-fold33
  • tonic dopamine firing enhanced by AMP depleting vesicular stores and promoting non-exocytotic release through reverse transport34
  • phasic dopamine firing: contradictory data
    • amplified by upregulating vesicular dopamine release34
    • Stimulants reduce the phasic release of dopamine33
    • AMP reduces vesicular release3536 (this can affect both tonic and phasic release)

2.1.1. Effect on DAT

2.1.1.1. Dopamine (re)uptake inhibition via DAT and NET

Stimulants (MPH such as AMP inhibit dopamine reuptake37 and thus lead (in low doses) to a 6-fold increase in extracellular dopamine levels.33
The increased extracellular dopamine level acts on presynaptic dopamine D2 autoreceptors at the nerve ending. The D2 autoreceptor activation causes a 2 to 3-fold increase in impulse-associated (phasic) dopamine release. This increase is therefore relatively smaller than the increase in extracellular dopamine. The (relatively smaller) increase in phasic dopamine acts on the postsynaptic D2 dopamine receptors and causes reduced locomotor activity. Higher doses of stimulants increase extracellular dopamine more strongly and result in marked behavioral stimulation that cannot be overcome by phasic activation of inhibitory postsynaptic D2 receptors. High D-Amp doses cause supersaturation of extracellular postsynaptic D1 and D2 receptors, so that they exceed the inhibitory presynaptic effect of low D-AMP doses.33

  • Amphetamine drugs block the dopamine and noradrenaline transporters in a different way to methylphenidate. While the reuptake inhibition of MPH is similar to that of antidepressants, amphetamine drugs act as a competitive inhibitor and pseudosubstrate on dopamine and noradrenaline transporters and bind to the same site where the monoamines bind to the transporter, thereby also inhibiting NE and DA reuptake.38
  • D-amphetamine works
    • Primarily as a dopamine reuptake inhibitor.39
    • Equally as a dopamine and noradrenaline reuptake inhibitor38
  • “Amphetamines can also stabilize dopamine and noradrenaline transporters in channel configurations, reverse flow through intracellular vesicular monoamine transporters, and cause internalization of dopamine transporters”40
    D-AMP drug doses cause a D-AMP plasma concentration of around 150 nM, which is sufficient to occupy a significant proportion of the dopamine transporters. This effect coincides with that of MPH.33
  • D-amphetamine has approximately three times the affinity for noradrenaline transporters (NET) for reuptake inhibition and two and a half times the affinity for dopamine transporters (DAT) compared to racemic methylphenidate.29
2.1.1.1.1. DAT inhibition via PKC
  • AMP possibly inhibits DAT via PKC41
    • Several protein kinases regulate DAT function4243
    • AMP increases the activity of striatal particulate PKC via a calcium-dependent signaling pathway44
    • PKC activation leads to phosphorylation in the N-terminal of the rat striatal DAT45
    • PKC activation stimulates DAT-mediated dopamine release41
    • PKC inhibitors and the downregulation of PKC41
      • Inhibit efflux
      • Leave dopamine uptake unchanged
2.1.1.2. Increased release of dopamine (DAT efflux)

The increased DAT efflux increases extracellular dopamine.

Amphetamine drugs release dopamine into the extracellular space 29 3937
Amphetamines therefore not only act as dopamine reuptake inhibitors, but also reverse the DAT function so that the DAT not only does not reabsorb dopamine, but also releases it from the cell (efflux).46
This is newly synthesized dopamine. There is no doubt that this is not a depletion of dopamine reserves, as amphetamine drugs would otherwise have no lasting effect.
It is questionable whether this is dopamine that was previously stored in vesicles. There is no doubt that amphetamine drugs (characteristic of drugs: high dose, rapid application, rapid onset of action) release dopamine. It is questionable whether this is also the case with amphetamine drugs (characteristic: drug-like = low dose, slow release, long-lasting effect), and if so, to what extent this is the case.

2.1.1.2.1. Via VMAT2 at high doses
  • (Only) at a very high dosage as a drug, amphetamines also act on the vesicular monoamine transporter 2 (VMAT2) for dopamine and noradrenaline and then trigger an accumulating release of dopamine from the synaptic vesicles. The high amount of dopamine is then swept out into the synaptic cleft by a reversal of action of the dopamine transporters. This mechanism does not take effect with the usual dosage as an ADHD medication.16 In other words: Amphetamines can enter presynaptic monoamine vesicles and cause an efflux of neurotransmitters towards the synapse.47
  • An administration of 1 mg/kg AMP (injected) already caused a dopamine DAT efflux that was significantly higher at 10 mg/kg.48
2.1.1.2.2. By increasing intracellular Ca2+

AMP increases intracellular Ca2+, which supports phosphorylation of DAT at the N-terminus of the transporter. Phosphorylation (by CaMKII and possibly also by PKCβ) increases probability of DAT efflux from cytoplasmic DA.49

2.1.1.2.3. Increased DAT efflux via TAAR1
  • AMP acts on DAT via TAAR1
    Amphetamine enables the trace amine-associated receptor 1 (TAAR1) to phosphorylate the DAT transporter. This interrupts the reuptake of dopamine and the DAT is stimulated to release dopamine (efflux).47
  • AMP also leads to increased intracellular accumulation of DAT50

2.1.2. Vesicular release

  • AMP reduces vesicular release because as a lipophilic weak base and as a substrate for VMAT, AMP promotes the redistribution of dopamine from the synaptic vesicles into the cytosol by collapsing the vesicular pH gradient.35 As a result, AMP reduces the number of dopamine molecules released per vesicle.36
  • Amphetamine initially reduces VMAT2, while prolonged administration increases it.51 MPH increases VMAT2 per se.5253
  • AMP can inhibit vesicular release by indirectly activating D2 autoreceptors. The activation of D2 autoreceptors regulates potassium channels, which in turn regulate the probability of exocytic dopamine release.36
  • A computer model determined54
    • A maximum release of dopamine at 0.5-1.0 mg/kg AMP (lower at lower doses than at higher doses)
    • Most of the dopamine released resulted from AMP-stimulated dopamine neosynthesis
      • The dopamine produced was immediately converted into DOPAC, which is excreted extracellularly
      • The dopamine was not stored in vesicles
  • According to Stahl, AMP does not release dopamine, at least at low doses16
  • AMP caused a gradual 10-fold increase in extracellular dopamine in the striatum over approximately 30 minutes in wild-type mice in vitro and in vivo, while simultaneously reducing the dopamine pool available for electrically stimulated release. If vesicular dopamine was previously released into the cytosol by reserpine, extracellular dopamine did not increase; however, AMP caused a rapid increase in dopamine within 5 minutes. In DAT-KO mice, extracellular dopamine did not increase, but at the same time electrically stimulable dopamine release was also reduced. DAT are therefore required for the dopamine-releasing effect of AMP, but not for the vesicle-emptying effect. Dopamine emptying of the vesicles is the rate-limiting step for the AMP effect on dopamine.55
  • AMP (10 microm) promoted the release of dopamine from vesicles by reducing the affinity of vesicles for dopamine uptake (from K(m) 0.8 to K(m) 32 microm). However, the amount of dopamine released per pulse was reduced by 82 % (according to another source by 25 to 50 %). The D2 antagonist sulpiride reduced the inhibition of release, i.e. promoted the release. This was reduced in D2-KO mice.
    In inhibited D2 autoreceptors, AMP increased the extracellular release of dopamine.56
  • Emptying of the vesicular DA stores through a weak alkaline effect on the intravesicular pH gradient. The intravesicular pH gradient is required for the concentration of DA.
  • Different effect on release-ready vesicles and reserve pool vesicles:34
    • stimulus-dependent effect in the dorsal striatum
      • stimulates vesicular dopamine release
        • by a firing of short duration
        • via vesicle pool ready for release
      • Release reduced
        • through a firing of long duration
        • which accesses the reserve pool
      • these opposing effects of vesicular dopamine release were associated with simultaneous increases in tonic and phasic dopamine responses
    • in the ventral striatum
      • only increased vesicular release and increased phasic signals

2.1.3. D2 autoreceptor activation

Basically, D-amphetamine activates D2 dopamine autoreceptors in the striatum.57
However, drug doses of D-AMP do not cause a significant reduction in dopamine release via activation of the D2 autoreceptors.5859

Since drugs such as levodopa or piribedil have no positive effect on ADHD, although they reduce the firing rate of the dopaminergic neurons of the substantia nigra pars compacta, it is doubtful whether the reduction of hyperactivity in ADHD by stimulants is based on presynaptic inhibition. It is more likely that the reduction in hyperactivity in ADHD caused by stimulants is due to an increase in dopamine release.58

2.1.4. Increase in tyrosine hydroxylase

Amphetamine drugs appear to have an activating effect on tyrosine hydroxylase in the dorsal striatum and nucleus accumbens, leading to increased L-dopa levels, but this does not appear to occur via a change in the phosphorylation of tyrosine hydroxylase.60

2.1.5. Increased DA firing / activation in dopaminergic brain regions

2.1.5.1. Increased DA firing in caudate nucleus / putamen (striatum)

High (well above drug dose) D-amphetamine administration (2.5 to 10 mg/kg in the rat into the abdominal cavity), leads to increased dopaminergic firing in the caudate nucleus and putamen and causes focused-repetitive (stereotypic) behavior.6162 The D2 antagonist haloperidol (2 mg/kg) terminates the excessive firing in the caudate nucleus and putamen and the reduced firing in the nucleus accumbens61

2.1.5.2. Increased DA firing in VTA and substantia nigra

D2 antagonists prevent increased firing in the substantia nigra and VTA (in vivo).63

2.1.5.3. Increased activation in the right orbitofrontal cortex, left middle frontal lobe, superior frontal lobe and precentral gyri

The improvement in ADHD symptoms with LDX was associated with significantly increased activation in a number of brain regions previously implicated in reinforcement processing under choice and feedback conditions (e.g., left caudate and putamen, right orbitofrontal cortex, left middle frontal lobe, superior frontal lobe, and precentral gyri).64

2.1.6. Reduced DA firing in the nucleus accumbens

In the nucleus accumbens, 7.5 mg/kg D-Amp led to a reduction in dopaminergic firing.61 The D2 antagonist haloperidol (2 mg/kg) terminated the excessive firing in the caudate nucleus and putamen and the reduced firing in the nucleus accumbens61

2.1.7. DA influence indirectly via effects on dopamine cells emanating from other brain regions

Amphetamine appears to influence the activity of dopamine cells indirectly via its effects on dopamine cells originating in other brain regions.65

Amphetamine can excite dopamine neurons by modulating glutamate neurotransmission. Amphetamine strongly inhibits inhibitory postsynaptic potentials in dopamine neurons mediated by the metabotropic glutamate receptor (mGluR), but has no effect on excitatory postsynaptic currents mediated by the ionotropic glutamate receptor. Amphetamine desensitizes mGluR-mediated hyperpolarization by:66

  • DA release
  • Activation of postsynaptic alpha1-adrenergic receptors
  • Suppression of InsP3-induced calcium release from internal stores
    By selectively suppressing the inhibitory component of glutamate-mediated transmission, amphetamine can promote burst firing of dopamine neurons and thus increase the phasic release of dopamine.

2.1.8. Downregulation of dopamine receptors?

Reports of immediate downregulation of dopamine receptors by administration of amphetamine are based on studies in which rats were given doses of amphetamine. This concerns the dosage level (5, 10, 15 mg/kg for 4 or 20 days twice daily) as well as the form (injection).67 Interestingly, a single dose of D-AMP even increased the number of receptors.6759
So far, we are not aware of any reports of downregulation when administered in the dose and form of medication.

Similarly, only studies with drug doses of amphetamines appear to change the dopamine receptor affinity or receptor status from high-affinity to low-affinity. Drug doses could alter the balance between receptor status towards low-affinity.59
More on receptor status at High-affinity and low-affinity receptor status In the article Dopamine effect on receptors

However, it is conceivable that amphetamine in drug doses does not cause a desensitization of the postsynaptic or extrasynaptic (the majority of dopamine receptors are located outside of synapses) D1 and D2 receptors directly, but via the detour of increasing the extracellular dopamine level. However, this hypothesis has not yet been experimentally proven.59 It is possible that this pathway leads to reduced psychomotor activity through amphetamine medication. In our view, however, this is contradicted by the fact that this effect already occurs with the first dose. On the other hand, this pathway could explain why many sufferers benefit from a slow and gradual titration of stimulants,

2.2. Noradrenaline with amphetamine medication

2.2.1. Noradrenaline reuptake inhibition via NET

  • Amphetamine drugs block the dopamine and noradrenaline transporters in a different way to methylphenidate. While the reuptake inhibition of MPH is similar to that of antidepressants, amphetamine drugs act as a competitive inhibitor and pseudosubstrate on dopamine and noradrenaline transporters and bind to the same site where the monoamines bind to the transporter, thereby also inhibiting NE and DA reuptake.3868
  • “Amphetamines can also stabilize dopamine and noradrenaline transporters in channel configurations, reverse flow through intracellular vesicular monoamine transporters, and cause internalization of dopamine transporters”40
  • D-amphetamine has about a third of the reuptake inhibition on the noradrenaline transporter (NET) and dopamine transporter (DAT) as racemic methylphenidate.29
  • Amphetamine (as well as ephedrine) also inhibit the intracellular noradrenaline transporter, which takes up noradrenaline from the nerve cell into the vesicles (the neurotransmitter stores)68

2.2.2. Noradrenaline release

  • Whether amphetamine has a noradrenaline-releasing effect when administered as a drug is the subject of controversial debate, as is the case with dopamine. There are voices against16 as well as in favor.3739
  • D-amphetamine secondarily increases the release of noradrenaline.57 This is always the case with dopaminergic drugs due to the conversion of dopamine (approx. 5 - 10 %) into noradrenaline.
  • There is no doubt that amphetamine drugs do not lead to a chronic depletion of noradrenaline reserves in the sense of a deficiency state. It is empirically proven that amphetamine medication does not cause any long-term habituation effects in ADHD

2.5 mg/kg AMP in mice:69

  • stereotypical behavior (a sign of strongly increased extracellular dopamine); as strong as 20 mg/kg MPH
  • extracellular dopamine increased
  • extracellular noradrenaline increased
  • extracellular serotonin increased

2.2.3. Reduction of noradrenaline metabolites only in responders

  • In several independent studies, D-amphetamine drugs were found to decrease the urinary metabolite of norepinephrine, MHPG. The decrease of MPHG in urine is thought to be an important indicator of stimulant onset, indicating a lowering of noradrenaline levels by dextroamphetamine drugs.70](https://psycnet.apa.org/psycinfo/1982-21744-001)
  • Furthermore, the reduction in noradrenaline metabolites only occurs in ADHD sufferers who respond positively to dexamphetamine (responders).71
  • Even with the administration of methylphenidate, only the responders showed a significant decrease in MPHG in the urine, while MPHG in the urine of the non-responders did not decrease.72
    The authors conclude from this that the level of noradrenaline in ADHD is reduced.
  • Furthermore, several studies with ADHD sufferers found that behavioral improvements were proportional to the reduced noradrenal metabolite levels (using D-amphetamine medication).73

In contrast to the reduction of metabolites in urine by D-amphetamine, the noradrenaline increase in PFC mediated by D-amphetamine is approximately as pronounced as that of MPH, but is significantly more dose-dependent and therefore more controllable.29

2.2.4. DA firing and DA bursting increased via noradrenaline α1 receptors

D-Amp (1 to 2 mg/kg) acts via alpha1-adrenoceptors74 (but not via alpha2- or beta-adrenoceptors) to increase dopaminergic firing and bursting in substantia nigra and VTA (in vivo). This adrenergic pathway is normally masked by the reduction in dopaminergic firing mediated by D2 autoreceptors and is visualized by D2 antagonists or by simultaneous administration of D1/D5 and D2/D3/D4 blockers. The selective norepinephrine uptake blocker nisoxetine did not increase the DA firing rate, but did increase DA bursts.6375

D-amphetamine appears to activate the noradrenaline α1-receptor in the PFC, as the α1-receptor antagonist prazosin completely neutralized the effect of D-amphetamine in the PFC. In contrast, D-amphetamine does not appear to target either the α2 receptor or the β receptor, as the effect of D-amphetamine persisted when the α2 or β receptors were blocked.76
D-amphetamine promotes the up-state of cortical neurons by activating77

  • Central α1A-adrenoceptors
  • D1 receptors
  • D2 receptors
  • But not by D1 or D2 receptors alone

In contrast, the dopamine/noradrenaline precursor L-DOPA did not promote the up state.

Arousal is associated with an increased up state, while slow-wave sleep, general anaesthesia and calm wakefulness are characterized by an oscillating change between up and down states. During arousal, the down states end and the up/down oscillation changes to a sustained up state.
The up/down oscillations appear to be relevant for memory consolidation, while the transition to a sustained up state is required for arousal and attention.77

2.3. Monoamine degradation inhibition via MAO

Amphetamine drugs act as MAO inhibitors,7830 differently than low-dose MPH. Whether high-dose MPH acts as an MAO inhibitor is unknown.29

MAO is an enzyme that breaks down dopamine and noradrenaline in the cell. MAO inhibitors thus increase the amount of dopamine and noradrenaline available in the cell. As dopamine and noradrenaline continue to be synthesized in the nerve cell, the noradrenaline and dopamine levels in the cell continue to rise. This leads to a reversal of the effect of the transporters (which actually return DA and NE from the synaptic cleft into the cell), so that they release NE and DA into the synaptic cleft, even without this being triggered by a nerve signal to be transmitted.78 This effect triggers peripheral hypertension and an increase in heart rate. As this mechanism of action occurs indirectly at the presynapse, ephedrine and amphetamine drugs are also called “indirect sympathomimetics”, while drugs that act directly on the postsynaptic receptors are called sympathomimetics.78

2.4. Serotonin release

Amphetamine drugs are said to release a small amount of serotonin.7915 Here, too, it is unclear whether this is really also the case when dosed at drug level, or whether this effect is only limited when dosed as drugs. In any case, Stahl does not report a serotonergic effect of amphetamine drugs.38

2.5 mg/kg AMP in mice:69

  • stereotypical behavior (a sign of strongly increased extracellular dopamine); as strong as 20 mg/kg MPH
  • extracellular dopamine increased
  • extracellular noradrenaline increased
  • extracellular serotonin increased

Serotonin release through amphetamine drugs

Amphetamine drugs (MDMA, MBDB) also increase the release of serotonin. It is assumed that amphetamine-induced serotonin release not only influences psychomotor activation, but also subjective well-being (and euphoria when taken as a drug).80 MDBD causes almost no dopamine release.

Hyperactivity induced by 5 mg or 10 mg / kg MDMA (= 10 to 20 times higher dosage than as medication) could be prevented by prior administration of 2.5 and 10 mg / kg of the selective serotonin reuptake inhibitor fluoxetine. Fluoxetine had the same effect on the interactive effect of MDMA and P-chloroamphetamine.81 This suggests that MDMA causes hyperactivity by increasing serotonin via the serotonin transporter, which was blocked by fluoxetine as a serotonin reuptake inhibitor.

  • There is evidence that an increased release of serotonin indirectly increases dopamine levels.81
  • Other sources point to a serotonin-increasing effect of amphetamine salts due to inhibition of monoamine oxidase.18
  • Amphetamine increases c-Fos expression in the mPFC, striatum and nucleus accumbens. A serotonin 1A receptor agonist reduced the c-Fos increase in the mPFC and striatum, but not in the nucleus accumbens.82
  • MPH itself has an agonistic effect on the 5-HT1A receptor.30

2.5. Effect on the HPA axis

2.5.1. ACTH increased

Lisdexamfetamine and d-amphetamine significantly increased plasma levels in healthy subjects of, among others:83

  • ACTH

2.5.2. Corticosteroids increased

  • D-amphetamine drugs such as lisdexamphetamine drugs (Elvanse) increase cortisol levels but not testosterone levels.83
  • The following were increased
    • Glucocorticoids (as with methylphenidate; the increase was even greater with the drugs MDMA or LSD)
      • Cortisol
      • Cortisone
      • Corticosterone
      • 11-Dehydrocorticosterone,
      • 11-Deoxycortisol
  • The following remained unchanged
    • Mineralocorticoids
      • Aldosterone
      • 11-Deoxycorticosterone

The increase in the cortisol level causes a stronger addressing of the glucocorticoid receptor (GR) by cortisol. Cortisol causes the HPA axis to be switched off again via GR at the end of the stress response.
In ADHD-HI and ADHD-C (both with hyperactivity), due to the flattened endocrine stress response of the adrenal gland, it can be assumed that the GR are not sufficiently addressed to switch off the HPA axis again after a stress reaction. In addition, in ADHD-HI (unlike ADHD-I) there is often deficient GR function, which makes HPA axis deactivation even more difficult.
Find out more at Medication for ADHD at Dexamethasone for ADHD. If the release of cortisol is increased by AMP, this could improve the resilencing of the HPA axis in ADHD-HI. However, as AMP also works in ADHD-I, the primary mechanism of action is likely to be different.

2.5.3. Increased steroid hormones

Lisdexamfetamine and d-amphetamine significantly increased plasma levels in healthy subjects of, among others:83

  • Androgens
    • Dehydroepiandrosterone
    • Dehydroepiandrosterone sulfate
    • Androstenedione (Δ4-androstene-3,17-dione)
    • Progesterone (only for men)

The androgen remained unchanged

  • Testosterone

Since aggression correlates with an increased testosterone to cortisol ratio, amphetamine drugs have an anti-aggressive effect due to the relative increase in cortisol levels.
More on this at Neurophysiological correlates of aggression

A study on juvenile rhesus monkeys found that both active substances increased testosterone levels as a result of 12 months of AMP or MPH administration in drug doses, MPH even more significantly than AMP.84 Another study in rhesus monkeys found reduced testosterone levels with MPH administration.85

A reduction in testosterone levels was observed in rodents following amphetamine administration.8687

2.6. Inhibition of OCT2

Basic information on uptake-2 transporters can be found at Dopamine degradation by organic cation transporters (OCT) In the article Dopamine reuptake, dopamine degradation

Organic cation transporter 2 (OCT2) is involved in the degradation of dopamine. OCTs take up dopamine and noradrenaline as well as serotonin and, to a somewhat greater extent, histamine in glial cells, where they are degraded by COMT. OCT2 and OCT3 are also located on (also dopaminergic) neurons.
While methylphenidate only binds to OCT1 (IC50: 0.36) and neither to OCT2, OCT3 nor PMAT88, d-amphetamine acts as a highly effective hOCT2 reuptake inhibitor (Ki: 10.5 mM) and moderately effective hOCT1 reuptake inhibitor (Ki: 202 mM), while it only interacted with hOCT3 from 100 μM (Ki: 460 mM) (hOCT: human OCT) 8889
d-Amphetamine binds approximately equally strongly to hOCT2 and hOCT3 and to these by an order of magnitude (factor 10) weaker than to DAT89

Binding of amphetamine to OCT may contribute to cellular and behavioral effects of amphetamine.89

OCT2 reuptake inhibitors have an antidepressant effect.90 In addition, even much lower doses of venlaflaxine or reboxetine have an antidepressant effect in OCT2-KO mice than in wild-type mice91
We believe it is worth considering whether this approach could also support the effect of dopamine reuptake inhibitors in ADHD.
These correlations could also explain why AMP, which also acts as an OCT2 inhibitor, has a better antidepressant effect than MPH, which only binds to OCT1.

2.7. Other effects on brain functions

  • D-amphetamine increases metabolism in the right caudate nucleus and decreases it in the right Rolandic region and in the right anterior inferior frontal regions.92
  • D-amphetamine (as well as L-dopa, which has no effect on ADHD, although it has a dopaminergic effect) is also suitable for restoring brain function after strokes, but only if suitable training measures are taken at the same time.93 D-amphetamine increases dopamine, which has a neurotrophic effect (promotes neuroplasticity). Dopaminergic drugs such as (D-)amphetamine drugs or MPH can therefore also support appropriate training measures (e.g. neurofeedback, cognitive behavioral therapy) in ADHD by reducing the restrictions on learning ability.
  • Methylphenidate and amphetamine drugs increase the power of alpha (in rats), while atomoxetine and guanfacine do not.94
  • Lisdexamfetamine (Elvanse) has the following effects95
    • Increased acetylcholine levels in the cortex
    • Increased histamine levels in the cortex and hippocampus (which escitalopram given in parallel only prevents in the hippocampus)

Amphetamine medication is therefore not just a substitute for methylphenidate, but has its own area of application.

2.8. Overview of AMP and neurotransmitters

2.8.1. Binding affinity of AMP, MPH, ATX to DAT / NET / SERT

The active ingredients methylphenidate (MPH), d-amphetamine (d-AMP), l-amphetamine (l-AMP) and atomoxetine (ATX) bind with different affinities to dopamine transporters (DAT), noradrenaline transporters (NET) and serotonin transporters (SERT). The binding causes an inhibition of the activity of the respective transporters.96
The values given in the following table by Easton et al. refer to values in the synaptosome as well as to the DAT in the striatum and the NET in the PFC.

Binding affinity: stronger with smaller number (KD = Ki) DAT NET SERT
MPH 34 - 20096 , 34125 23825, 33996 > 10,00096
d-AMP (Elvanse, Attentin) 34 - 4196 , 206 (sulphate) 25 ** 23.3 - 38.9**96 , 54.8 (sulphate)25 3,830 - 11,00096
l-AMP 13896 , 1435 (sulphate) 25 ** 30.1**96 , 259 (sulphate)25 57,00096
ATX 1451 - 160096 235525 ** 2.6 - 5**96 , 20.625 ** 48 - 77**96
GBR-12909 40.225
Desipramine 4.925

2.8.2. Effect of AMP, MPH, ATX on dopamine / noradrenaline per brain region

The active substances methylphenidate (MPH), amphetamine (AMP) and atomoxetine (ATX) alter extracellular dopamine (DA) and noradrenaline (NE) to different degrees in different regions of the brain. Table based on Madras,96 modified.

PFC Striatum Nucleus accumbens
MPH DA +
NE (+)
DA +
NE +/- 0
DA +
NE +/- 0
AMP DA +
NE +
DA +
NE +/- 0
DA +
NE +/- 0
ATX DA +
NE +
DA +/- 0
NE +/- 0
DA +/- 0
NE +/- 0

3. Effect of amphetamine medication compared to MPH / atomoxetine

In MPH nonresponders, lisdexamfetamine (EU: Elvanse) and atomoxetine were compared in a randomized double-blind study with n = 200 subjects. Lisdexamfetamine was significantly more effective than atomoxetine in 2 of 6 categories and in the overall assessment.97
Lisdexamfetamine (EU: Elvanse) also had a good effect on comorbid depression symptoms in a double-blind study.98 MPH is not known to have any positive effects on depression symptoms.
A 2-year study in children and adolescents (n = 314) showed a responder rate of between 70 and 77 % with good efficacy and manageable side effects.99

4. Effect on ADHD symptoms

In ADHD sufferers who respond positively to D-amphetamine medication as well as MPH, the effect of D-amphetamine medication is at least on a par with MPH100, in our experience even significantly better in adults.

For a comparison of the effectiveness of individual medications and forms of treatment, see Effect strength of different forms of ADHD treatment.

According to the current European consensus, amphetamines are the first choice ADHD medication for adults (before methylphenidate) and the second choice for children (after methylphenidate).45

Amphetamine medication should also always be tried if MPH does not work (non-responder).

4.1. ADHD-I (without hyperactivity)

MPH has a stronger activating and drive-enhancing effect on most ADHD sufferers than AMP medication. Contrary reports101 are not consistent with our experience.
From our experience, we also cannot confirm statements in the specialist literature that amphetamine medication is more suitable for ADHD-I sufferers than MPH, partly because ADHD-I sufferers are above-average AMP nonresponders,102
We know a number of ADHD-HI sufferers who are significantly better helped by amphetamine medication than MPH and ADHD-I sufferers who cope better with MPH. We are not aware of any subtype-specific effect of amphetamine medication or methylphenidate. In our experience, amphetamine medication works just as well for ADHD-HI as for ADHD-I.

4.2. Attention control

ADHD sufferers have a reduced extrinsic and intrinsic motivational capacity. For example, they need higher rewards to be just as motivated for something as non-affected people. However, if the motivation of ADHD sufferers is aroused, their attention and controllability can no longer be reliably distinguished from that of non-affected individuals. Motivational shift towards own needs explains regulation problems
Attention correlates with a deactivation of the default mode network (DMN), among other things. Stimulants are able to align the attentional control of ADHD sufferers (or the ability to motivate, from which attention follows) with that of non-affected people, which is then also reflected in a normalization of DMN deactivation.103

More on the deviant function of the DMN in ADHD and its normalization by stimulants, including further sources at DMN (Default Mode Network) In the article Neurophysiological correlates of hyperactivity.
The cited references refer to the effect of methylphenidate. However, it can be assumed that the effect is achieved by stimulants in general.

Those affected report that MPH enables greater focus, while amphetamine medication (Elvanse) tends to create a more relaxed general alertness and has a somewhat more pleasant effect overall.

4.3. Comorbid depression or dysthymia

Amphetamine medications probably also have a mild serotonergic effect and thus have a special area of application in comorbid dysthymia or depression, especially since serotonin reuptake inhibitors (SSRIs) can have adverse effects in ADHD (especially ADHD-I) (see there).

In forums, a number of sufferers report a significant antidepressant effect from amphetamine medication, which they are not familiar with from MPH.104 This is consistent with the experiences of users known to us.

As amphetamines can have a stronger drive-boosting effect than MPH, this can release an existing suicidal tendency that was not previously carried out due to the existing depression. Amphetamine medication should therefore be used with caution in cases of (even concealed) severe depression.

Caution: a supposed dysthymia (mild chronic depression) in ADHD sufferers must be clearly differentiated from the original ADHD symptom of dysphoria during inactivity.
Find out more at Depression and dysphoria in ADHD In the section Differential diagnosis of ADHD.

4.4. Comorbid anxiety disorders / depression

Comorbid anxiety disorders or depression can be exacerbated by stimulants, as anxiety and moods are regulated by the dopaminergic activity of the ventromedial PFC in conjunction with the limbic system.38

4.5. Comorbid sleep disorders

Amphetamine drugs have a very long duration of action (up to 13 hours). Taking it too late (less than 14 hours before going to bed) could therefore cause problems falling asleep. On the other hand, some people taking amphetamine medication report feeling pleasantly tired in the evening and that they no longer have problems falling asleep.

Studies show that amphetamine medication improves the overall quality of sleep in ADHD.105106

4.6. Impulsiveness

Those affected reported in forums that MPH works better against impulsivity than Elvanse (lisdexamfetamine).107

5. Response rate (responding / non-responding)

Response here means whether there is an effect on the ADHD symptoms. Patients who do not respond sufficiently to a medication are called non-responders.
Non-responding does not mean having no effect, but merely that the effect remains below the level of symptom improvement specified in the respective study.

One study reported a responder rate of 80% (defined as an improvement of more than 30% in ADHD-RS-IV scores and CGI-I scores of greatly improved or very greatly improved)108
A summary of several studies reports a 69% response rate to amphetamine medication and a 59% response rate to methylphenidate. 87% of those affected responded to one of the two types of active substance.10
A 2-year study of L-amphetamine medication in children and adolescents (n = 314) showed a responder rate of between 70 and 77% with good efficacy and manageable side effects.99
For MPH non-responders, it is therefore highly recommended to test a medication with amphetamine drugs (see 1.2.), and vice versa.

In carriers of the COMT Val-158-Met gene polymorphism, amphetamine increased the efficiency of the PFC in subjects with presumably low levels of dopamine in the PFC. In contrast, in carriers of the COMT Met-158-Met polymorphism, amphetamine had no effect on cortical efficiency at low to moderate working memory load and caused a deterioration at high working memory load. Individuals with the Met-158-Met polymorphism appear to be at increased risk for an adverse response to amphetamine.109

6. No gender-specific differences in effect

Amphetamine drugs do not appear to show any gender-specific differences in effect.110

7. Calming effect at low doses, activating at high doses

D-amphetamine appears to have a biphasic action profile. Low doses of 0.5 to 1 mg/kg in rats (equivalent to about 0.2 to 0.6 mg/kg in humans) reduce (hyper)activity, while higher doses increase it.33

8. Dosage of amphetamine medication or MPH

Around 66% of all ADHD sufferers respond equally well to MPH and amphetamine medication.
22% respond better to amphetamine drugs than to MPH.
11% respond better to MPH than to amphetamine drugs.111
Around 15% of ADHD sufferers respond best to the active ingredient D-amphetamine.112

According to this result, it would make more sense to first try treatment with amphetamine medication and only try MPH as a second option in the event of non-response, as ADHD sufferers respond somewhat better to amphetamine medication than to MPH.

Highly gifted people with ADHD (here: IQ > 120) are said to respond better to amphetamine medication than less gifted people with ADHD.113

An interesting study discusses the effectiveness of lisdexamfetamine.114

It is advisable to start with a very low dosage, which is then slowly increased. Even if the optimal dosage were known, an immediate optimal dosage would possibly lead to excessive demands.115 The symptoms of ADHD are caused by signal transmission problems between the brain nerves because the neurotransmitter level (dopamine, noradrenaline) is too low. An optimal neurotransmitter level corrects the signal transmission problems. If the neurotransmitter level is too high due to an overdose, signal transmission is just as impaired as if the level is too low.
This explains why low doses should be given at the beginning and then, with patience, higher doses should be given until a worsening of symptoms is observed.

As the number of dopamine transporters in adults is half that of 10-year-olds, it is advisable to start with a much lower dosage than in children.

9. Effect profile (temporal) / duration of action

In replicated studies on the duration of action of amphetamine drugs, children had a shorter half-life of around 7 hours, while adults had a longer half-life of around 10 to 12 hours116

The time course of the effect (effect profile) depends less on the active ingredients than on the specific composition of the medication.
Elvanse has a very elongated effect profile without pronounced peaks, so that there are hardly any taper or rebound effects. See: Graphic representation of the Elvanse effect profile. However, the graph from Shire’s patent application refers to the plasma level in rats at an extremely high dose of 3 mg/kg.

Another graph shows the Of the active ingredients at 30 mg, 50 mg and 70 mg Elvanseon page 20.

The extent to which the binding of D-amphetamine to lysine in lisdexamfetamine really leads to a flattened and prolonged concentration of amphetamine in the blood plasma remains to be seen. A single dose of 40 mg D-amphetamine or 100 mg lisdexamfetamine (above the medically appropriate doses) in healthy people showed no relevant differences in amphetamine blood plasma concentration.117 Furthermore, the study data probably indicate a subjective impression of a gentler and longer effect of lisdexamfetamine on the part of the test subjects, although the authors do not report this. A further limitation of the study is that the subjects were treated with a single dose and there was no dosing to the tested dosage. The authors themselves cite studies showing that amphetamine drugs require familiarization phases or show (initial) habituation effects. The results of the study are therefore primarily of pharmacological interest, but only of limited practical use.

Empirically, adults report quite unanimously of a gentler and prolonged effect of lisdexamfetamine. The majority cite 5 to 7 hours as the duration of action of a single dose. There is also a fairly unanimous report of a very slow onset of action, with 1 to 2 hours being mentioned in most cases.

An internal (and not representative) Survey at adhs-forum.adx.org on the duration of effect of Elvanse (n= 80) and a further survey in a sub-reddit on Elvanse (n = 467) yielded the following result (n = 547):

Duration of action of a single dose of Elvanse % of participants
5 hours and less 40.8 %
6 to 7 hours 26.7 %
8 to 9 hours 15.4 %
10 to 11 hours 11 %
12 hours and more 6.2 %

The surveys are not representative (no consideration of age, weight, dose level or gender), but clearly show that a duration of action of 13 or 14 hours, as stated by the manufacturer, is only exceptionally achieved in adults in practice.
A more detailed Survey on the single-dose duration of action of all ADHD medicationswhich also includes the aforementioned secondary factors, has been running since March 2023 and could show initial results in fall 2023.

Many patients (we know many cases from the forum) take 2 or 3 single doses of Elvanse per day to achieve the required all-day coverage, even if this does not correspond to the manufacturer’s instructions. The individually shortened duration of action could also be the result of a low dosage of often 30 mg and less per single dose, which was chosen when an overdose was perceived at a higher single dose during the phase of high D-AMP blood plasma levels. The sum of the individual doses does not exceed 70 mg/day in almost all patients.
The result of a multiple intake of smaller Elvanse doses on the D-AMP blood plasma level could (purely hypothetically) look like this:

10. Areas of application of amphetamine drugs in relation to MPH

According to the current European consensus on the diagnosis and treatment of ADHD in adults, amphetamine medication is the first-choice ADHD medication for adults (before methylphenidate) and the second-choice medication for children (after methylphenidate)45
In children who are MPH non-responders, i.e. who do not respond to MPH, the efficacy of amphetamine medication should be tested.
Patients with pronounced dysphoria during inactivity or with comorbid depression benefit particularly from amphetamine medication.
In addition, sufferers who require stronger activation may be able to cope better with amphetamine medication.
Highly gifted people are said to respond better to amphetamine medication than to MPH.113

11. Side effects

11.1. No liver damage with normal medication dosage

High doses of amphetamines may be associated with liver damage and certain forms of clinically apparent liver damage. This is most commonly reported with methylenedioxymetamphetamine (MDMA: “ecstasy”).118

Amphetamine drugs, on the other hand, are dosed so low that this does not occur: the dose makes the poison. See also under Amphetamine medication versus amphetamine as a drug.

11.2 AMP increases histamine

AMP increases histamine,119120 like all other known ADHD medications:

  • Atomoxetine
  • Methylphenidate
  • Modafinil
  • Nicotine
  • Caffeine

This is why people with histamine intolerance often have problems when taking ADHD medication.
An ADHD sufferer with histamine intolerance reported that she could not tolerate AMP and sustained-release MPH at all, but was able to tolerate low doses of sustained-release MPH.

11.3. No increased cardiovascular risks

Several large studies found no increased risk of serious cardiovascular events such as stroke, heart attack or cardiac arrhythmia for amphetamine drugs.121122
A study over 14 years found an increase in the risk of cardiovascular problems of 4% per year of taking stimulants (methylphenidate, amphetamine drugs) and, to a lesser extent, the non-stimulant atomoxetine.123

11.4. Miscellaneous

Individual cases of trichotillomania (pulling out hair) have been reported.124 Trichotillomania is a specific form of impulse control disorder.

The drug MDMA (unlike amphetamine drugs) can damage nerve cells and attack the blood-brain barrier.125

Two male patients reported a loss of sensitivity in the genital area following the consumption of red wine outside the active period of the regularly taken Elvanse. In one of the patients, low nicotine consumption outside the active period is a further suspicious factor.
The package insert for Elvanse mentions erectile dysfunction in 1 in 10 out of 100 men.
Amphetamine drugs also bind to alpha1-adrenoceptors (see above).
A blockade of alpha1-adrenoceptors leads to a delayed detumescence of the erectile tissue and thus to a reduced ability to ejaculate and orgasm, both in women and in men.126 Blockade is the opposite of binding. Dopamine agonists such as L-dopa or bromocriptine cause an increase in sexual desire and sexual activity.

The literature or studies do not report any sexual impairment with amphetamine medication. A pilot study in men with sexual problems reported improvements in subjective sexual experience (reduced time to orgasm or increased frequency of orgasm) with 5 to 20 mg amphetamine salts (Adderall) 1 to 4 hours before sexual activity (up to 10 doses/month)127
In 5 individual cases, the resolution of SSRI-induced sexual dysfunction by small doses of dextroamphetamine or methylphenidate was reported.128 Further case studies report multiple erections (15-year-old), hypersexual behavior (8-year-old) due to OROS-MPH (Concerta)129 and priapism (14-year-old).130

Common side effects of amphetamine mixed salts are:27

  • Loss of appetite
  • Mood swings

Rare serious side effects of amphetamine mixed salts are:27

  • psychotic symptoms
  • Seizures
  • Risk of abuse

One study reported a doubled rate of testosterone deficiency in adult ADHD sufferers after 5 years of stimulant use (1.2%) compared to ADHD sufferers without stimulant use (0.67%) or non-affected individuals (0.68%).131

11.5. Overdose

Symptoms of a (severe) overdose of amphetamines (in the sense of intoxication) include

  • Agitation132
  • Hyperactivity133
  • Movement disorders132
  • Tremor132
  • Hyperthermia133
  • Tachycardia (rapid heartbeat)133
  • Tachypnea (increased respiratory rate)133
  • Mydriasis (pupil enlargement)133132
  • Trembling133
  • Seizures133, in extreme cases up to epileptic forms132
  • Hyperreflexia (excessive reflex response)132
  • combative behavior132
  • Confusion132
  • Hallucinations132
  • Delirium132
  • Fear132
  • Paranoia132

12. Breakdown of amphetamine

12.1. Reduction of LDX

Lisdexamfetamine (Elvanse) is converted to d-AMP in the blood cytosol of erythrocytes by an unknown amino acid (presumably an aminopeptidase)134135 by cleaving the covalent bond between d-amphetamine and L-lysine. Only d-AMP is pharmacologically active.

96% of LDX is excreted in the urine, of which23

  • 42 % of the dose as AMP
  • 25 % as hippuric acid
  • 2 % as intact LDX.

In contrast to AMP, LDX is less sensitive to changes in urine pH.
The half-life of LDX is typically less than 1 hour.

12.2. Degradation of D-AMP and L-AMP

D-AMP is metabolized faster than l-AMP, so that the exposure of d-AMP lasts 9-11 hours and of l-AMP 11-14 hours.
Taking it together with a high-fat meal can extend the half-life of d-AMP by one hour.

AMP is broken down in two ways23

  • Hydroxylation by CYP2D6:136135
    • 4-Hydroxyamphetamine
    • Noradrenaline (alpha-hydroxyamphetamine, norepinephrine)
    • both are subject to a further metabolism
  • oxidative deamination

A study came to different conclusions, according to which CYP2D6 is possibly hardly involved in the degradation of AMP.137 Other studies also tend to indicate that amphetamine itself is metabolized much less by CYP2D6 than some amphetamine analogues.138139
Nevertheless, amphetamine is a strong CYP2D6 inhibitor.139

AMP is primarily excreted via the kidneys.
Since AMP is slightly basic (pKA = 9.9), AMP excretion is highly dependent on urine pH and flow rate, with recovery of AMP in urine ranging from 1% to 75% and the remainder being metabolized hepatically:23

  • normal urine pH values
    • 30 to 40 % of the AMP dose is largely excreted as unchanged parent compound
    • 50% of the dose is excreted as alpha-hydroxyamphetamine or its downstream inactive metabolite, hippuric acid.
  • acidic urine (pH <6.0)
    • accelerated AMP excretion
  • alkaline urine (pH >7.5)
    • delayed AMP excretion

The half-life of AMP should increase by 7 hours per unit of pH increase. Acidifying or alkalizing agents can therefore significantly alter the effect of AMP.

12.3. Duration of action of AMP; influence of CYP2P6 metabolism types

The CYP2D6 gene is highly polymorphic. In Central Europe, the following alleles are particularly relevant140

  • CYP2D6*3
  • CYP2D6*4
  • CYP2D6*5
  • CYP2D6*6
  • CYP2D6*9
  • CYP2D6*41

Poor metabolizers are likely to require lower AMP doses and ultra-rapid metabolizers are likely to require higher AMP doses. However, the effects of CYP2D6 polymorphisms on AMP metabolism are still unclear.23

Based on the experience with the influence of CYP2D6 on the effect of other drugs (CYP2D6 is responsible for the metabolization of 20 - 30 % of all drugs), the different CYP2D6 gene variants lead to different types of metabolization140

  • Slow metabolizers - approx. 7 %
    • particularly slow dosing is important
    • particularly low dosage helpful
  • moderately fast metabolizers - approx. 40 %
  • Fast metabolizers - approx. 46 %
  • Ultra-fast metabolizers - approx. 7 %
    • CYP2D6*XN allele
    • increased enzyme activity
    • is associated with therapy resistance (non-responders)
    • increased dose can be helpful

More on this under –&gt CYP2D6 metabolizing enzyme

Two online surveys of a total of around 550 ADHD sufferers taking Elvanse showed that around 40% have a duration of action of 5 hours or less and two thirds have a single-dose duration of action of 7 hours or less. More on this under Effect and duration of action of ADHD medication

13. Contraindications and interactions

As with every drug described here, there are also contraindications for amphetamine drugs.
It should not be taken without prior medical consultation.

For lisdexamfetamine:141

  • Pregnancy / breastfeeding
    • A cohort study found no increased risk of ADHD or other neuronal developmental disorders due to MPH or AMP use during pregnancy.142
    • One study found no reduction in the weight of newborns of mothers with ADHD who took amphetamine medication during pregnancy.143 This is consistent with results from a large cohort study of MPH use during pregnancy.144
      Another comprehensive study found a slight reduction in birth weight and a slight increase in the risks of pre-eclampsia, placental abruption or premature birth when taking stimulants (AMP or MPH) during pregnancy, although this was so small that the authors did not recommend discontinuing stimulant use during pregnancy.145 Atomoxetine did not show these slight increases in risk.
      Another Danish cohort study found a doubled risk of miscarriage when taking stimulants during pregnancy.146
      Another Danish cohort study found an increase in malformations in children of mothers who had taken MPH in the first trimester of pregnancy, but the authors did not consider this to be relevant.147 A smaller study found no increased risk.148
      One study found no disadvantages for the child if the mother continued to take D-Amp during pregnancy. In contrast, an increased risk of abortion was found if D-Amp intake was discontinued during pregnancy. There were advantages if no D-Amp was taken before and during pregnancy. Early discontinuation could therefore be helpful if you wish to have children.149
  • Hypersensitivity to the active substance
  • Monoamine oxidase inhibitors (MAO inhibitors) at the same time or 14 days before use
    • Risk: hypertensive crisis
  • Hyperthyroidism / thyrotoxicosis
  • Excitation states
  • Symptomatic cardiovascular disease
  • Advanced arteriosclerosis
  • Moderate to severe hypertension
  • Glaucoma
  • Serotonin reuptake inhibitors
    • The risk of serotonin syndrome should be taken into account when administering SSRIs and amphetamine medication at the same time.18

According to a very large study, the risk of developing psychosis is lower for ADHD sufferers taking MPH (0.10%) than for those treated with amphetamine medication (0.21%).150 While ADHD sufferers treated with stimulants have 2.4 cases of psychosis per 1000 person-years, the figure for the population as a whole is 0.214%.151 The studies do not allow any conclusion to be drawn as to whether the increased prevalence of psychosis is attributable to ADHD or stimulants.

13.1. Shortened half-life (reduced effect)

Attenuation of the effects of dexamphetamine:152

  • Adrenoreceptor blockers (beta blockers)
    • e.g.:
      • Propranolol
  • Lithium
  • Phenothiazines
  • Haloperidol
  • Substances that lower the pH value in the gastrointestinal tract
    • e.g:
      • Guanethidine
      • Reserpine
      • Glutamic acid
      • Hydrochloric acid
      • Ascorbic acid
      • Fruit juice
    • cause reduced absorption of dexamphetamine
  • Substances that acidify urine (ammonium chloride, sodium dihydrogen phosphate, etc.)
    • increase ionized excretion products of dexamphetamine in urine, resulting in increased renal excretion
  • Acidification of the urine (reduced pH value)141
    e.g. through
    • Ascorbic acid
    • Thiazide diuretics
    • High protein diet
    • Diabetes mellitus

Caution: Foods that taste acidic often have an alkalizing effect in the body beyond the digestive tract.
Example: Lemon juice has a pH value of 2.4 and therefore has an acidic effect on the mouth and stomach. After digestion, however, only an alkaline residue remains in the rest of the body, which increases the pH value.
The effect of food after digestion on the acid load of minerals and protein on the kidneys is indicated by the PRAL value (potential renal acid load). This value is not suitable for assessing the acid load of the mouth and stomach (as is relevant for heartburn).
The higher the PRAL value, the more acidic the effect on the kidneys and the rest of the body after the digestive organs.

Urine pH has been shown to be a good PRAL marker. An alkaline urine pH value correlates with a diet with a negative PRAL value, while urine pH values below 6.0 correlate with an acidifying diet.
A distinction must be made between plant and animal proteins. After 7 days of a vegetarian diet, the pH urine value increases and the PRAL value decreases, as does 2 or 3 days of a vegetarian diet per week.153 A vegetarian diet thus correlates with a prolonged amphetamine drug effect.
Foods with a high oxalate content can increase acid formation.154
One study gives the following calculation method:155 PRAL (mEq/d) = 0.49 x protein (g/d) + 0.037 x phosphorus (mg/d) - 0.021 x potassium (mg/d) - 0.026 x magnesium (mg/d) - 0.013 x calcium (mg/d).

In other words, foods with a strongly negative PRAL value cause alkaline (less acidic) urine and thus promote a prolonged effect of amphetamine drugs. Foods with a high PRAL value cause acidic urine and thus promote a shortened effect of amphetamine drugs. According to this model, hard cheese is suitable for shortening the effects of amphetamine drugs, while raisins could prolong them.

More on this under Acid balance and amphetamine medication In the article Effect and duration of action of ADHD medication

13.2. Prolonged half-life (increased effect)

  • With alkalized urine (increased pH value)141
    e.g. through
    • Sodium hydrogen carbonate (baking powder, soda)
    • Diet with a high fruit / vegetable content
    • Urinary tract infections
    • Vomiting
    • Clonidine152

Enhanced effect of dexamphetamine due to:152

  • Disulfiram
  • Substances that increase the pH value in the gastrointestinal tract increase dexamphetamine uptake
    • e.g:
      • Sodium bicarbonate (baking powder)
  • Substances that increase urine pH increase non-ionized excretion products in urine, which decreases renal excretion and thus increases blood levels of dexamphetamine
    • e.g:
      • Acetazolamide
      • some thiazides

There is evidence that reduced expression of the CACNA1C gene can lead to a prolonged effect of dopamine reuptake inhibitors.156 Conversely, increased CACNA1C expression may lead to a shortened effect.

13.3. Delayed effect

Lisdexamfetamine (Elvanse) has a maximum blood level that is delayed by one hour with high-fat meals (4.7 hours instead of 3.8 hours after ingestion).157 However, other parameters, such as the duration of action, do not change.

13.4. Amplifying effect on amphetamines

13.4.1. Alcohol increases amphetamine levels

Alcohol can increase amphetamine levels.158

13.4.2. CYP2D6 inhibitors increase amphetamine levels

Since amphetamine is broken down by CYP2D6, drugs that are also broken down by CYP2D6 can slow down the breakdown of amphetamine as well as their own breakdown, as competition for the CYP2D6 enzyme arises.
CYP2D6 inhibitors can increase amphetamine levels, making a dose reduction necessary. After discontinuation of CYP2D6 inhibitors, an increase in the dose of amphetamine medication may be necessary.158
CYP2D6 inducers can accelerate degradation and thus reduce the effect.

See under CYP2D6 metabolizing enzyme

13.5. Attenuating effect on amphetamines

Have an attenuating effect on amphetamine:159

  • Chlorpromazine
  • Haloperidol
  • Lithium carbonate

A single patient reported a loss of efficacy of Elvanse due to Dienogest 2 mg (Zafrilla) in endometriosis, while the effect of Attentin remained unchanged.

13.6. Few interactions of AMP with other medications

In contrast to the above-mentioned interactions of amphetamine medications with other medications, hardly any interactions of amphetamine medications with other medications are known.158

Amphetamine is said to have a slight inhibitory effect on the cytochromes

  • CYP2D6
  • CYP1A2
  • CYP3A4.

The clinical relevance is classified as low.159

Attenuating effect on159

  • Antihypertensives such as guanethidine

Reinforcing on159

  • Analgesic effect of opioids

14. Long-term effect: No habituation effects of amphetamine medication

A meta-analysis of 87 randomized placebo-controlled double-blind studies found no evidence of a decrease in the effect of methylphenidate, amphetamine drugs, atomoxetine or α2 antagonists with prolonged use.160

15. Preparations

Amphetamine medications are available as various preparations.

In the USA, amphetamine drugs are available as:1

  • Mixture of D- and L-amphetamine isomers (racemic mixture)
  • Mixed sulfates and saccharinates of D-L-amphetamine isomers (Adderall)
  • Pure D-amphetamine sulphate
    • Dexamfetamine hemisulfate (Attentin)
  • D-amphetamine as lisdexamfetamine in lysine-bound form (Elvanse, Tyvense)
  • Racemic methamphetamine sulphate (Desoxyn)

Elvanse, Tyvense

Elvanse / Tyvense contains lisdexamfetamine. Lisdexamfetamine is dextroamphetamine bound to lysine. The lysine binding causes a very slow and even release of dextroamphetamine in the blood and thus a prolonged effect.
Available in the USA:
Capsules: 10, 20, 30, 40, 50, 60, 70 mg
Chewable tablets: 10, 20, 30, 40, 50, 60 mg

16. Taking amphetamine medication abroad

See under Taking stimulants abroad


  1. Edel, Vollmoeller (2006): Aufmerksamkeitsdefizit-/Hyperaktivitätsstörung bei Erwachsenen, Springer, Seite 57, Stand 2006

  2. so noch Edel, Vollmoeller (2006): Aufmerksamkeitsdefizit-/Hyperaktivitätsstörung bei Erwachsenen, Springer, Seite 57

  3. Rösler, Retz (2020): Medikamentöse Therapie der ADHS bei Erwachsenen; Psychiatrie up2date 2020; 14: 59–75

  4. Kooij, Bijlenga, Salerno, Jaeschke, Bitter, Balázs, Thome, Dom, Kasper, Filipe, Stes, Mohr, Leppämäki, Brugué, Bobes, Mccarthy, Richarte, Philipsen, Pehlivanidis, Niemela, Styr, Semerci, Bolea-Alamanac, Edvinsson, Baeyens, Wynchank, Sobanski, Philipsen, McNicholas, Caci, Mihailescu, Manor, Dobrescu, Krause, Fayyad, Ramos-Quiroga, Foeken, Rad, Adamou, Ohlmeier, Fitzgerald, Gill, Lensing, Mukaddes, Brudkiewicz, Gustafsson, Tania, Oswald, Carpentier, De Rossi, Delorme, Simoska, Pallanti, Young, Bejerot, Lehtonen, Kustow, Müller-Sedgwick, Hirvikoski, Pironti, Ginsberg, Félegeházy, Garcia-Portilla, Asherson (2018): Updated European Consensus Statement on diagnosis and treatment of adult ADHD, European Psychiatrie, European Psychiatry 56 (2019) 14–34, http://dx.doi.org/10.1016/j.eurpsy.2018.11.001, Seite 22, 7.4.1.

  5. Cortese, Adamo, Del Giovane, Mohr-Jensen, Hayes, Carucci, Atkinson, Tessari, Banaschewski, Coghill, Hollis, Simonoff, Zuddas, Barbui, Purgato, Steinhausen, Shokraneh, Xia, Cipriani (2018): Comparative efficacy and tolerability of medications for attention-deficit hyperactivity disorder in children, adolescents, and adults: a systematic review and network meta-analysis. Lancet Psychiatry. 2018 Sep;5(9):727-738. doi: 10.1016/S2215-0366(18)30269-4.

  6. https://www.awmf.org/uploads/tx_szleitlinien/028-045k_S3_ADHS_2018-06.pdf, S 40/81

  7. Banaschewski T (Leitlinienkoordinator), Hohmann, Millenet et al (2018): Langfassung der interdisziplinären evidenz- und konsensbasierten (S3) Leitlinie „Aufmerksamkeitsdefizit- / Hyperaktivitätsstörung (ADHS) im Kindes-, Jugend- und Erwachsenenalter“ AWMF-Registernummer 028-045, S. 72/198

  8. Lisdexamfetamin – jetzt auch für Erwachsene zugelassen – Kompendium psychiatrische Pharmakotherapie, Springer.

  9. Frölich, Banaschewski, Spanagel, Döpfner, Lehmkuhl (2012): Die medikamentöse Behandlung der Aufmerksamkeitsdefizit-Hyperaktivitätsstörung im Kindes- und Jugendalter mit Amphetaminpräparaten; Zeitschrift für Kinder- und Jugendpsychiatrie und Psychotherapie (2012), 40, pp. 287-300. https://doi.org/10.1024/1422-4917/a000185

  10. Arnold: Journal of Attention Disorders Vol. 3(4):200-211 (2000) Methylphenidate vs. amphetamine: Comparative review, n = 174

  11. Krause, Krause (2014): ADHS im Erwachsenenalter, Schattauer. Fallbeispiel S. 155

  12. Madhoo, Keefe, Roth, Sambunaris, Wu, Trivedi, Anderson, Lasser (2014): Lisdexamfetamine dimesylate augmentation in adults with persistent executive dysfunction after partial or full remission of major depressive disorder; Neuropsychopharmacology. 2014 May;39(6):1388-98. doi: 10.1038/npp.2013.334. n=143 Erwachsene

  13. Castells X1, Ramos-Quiroga JA, Bosch R, Nogueira M, Casas (2011): Amphetamines for Attention Deficit Hyperactivity Disorder (ADHD) in adults; Cochrane Database Syst Rev. 2011 Jun 15;(6):CD007813. doi: 10.1002/14651858.CD007813.pub2. n = 1071

  14. Childress, Komolova, Sallee (2019): An update on the pharmacokinetic considerations in the treatment of ADHD with long-acting methylphenidate and amphetamine formulations. Expert Opin Drug Metab Toxicol. 2019 Nov;15(11):937-974. doi: 10.1080/17425255.2019.1675636. PMID: 31581854. REVIEW

  15. Edel, Vollmoeller (2006): Aufmerksamkeitsdefizit-/Hyperaktivitätsstörung bei Erwachsenen, Springer, Seite 57

  16. Stahl (2013): Stahl’s Essential Psychopharmacology, 4. Auflage, Chapter 12: Attention deficit hyperactivity disorder and its treatment, Seite 491

  17. Castellanos, Tannock (2002): Neuroscience of attention-deficit/hyperactivity disorder: The search for endophenotypes; Article in Nature reviews Neuroscience 3(8):617-28 · September 2002; DOI: 10.1038/nrn896, Seite 621

  18. Davis, Hernandez, Stock (2020): Adolescent Polypharmacy and Serotonin Syndrome. Clin Neuropharmacol. 2020 Jan/Feb;43(1):28-30. doi: 10.1097/WNF.0000000000000375. PMID: 31934921.

  19. Prox-Vagedes, Ohlmeier in Ohlmeier, Roy (Hrsg.) (2012): ADHS bei Erwachsenen – Ein Leben in Extremen, Kapitel 5: Die Suche nach dem Rausch: Substanzabhängigkeit bei ADHS Seite 101, mwNw

  20. Sharman J, Pennick M (2023): Lisdexamfetamine prodrug activation by peptidase-mediated hydrolysis in the cytosol of red blood cells. Neuropsychiatr Dis Treat. 2014 Nov 28;10:2275-80. doi: 10.2147/NDT.S70382. PMID: 25489246; PMCID: PMC4257105.

  21. https://www.adhspedia.de/wiki/Umrechnungstabelle_Medikamente

  22. Pharmazeutische Zeitung: Lisdexamfetamin|Elvanse®|71|2013

  23. Childress, Komolova, Sallee (2019): An update on the pharmacokinetic considerations in the treatment of ADHD with long-acting methylphenidate and amphetamine formulations. Expert Opin Drug Metab Toxicol. 2019 Nov;15(11):937-974. doi: 10.1080/17425255.2019.1675636. PMID: 31581854.

  24. Medikamio.com: Elvanse Abruf 09.07.2023

  25. Easton N, Steward C, Marshall F, Fone K, Marsden C (2007): Effects of amphetamine isomers, methylphenidate and atomoxetine on synaptosomal and synaptic vesicle accumulation and release of dopamine and noradrenaline in vitro in the rat brain. Neuropharmacology. 2007 Feb;52(2):405-14. doi: 10.1016/j.neuropharm.2006.07.035. PMID: 17020775.

  26. Huberman (2023): Adderall, Stimulants & Modafinil for ADHD: Short- & Long-Term Effects | Huberman Lab Podcast, english

  27. Buoli, Serati, Cahn (2016): Alternative pharmacological strategies for adult ADHD treatment: a systematic review. Expert Rev Neurother. 2016;16(2):131-44. doi: 10.1586/14737175.2016.1135735. PMID: 26693882. REVIEW

  28. Fouladvand, Hankosky, Bush, Chen, Dwoskin, Freeman, Henderson, Kantak, Talbert, Tao, Zhang (2019): Predicting substance use disorder using long-term attention deficit hyperactivity disorder medication records in Truven. Health Informatics J. 2019 May 19:1460458219844075. doi: 10.1177/1460458219844075.

  29. Heal, Cheetham, Smith (2009): The neuropharmacology of ADHD drugs in vivo: insights on efficacy and safety. Neuropharmacology. 2009 Dec;57(7-8):608-18. doi: 10.1016/j.neuropharm.2009.08.020.

  30. Faraone (2018): The pharmacology of amphetamine and methylphenidate: Relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev. 2018 Apr;87:255-270. doi: 10.1016/j.neubiorev.2018.02.001. PMID: 29428394.

  31. Visentin R, Dalla Man C, Kovatchev B, Cobelli C (2014): The university of Virginia/Padova type 1 diabetes simulator matches the glucose traces of a clinical trial. Diabetes Technol Ther. 2014 Jul;16(7):428-34. doi: 10.1089/dia.2013.0377. PMID: 24571584; PMCID: PMC4074748.

  32. Gutiérrez-Casares JR, Quintero J, Segú-Vergés C, Rodríguez Monterde P, Pozo-Rubio T, Coma M, Montoto C (2023): In silico clinical trial evaluating lisdexamfetamine’s and methylphenidate’s mechanism of action computational models in an attention-deficit/hyperactivity disorder virtual patients’ population. Front Psychiatry. 2023 Jun 2;14:939650. doi: 10.3389/fpsyt.2023.939650. PMID: 37333910; PMCID: PMC10273406.

  33. Seeman P, Madras BK (1998): Anti-hyperactivity medication: methylphenidate and amphetamine. Mol Psychiatry. 1998 Sep;3(5):386-96. doi: 10.1038/sj.mp.4000421. PMID: 9774771. REVIEW

  34. Covey DP, Juliano SA, Garris PA (2013): Amphetamine elicits opposing actions on readily releasable and reserve pools for dopamine. PLoS One. 2013 May 3;8(5):e60763. doi: 10.1371/journal.pone.0060763. PMID: 23671560; PMCID: PMC3643976.

  35. Sulzer D, Rayport S (1990): Amphetamine and other psychostimulants reduce pH gradients in midbrain dopaminergic neurons and chromaffin granules: a mechanism of action. Neuron. 1990 Dec;5(6):797-808. doi: 10.1016/0896-6273(90)90339-h. PMID: 2268433.

  36. Schmitz Y, Schmauss C, Sulzer D. Altered dopamine release and uptake kinetics in mice lacking D2 receptors. J Neurosci. 2002 Sep 15;22(18):8002-9. doi: 10.1523/JNEUROSCI.22-18-08002.2002. PMID: 12223553; PMCID: PMC6758092.

  37. Frölich, Banaschewski, Spanagel, Döpfner, Lehmkuhl (2012): Die medikamentöse Behandlung der Aufmerksamkeitsdefizit-Hyperaktivitätsstörung im Kindes- und Jugendalter mit Amphetaminpräparaten; Zeitschrift für Kinder- und Jugendpsychiatrie und Psychotherapie (2012), 40, pp. 287-300. https://doi.org/10.1024/1422-4917/a000185

  38. Stahl (2013): Stahl’s Essential Psychopharmacology, 4. Auflage, Chapter 12: Attention deficit hyperactivity disorder and its treatment, Seite 490

  39. Steinhausen, Rothenberger, Döpfner (2010): Handbuch ADHS, Seite 84, 85

  40. Castellanos, Tannock (2002): Neuroscience of attention-deficit/hyperactivity disorder: The search for endophenotypes; Article in Nature reviews Neuroscience 3(8):617-28 · September 2002; DOI: 10.1038/nrn896, Seite 621, mwNw

  41. Khoshbouei H, Sen N, Guptaroy B, Johnson L’, Lund D, Gnegy ME, Galli A, Javitch JA (2004): N-terminal phosphorylation of the dopamine transporter is required for amphetamine-induced efflux. PLoS Biol. 2004 Mar;2(3):E78. doi: 10.1371/journal.pbio.0020078. PMID: 15024426; PMCID: PMC368172.

  42. Daniels GM, Amara SG (1999):. Regulated trafficking of the human dopamine transporter. Clathrin-mediated internalization and lysosomal degradation in response to phorbol esters. J Biol Chem. 1999 Dec 10;274(50):35794-801. doi: 10.1074/jbc.274.50.35794. PMID: 10585462.

  43. Granas C, Ferrer J, Loland CJ, Javitch JA, Gether U (2003): N-terminal truncation of the dopamine transporter abolishes phorbol ester- and substance P receptor-stimulated phosphorylation without impairing transporter internalization. J Biol Chem. 2003 Feb 14;278(7):4990-5000. doi: 10.1074/jbc.M205058200. PMID: 12464618.

  44. Giambalvo CT (2003): Differential effects of amphetamine transport vs. dopamine reverse transport on particulate PKC activity in striatal synaptoneurosomes. Synapse. 2003 Aug;49(2):125-33. doi: 10.1002/syn.10223. PMID: 12740868.

  45. Foster JD, Pananusorn B, Vaughan RA (2002): Dopamine transporters are phosphorylated on N-terminal serines in rat striatum. J Biol Chem. 2002 Jul 12;277(28):25178-86. doi: 10.1074/jbc.M200294200. PMID: 11994276.

  46. Mulvihill (2019): Presynaptic regulation of dopamine release: Role of the DAT and VMAT2 transporters. Neurochem Int. 2019 Jan;122:94-105. doi: 10.1016/j.neuint.2018.11.004. PMID: 30465801.

  47. Edinoff, Akuly, Wagner, Boudreaux, Kaplan, Yusuf, Neuchat, Cornett, Boyer, Kaye, Kaye (2021): Viloxazine in the Treatment of Attention Deficit Hyperactivity Disorder. Front Psychiatry. 2021 Dec 17;12:789982. doi: 10.3389/fpsyt.2021.789982. PMID: 34975586; PMCID: PMC8718796., REVIEW

  48. Daberkow DP, Brown HD, Bunner KD, Kraniotis SA, Doellman MA, Ragozzino ME, Garris PA, Roitman MF (2013): Amphetamine paradoxically augments exocytotic dopamine release and phasic dopamine signals. J Neurosci. 2013 Jan 9;33(2):452-63. doi: 10.1523/JNEUROSCI.2136-12.2013. PMID: 23303926; PMCID: PMC3711765.

  49. Mergy MA, Gowrishankar R, Davis GL, Jessen TN, Wright J, Stanwood GD, Hahn MK, Blakely RD (2014): Genetic targeting of the amphetamine and methylphenidate-sensitive dopamine transporter: on the path to an animal model of attention-deficit hyperactivity disorder. Neurochem Int. 2014 Jul;73:56-70. doi: 10.1016/j.neuint.2013.11.009. PMID: 24332984; PMCID: PMC4177817.

  50. Saunders C, Ferrer JV, Shi L, Chen J, Merrill G, Lamb ME, Leeb-Lundberg LM, Carvelli L, Javitch JA, Galli A (2000): Amphetamine-induced loss of human dopamine transporter activity: an internalization-dependent and cocaine-sensitive mechanism. Proc Natl Acad Sci U S A. 2000 Jun 6;97(12):6850-5. doi: 10.1073/pnas.110035297. PMID: 10823899; PMCID: PMC18764.

  51. Mulvihill (2019): Presynaptic regulation of dopamine release: Role of the DAT and VMAT2 transporters. Neurochem Int. 2019 Jan;122:94-105. doi: 10.1016/j.neuint.2018.11.004. PMID: 30465801. REVIEW

  52. Quansah, Ruiz-Rodado, Grootveld, Zetterström (2018): Methylphenidate alters monoaminergic and metabolic pathways in the cerebellum of adolescent rats. Eur Neuropsychopharmacol. 2018 Feb 22. pii: S0924-977X(18)30043-9. doi: 10.1016/j.euroneuro.2018.02.002.

  53. Quansah, Zetterström (2019): Chronic methylphenidate preferentially alters catecholamine protein targets in the parietal cortex and ventral striatum. Neurochem Int. 2019 Jan 17;124:193-199. doi: 10.1016/j.neuint.2019.01.016.

  54. Felmer AC, Janson MT, Summers KE, Wallace LJ (2019):. Extracellular dopamine kinetic parameters consistent with amphetamine effects. Synapse. 2019 Dec;73(12):e22129. doi: 10.1002/syn.22129. PMID: 31449701.

  55. Jones SR, Gainetdinov RR, Wightman RM, Caron MG (1998): Mechanisms of amphetamine action revealed in mice lacking the dopamine transporter. J Neurosci. 1998 Mar 15;18(6):1979-86. doi: 10.1523/JNEUROSCI.18-06-01979.1998. PMID: 9482784; PMCID: PMC6792915.

  56. Schmitz Y, Lee CJ, Schmauss C, Gonon F, Sulzer D (2001): Amphetamine distorts stimulation-dependent dopamine overflow: effects on D2 autoreceptors, transporters, and synaptic vesicle stores. J Neurosci. 2001 Aug 15;21(16):5916-24. doi: 10.1523/JNEUROSCI.21-16-05916.2001. PMID: 11487614; PMCID: PMC6763160.

  57. Müller, Candrian, Kropotov (2011), ADHS – Neurodiagnostik in der Praxis, Springer, Seiten 21, 86

  58. Ruskin DN, Bergstrom DA, Shenker A, Freeman LE, Baek D, Walters JR (2001): Drugs used in the treatment of attention-deficit/hyperactivity disorder affect postsynaptic firing rate and oscillation without preferential dopamine autoreceptor action. Biol Psychiatry. 2001 Feb 15;49(4):340-50. doi: 10.1016/s0006-3223(00)00987-2. PMID: 11239905.

  59. Seeman P, Madras BK (1998): Anti-hyperactivity medication: methylphenidate and amphetamine. Mol Psychiatry. 1998 Sep;3(5):386-96. doi: 10.1038/sj.mp.4000421. PMID: 9774771. REVIEW

  60. Janenaite, Vengeliene, Bespalov, Behl (2017): Potential role of tyrosine hydroxylase in the loss of psychostimulant effect of amphetamine under conditions of impaired dopamine transporter activity. Behav Brain Res. 2017 Sep 15;334:105-108. doi: 10.1016/j.bbr.2017.07.028. PMID: 28750831.

  61. Rebec GV, Zimmerman KS (1980): Opposite effects of D-amphetamine on spontaneous neuronal activity in the neostriatum and nucleus accumbens. Brain Res. 1980 Nov 17;201(2):485-91. doi: 10.1016/0006-8993(80)91058-6. PMID: 7191347.

  62. Hansen EL, McKenzie GM (1979): Dexamphetamine increases striatal neuronal firing in freely moving rats. Neuropharmacology. 1979 Jun;18(6):547-52. doi: 10.1016/0028-3908(79)90099-6. PMID: 573376.

  63. Shi WX, Pun CL, Zhang XX, Jones MD, Bunney BS (2000): Dual effects of D-amphetamine on dopamine neurons mediated by dopamine and nondopamine receptors. J Neurosci. 2000 May 1;20(9):3504-11. doi: 10.1523/JNEUROSCI.20-09-03504.2000. PMID: 10777813; PMCID: PMC6773133.

  64. Newcorn JH, Ivanov I, Krone B, Li X, Duhoux S, White S, Schulz KP, Bédard AV, Pedraza J, Adler L, Blair RJ (2023): Neurobiological basis of reinforcement-based decision making in adults with ADHD treated with lisdexamfetamine dimesylate: Preliminary findings and implications for mechanisms influencing clinical improvement. J Psychiatr Res. 2023 Dec 3;170:19-26. doi: 10.1016/j.jpsychires.2023.11.037. PMID: 38101205.

  65. Marinelli M, McCutcheon JE (2014): Heterogeneity of dopamine neuron activity across traits and states. Neuroscience. 2014 Dec 12;282:176-97. doi: 10.1016/j.neuroscience.2014.07.034. PMID: 25084048; PMCID: PMC4312268. REVIEW

  66. Paladini CA, Fiorillo CD, Morikawa H, Williams JT (2001): Amphetamine selectively blocks inhibitory glutamate transmission in dopamine neurons. Nat Neurosci. 2001 Mar;4(3):275-81. doi: 10.1038/85124. PMID: 11224544.

  67. Howlett DR, Nahorski SR (1979): Acute and chronic amphetamine treatments modulate striatal dopamine receptor binding sites. Brain Res. 1979 Jan 26;161(1):173-8. doi: 10.1016/0006-8993(79)90206-3. PMID: 758968.

  68. Mang (2018): 05. Monoamine 2: Amphetamin, Ritalin (ADHS), Cocain, Tricyclika, Videovorlesung. ca. bei Minute 14:50.

  69. Kuczenski R, Segal DS (1997): Effects of methylphenidate on extracellular dopamine, serotonin, and norepinephrine: comparison with amphetamine. J Neurochem. 1997 May;68(5):2032-7. doi: 10.1046/j.1471-4159.1997.68052032.x. Erratum in: J Neurochem 1997 Sep;69(3):1332. PMID: 9109529.

  70. Lahey (Hrsg.) (2012): Advances in Clinical Child Psychology, Band 9, S. 187, mit Verweis auf [Brown, Ebert, Hunt, Rapoport (1981): Urinary 3-methoxy-4-hydroxyphenylglycol and homovanillic acid response to d-amphetamine in hyperactive children; Biological Psychiatry, Vol 16(8), Aug 1981, 779-787

  71. Shekim, Dekirmenjian, Chapel, (1979): Urinary MHPG excretion in minimal brain dysfunction and its modification by d-amphetamine; The American Journal of Psychiatry, Vol 136(5), May 1979, 667-671. http://dx.doi.org/10.1176/ajp.136.5.667

  72. Shen, Wang (1984): Urinary 3-methoxy-4-hydroxyphenylglycol sulfate excretion in seventy-three schoolchildren with minimal brain dysfunction syndrome.Biological Psychiatry [1984, 19(6):861-870]; PMID:6743722

  73. Lahey (Hrsg.) (2012): Advances in Clinical Child Psychology, Band 9, S. 187, mit Verweis u.a. auf Shekim, Dekirmenjian, Chapel, (1979): Urinary MHPG excretion in minimal brain dysfunction and its modification by d-amphetamine; The American Journal of Psychiatry, Vol 136(5), May 1979, 667-671. http://dx.doi.org/10.1176/ajp.136.5.667

  74. Paladini CA, Williams JT (2004): Noradrenergic inhibition of midbrain dopamine neurons. J Neurosci. 2004 May 12;24(19):4568-75. doi: 10.1523/JNEUROSCI.5735-03.2004. PMID: 15140928; PMCID: PMC6729397.

  75. Shi WX, Zhang XY, Pun CL, Bunney BS (2007): Clozapine blocks D-amphetamine-induced excitation of dopamine neurons in the ventral tegmental area. Neuropsychopharmacology. 2007 Sep;32(9):1922-8. doi: 10.1038/sj.npp.1301334. PMID: 17299514.

  76. Shen, Shi (2020): Amphetamine promotes cortical Up state: Role of adrenergic receptors. Addict Biol. 2020 Jan 31;e12879. doi: 10.1111/adb.12879. PMID: 32003119.

  77. Shen, Shi (2021): Amphetamine Promotes Cortical Up State in Part Via Dopamine Receptors. Front Pharmacol. 2021 Aug 19;12:728729. doi: 10.3389/fphar.2021.728729. PMID: 34489713; PMCID: PMC8417369.

  78. Mang (2018): 05. Monoamine 2: Amphetamin, Ritalin (ADHS), Cocain, Tricyclika, Videovorlesung. ca. bei Minute 14:30.

  79. Castellanos FX, Acosta MT (2011): Hacia un entendimiento de los mecanismos moleculares de los tratamientos farmacologicos del trastorno por deficit de atencion/hiperactividad [Towards an understanding of the molecular mechanisms underlying the pharmacological treatments of attention deficit hyperactivity disorder]. Rev Neurol. 2011 Mar 1;52 Suppl 1:S155-60. Spanish. PMID: 21365598. REVIEW

  80. Heinz (2000, 2013): Das dopaminerge Verstärkungssystem – Funktion, Interaktion mit anderen Neurotransmittersystemen und psychopathologische Korrelate, Seite 61

  81. Callaway, Johnson, Gold, Nichols, Geyer (1991): Amphetamine derivatives induce locomotor hyperactivity by acting as indirect serotonin agonists; Psychopharmacology (Berl). 1991;104(3):293-301

  82. Tsuchida, Kubo, Shintani, Abe, Köves, Uetsuki, Kuroda, Hashimoto, Baba (2009): Inhibitory effects of osemozotan, a serotonin 1A-receptor agonist, on methamphetamine-induced c-Fos expression in prefrontal cortical neurons. Biol Pharm Bull. 2009 Apr;32(4):728-31. doi: 10.1248/bpb.32.728. PMID: 19336914.

  83. Strajhar, Vizeli, Patt, Dolder, Kratschmar, Liechti, Odermatt (2018): Effects of lisdexamfetamine on plasma steroid concentrations compared with d-amphetamine in healthy subjects: A randomized, double-blind, placebo-controlled study. J Steroid Biochem Mol Biol. 2019 Feb;186:212-225. doi: 10.1016/j.jsbmb.2018.10.016. PMID: 30381248.

  84. Soto, Wilcox, Zhou, Kumar, Ator, Riddle, Wong, Weed (2013): Long-term exposure to oral methylphenidate or dl-amphetamine mixture in peri-adolescent rhesus monkeys: effects on physiology, behavior, and dopamine system development. Neuropsychopharmacology. 2012 Nov;37(12):2566-79. doi: 10.1038/npp.2012.119. Erratum in: Neuropsychopharmacology. 2013 May;38(6):1141. Kumar, Anil [added]. PMID: 22805599; PMCID: PMC3473325.

  85. Mattison, Plant, Lin, Chen, Chen, Twaddle, Doerge, Slikker, Patton, Hotchkiss, Callicott, Schrader, Turner, Kesner, Vitiello, Petibone, Morris (2011): Pubertal delay in male nonhuman primates (Macaca mulatta) treated with methylphenidate. Proc Natl Acad Sci U S A. 2011 Sep 27;108(39):16301-6. doi: 10.1073/pnas.1102187108. PMID: 21930929; PMCID: PMC3182701.

  86. Tsai, Chiao, Lu, Doong, Chen, Shih, Liaw, Wang, Wang (1996): Inhibition by amphetamine of testosterone secretion through a mechanism involving an increase of cyclic AMP production in rat testes. Br J Pharmacol. 1996 Jun;118(4):984-8. doi: 10.1111/j.1476-5381.1996.tb15496.x. PMID: 8799572; PMCID: PMC1909523.

  87. Tsai, Chen, Chiao, Lu, Lin, Yeh, Lo, Kau, Wang, Wang (1997): The role of cyclic AMP production, calcium channel activation and enzyme activities in the inhibition of testosterone secretion by amphetamine. Br J Pharmacol. 1997 Nov;122(5):949-55. doi: 10.1038/sj.bjp.0701463. PMID: 9384514; PMCID: PMC1565017.

  88. Angenoorth TJF, Stankovic S, Niello M, Holy M, Brandt SD, Sitte HH, Maier J (2021): Interaction Profiles of Central Nervous System Active Drugs at Human Organic Cation Transporters 1-3 and Human Plasma Membrane Monoamine Transporter. Int J Mol Sci. 2021 Nov 30;22(23):12995. doi: 10.3390/ijms222312995. PMID: 34884800; PMCID: PMC8657792.

  89. Amphoux A, Vialou V, Drescher E, Brüss M, Mannoury La Cour C, Rochat C, Millan MJ, Giros B, Bönisch H, Gautron S (2006): Differential pharmacological in vitro properties of organic cation transporters and regional distribution in rat brain. Neuropharmacology. 2006 Jun;50(8):941-52. doi: 10.1016/j.neuropharm.2006.01.005. PMID: 16581093.

  90. Orrico-Sanchez A, Chausset-Boissarie L, Alves de Sousa R, Coutens B, Rezai Amin S, Vialou V, Louis F, Hessani A, Dansette PM, Zornoza T, Gruszczynski C, Giros B, Guiard BP, Acher F, Pietrancosta N, Gautron S (2020): Antidepressant efficacy of a selective organic cation transporter blocker in a mouse model of depression. Mol Psychiatry. 2020 Jun;25(6):1245-1259. doi: 10.1038/s41380-019-0548-4. PMID: 31619760.

  91. Bacq A, Balasse L, Biala G, Guiard B, Gardier AM, Schinkel A, Louis F, Vialou V, Martres MP, Chevarin C, Hamon M, Giros B, Gautron S (2012): Organic cation transporter 2 controls brain norepinephrine and serotonin clearance and antidepressant response. Mol Psychiatry. 2012 Sep;17(9):926-39. doi: 10.1038/mp.2011.87. PMID: 21769100.

  92. Matochik, Liebenauer, King, Szymanski, Cohen, Zametkin (1994): Cerebral glucose metabolism in adults with attention defi cit hyperactivity disorder after chronic stimulant treatment. Am J Psychiatry 151: 658–664; Achtung, geringes N von 37; geringe Stichprobe mit n = 18; zitiert nach Edel, Vollmoeller (2006): Aufmerksamkeitsdefizit-/Hyperaktivitätsstörung bei Erwachsenen, Springer, Seite 10

  93. Stroemer, Kent, Hulsebosch (1998): Enhanced neocortical neural sprouting, synaptogenesis, and behavioral recovery with D-amphetamine therapy after neocortical infarction in rats; Stroke. 1998 Nov;29(11):2381-93; discussion 2393-5.

  94. Takahashi, Ohmiya, Honda, Ni (2018): The KCNH3 inhibitor ASP2905 shows potential in the treatment of attention deficit/hyperactivity disorder. PLoS One. 2018 Nov 21;13(11):e0207750. doi: 10.1371/journal.pone.0207750. eCollection 2018.

  95. Hutson, Heins, Folgering (2015): Effects of lisdexamfetamine alone and in combination with s-citalopram on acetylcholine and histamine efflux in the rat pre-frontal cortex and ventral hippocampus. J Neurochem. 2015 Aug;134(4):693-703. doi: 10.1111/jnc.13157.

  96. Madras, Miller, Fischman (2005): The dopamine transporter and attention-deficit/hyperactivity disorder. Biol Psychiatry. 2005 Jun 1;57(11):1397-409. doi: 10.1016/j.biopsych.2004.10.011. PMID: 15950014.

  97. Nagy, Häge, Coghill, Caballero, Adey, Anderson, Sikirica, Cardo (2015): Functional outcomes from a head-to-head, randomized, double-blind trial of lisdexamfetamine dimesylate and atomoxetine in children and adolescents with attention-deficit/hyperactivity disorder and an inadequate response to methylphenidate.Eur Child Adolesc Psychiatry. 2016 Feb;25(2):141-9. doi: 10.1007/s00787-015-0718-0.

  98. Madhoo, Keefe, Roth, Sambunaris, Wu, Trivedi, Anderson, Lasser (2014): Lisdexamfetamine dimesylate augmentation in adults with persistent executive dysfunction after partial or full remission of major depressive disorder; Neuropsychopharmacology. 2014 May;39(6):1388-98. doi: 10.1038/npp.2013.334. n=143 Erwachsene

  99. Coghill, Banaschewski, Nagy, Otero, Soutullo, Yan, Caballero, Zuddas (2017): Long-Term Safety and Efficacy of Lisdexamfetamine Dimesylate in Children and Adolescents with ADHD: A Phase IV, 2-Year, Open-Label Study in Europe.CNS Drugs. 2017 Jun 30. doi: 10.1007/s40263-017-0443-y; n = 314

  100. Arnold: Journal of Attention Disorders Vol. 3(4):200-211 (2000) Methylphenidate vs. amphetamine: Comparative review n =174

  101. Müller, Candrian, Kropotov (2011), ADHS – Neurodiagnostik in der Praxis, Seite 21

  102. Diamond: Attention-deficit disorder (attention-deficit/hyperactivity disorder without hyperactivity): A neurobiologically and behaviorally distinct disorder from attention-deficit (with hyperactivity), Development and Psychopathology 17 (2005), 807–825, Seite 811

  103. Liddle, Hollis, Batty, Groom, Totman, Liotti, Scerif, Liddle (2011): Task-related default mode network modulation and inhibitory control in ADHD: effects of motivation and methylphenidate. J Child Psychol Psychiatry. 2011 Jul;52(7):761-71. doi: 10.1111/j.1469-7610.2010.02333.x.

  104. ADHS-Chaoten;Thread: Amphetamin und Methylphenidat für Depressionen

  105. Adler, Goodman, Weisler, Hamdani, Roth (2009): Effect of lisdexamfetamine dimesylate on sleep in adults with attention-deficit/hyperactivity disorder. Behav Brain Funct. 2009 Aug 3;5:34. doi: 10.1186/1744-9081-5-34. PMID: 19650932; PMCID: PMC2732626.

  106. Giblin, Strobel (2011): Effect of lisdexamfetamine dimesylate on sleep in children with ADHD. J Atten Disord. 2011 Aug;15(6):491-8. doi: 10.1177/1087054710371195. PMID: 20574056.

  107. http://www.adhs-anderswelt.de/viewtopic.php?p=659654

  108. López FA, Leroux JR (2013): Long-acting stimulants for treatment of attention-deficit/hyperactivity disorder: a focus on extended-release formulations and the prodrug lisdexamfetamine dimesylate to address continuing clinical challenges. Atten Defic Hyperact Disord. 2013 Sep;5(3):249-65. doi: 10.1007/s12402-013-0106-x. PMID: 23564273; PMCID: PMC3751218. REVIEW

  109. Mattay, Goldberg, Fera, Hariri, Tessitore, Egan, Kolachana, Callicott, Weinberger (2003): Catechol O-methyltransferase val158-met genotype and individual variation in the brain response to amphetamine; doi: 10.1073/pnas.0931309100; PNAS May 13, 2003 vol. 100 no. 10 6186-6191

  110. Childress, Newcorn, Cutler (2019): Gender Effects in the Efficacy of Racemic Amphetamine Sulfate in Children with Attention-Deficit/Hyperactivity Disorder. Adv Ther. 2019 Apr 10. doi: 10.1007/s12325-019-00942-5.

  111. Arnold (2000): L.E. Methylphenidate vs amphetamine: Comparative review, J. of Att. Disorders, 2000, Vol 3, 200ff, n = 174

  112. Müller, Candrian, Kropotov (2011), ADHS – Neurodiagnostik in der Praxis, Springer, Seite 21

  113. Castello et al. 1992, zitiert nach Arnold: Journal of Attention Disorders Vol. 3(4):200-211 (2000) Methylphenidate vs. amphetamine: Comparative review

  114. Brams, Weisler, Findling, Gasior, Hamdani, Ferreira-Cornwell, Squires: Maintenance of Efficacy of Lisdexamfetamine Dimesylate in Adults With Attention-Deficit/Hyperactivity Disorder: Randomized Withdrawal Design, J Clin Psychiatry 2012;73(7):977-983; 10.4088/JCP.11m07430

  115. http://www.ads-hyperaktivitaet.de/FAQ/Infos/Medis/medis.html#1

  116. Markowitz, Patrick (2017): The Clinical Pharmacokinetics of Amphetamines Utilized in the Treatment of Attention-Deficit/Hyperactivity Disorder. J Child Adolesc Psychopharmacol. 2017 Oct;27(8):678-689. doi: 10.1089/cap.2017.0071. PMID: 28910145. REVIEW

  117. Dolder, Strajhar, Vizeli, Hammann, Odermatt, Liechti (2017): Pharmacokinetics and Pharmacodynamics of Lisdexamfetamine Compared with D-Amphetamine in Healthy Subjects. Front Pharmacol. 2017 Sep 7;8:617. doi: 10.3389/fphar.2017.00617. PMID: 28936175; PMCID: PMC5594082. n = 24) Die Studie benennt allerdings nicht das Gewicht der Probanden, sodass keine Aussage über die Dosierung / kg Körpergewicht möglich ist. Das Gewicht ist jedoch bei der Amphetamindosierung ein relevanter Faktor.((Markowitz, Patrick (2017): The Clinical Pharmacokinetics of Amphetamines Utilized in the Treatment of Attention-Deficit/Hyperactivity Disorder. J Child Adolesc Psychopharmacol. 2017 Oct;27(8):678-689. doi: 10.1089/cap.2017.0071. PMID: 28910145. REVIEW

  118. Amphetamines. LiverTox: Clinical and Research Information on Drug-Induced Liver Injury [Internet]. Bethesda (MD): National Institute of Diabetes and Digestive and Kidney Diseases; 2012-2016

  119. Ito, Onodera, Yamatodani, Watanabe, Sato (1997): The effect of methamphetamine on histamine release in the rat hypothalamus. Psychiatry Clin Neurosci. 1997 Apr;51(2):79-81. doi: 10.1111/j.1440-1819.1997.tb02911.x. PMID: 9141145.

  120. Ito, Onodera, Sakurai, Sato, Watanabe (1996): The effect of methamphetamine on histamine level and histidine decarboxylase activity in the rat brain. Brain Res. 1996 Sep 23;734(1-2):98-102. PMID: 8896814.

  121. Houghton, de Vries, Loss (2019): Psychostimulants/Atomoxetine and Serious Cardiovascular Events in Children with ADHD or Autism Spectrum Disorder. CNS Drugs. 2019 Nov 25. doi: 10.1007/s40263-019-00686-4. n = 2.566.995

  122. Forns, Dudukina, Hägg, Szentkúti, Gembert, Plana, Gilsenan, Horváth-Puhó, Ehrenstein, Reutfors, Rebordosa )2022): Risk of Major Cardiovascular and Cerebrovascular Events in Users of Lisdexamfetamine and Other Medications for Attention-Deficit/Hyperactivity Disorder in Denmark and Sweden: A Population-Based Cohort Study. Neurol Ther. 2022 Aug 26. doi: 10.1007/s40120-022-00396-y. PMID: 36028603. n = 273.000

  123. Harris E. Long-Term ADHD Medications and Cardiovascular Disease Risk. JAMA. 2023 Dec 26;330(24):2331. doi: 10.1001/jama.2023.24173. PMID: 38055293. n = 60.000

  124. Manso, Morcillo, Pereira, Maldonado (2020): Tricotilomanía de nueva aparición durante el tratamiento con fármacos estimulantes. A propósito de dos casos clínicos pediátricos [New-onset trichotillomania during treatment with stimulant drugs. About two pediatric clinical cases]. Arch Argent Pediatr. 2020 Feb;118(1):e61-e62. Spanish. doi: 10.5546/aap.2020.e61. PMID: 31984712.

  125. Mang (2018): 05. Monoamine 2: Amphetamin, Ritalin (ADHS), Cocain, Tricyclika, Videovorlesung. ca. bei Minute 38.

  126. Assem-Hilger, Kasper (2005): Psychopharmaka und sexuelle Dysfunktion. Journa lfür Neurologie, Neurochirurgie und Psychiatrie; 2005, 30-36

  127. Levine LA, Betcher HK, Ziegelmann MJ, Bajic P (2020): Amphetamine/Dextroamphetamine Salts for Delayed Orgasm and Anorgasmia in Men: A Pilot Study. Urology. 2020 Aug;142:141-145. doi: 10.1016/j.urology.2020.04.081. PMID: 32360625. n = 17

  128. Bartlik BD, Kaplan P, Kaplan HS (1995): Psychostimulants apparently reverse sexual dysfunction secondary to selective serotonin re-uptake inhibitors. J Sex Marital Ther. 1995 Winter;21(4):264-71. doi: 10.1080/00926239508414646. PMID: 8789508.

  129. Coskun M, Zoroglu S (2009): A report of two cases of sexual side effects with OROS methylphenidate. J Child Adolesc Psychopharmacol. 2009 Aug;19(4):477-9. doi: 10.1089/cap.2008.0161. PMID: 19702503.

  130. Cakin-Memik N, Yildiz O, Sişmanlar SG, Karakaya I, Ağaoğlu B (2010): Priapism associated with methylphenidate: a case report. Turk J Pediatr. 2010 Jul-Aug;52(4):430-4. PMID: 21043394.

  131. Ostdiek-Wille GP, Bavitz KC, Kohn TP, Deibert CM (2023): Attention-deficit hyperactivity disorder medication use is associated with testosterone hypofunction-results from a national claims database analysis. Int J Impot Res. 2023 Dec 21. doi: 10.1038/s41443-023-00805-2. PMID: 38129694. n = 34.448

  132. Spiller HA, Hays HL, Aleguas A Jr (2013): Overdose of drugs for attention-deficit hyperactivity disorder: clinical presentation, mechanisms of toxicity, and management. CNS Drugs. 2013 Jul;27(7):531-43. doi: 10.1007/s40263-013-0084-8. PMID: 23757186. REVIEW

  133. Fitzgerald KT, Bronstein AC (2013): Adderall® (amphetamine-dextroamphetamine) toxicity. Top Companion Anim Med. 2013 Feb;28(1):2-7. doi: 10.1053/j.tcam.2013.03.002. PMID: 23796480.

  134. Sharman J, Pennick M (2014): Lisdexamfetamine prodrug activation by peptidase-mediated hydrolysis in the cytosol of red blood cells. Neuropsychiatr Dis Treat. 2014 Nov 28;10:2275-80. doi: 10.2147/NDT.S70382. PMID: 25489246; PMCID: PMC4257105.

  135. Ward K, Citrome L (2018): Lisdexamfetamine: chemistry, pharmacodynamics, pharmacokinetics, and clinical efficacy, safety, and tolerability in the treatment of binge eating disorder. Expert Opin Drug Metab Toxicol. 2018 Feb;14(2):229-238. doi: 10.1080/17425255.2018.1420163. PMID: 29258368. REVIEW

  136. Konstantinidis: CYP-450-Interaktionen: Die Isoenzyme CYP1A2 und CYP2D6; Österreichische Gesellschaft für Neuropsychopharmakologie und Biologische Psychiatrie; Webseitenabruf 23.12.19

  137. Law R, Lewis D, Hain D, Daut R, DelBello MP, Frazier JA, Newcorn JH, Nurmi E, Cogan ES, Wagner S, Johnson H, Lanchbury J (2022): Characterisation of seven medications approved for attention-deficit/hyperactivity disorder using in vitro models of hepatic metabolism. Xenobiotica. 2022 Nov 1:1-32. doi: 10.1080/00498254.2022.2141151. PMID: 36317558.

  138. Bach MV, Coutts RT, Baker GB (1999): Involvement of CYP2D6 in the in vitro metabolism of amphetamine, two N-alkylamphetamines and their 4-methoxylated derivatives. Xenobiotica. 1999 Jul;29(7):719-32. doi: 10.1080/004982599238344. PMID: 10456690.

  139. Wu D, Otton SV, Inaba T, Kalow W, Sellers EM (1997): Interactions of amphetamine analogs with human liver CYP2D6. Biochem Pharmacol. 1997 Jun 1;53(11):1605-12. doi: 10.1016/s0006-2952(97)00014-2. PMID: 9264312.

  140. Phamarkogenetik.de: Cytochrom P450 2D6 (CYP2D6) [T88.7] Abruf 25.03.2022

  141. Wirkstoff aktuell (2015): Lisdexamfetamin

  142. Suarez EA, Bateman BT, Hernandez-Diaz S, Straub L, McDougle CJ, Wisner KL, Gray KJ, Pennell PB, Lester B, Zhu Y, Mogun H, Huybrechts KF (2024): Prescription Stimulant Use During Pregnancy and Risk of Neurodevelopmental Disorders in Children. JAMA Psychiatry. 2024 Jan 24:e235073. doi: 10.1001/jamapsychiatry.2023.5073. PMID: 38265792; PMCID: PMC10809143. n = 4,3 Mio

  143. Rose, Hathcock, White, Borowski, Rivera-Chiauzzi (2020): Amphetamine-Dextroamphetamine and Pregnancy: Neonatal Outcomes After Prenatal Prescription Mixed Amphetamine Exposure. J Atten Disord. 2020 Jan 13:1087054719896857. doi: 10.1177/1087054719896857.

  144. Bro, Kjaersgaard, Parner, Sørensen, Olsen, Bech, Pedersen, Christensen, Vestergaard (2015): Adverse pregnancy outcomes after exposure to methylphenidate or atomoxetine during pregnancy. Clin Epidemiol. 2015 Jan 29;7:139-47. doi: 10.2147/CLEP.S72906. eCollection 2015.

  145. Cohen, Hernández-Díaz, Bateman, Park, Desai, Gray, Patorno, Mogun, Huybrechts (2017): Placental Complications Associated With Psychostimulant Use in Pregnancy. Obstet Gynecol. 2017 Dec;130(6):1192-1201. doi: 10.1097/AOG.0000000000002362.

  146. Haervig, Mortensen, Hansen, Strandberg-Larsen (2014): Use of ADHD medication during pregnancy from 1999 to 2010: a Danish register-based study. Pharmacoepidemiol Drug Saf. 2014 May;23(5):526-33. doi: 10.1002/pds.3600. n = 480 unter 1.054.494 Geburten

  147. Pottegård, Hallas, Andersen, Løkkegaard, Dideriksen, Aagaard, Damkier (2014): First-trimester exposure to methylphenidate: a population-based cohort study. J Clin Psychiatry. 2014 Jan;75(1):e88-93. doi: 10.4088/JCP.13m08708.

  148. Szpunar MJ, Freeman MP, Kobylski LA, Rossa ET, Gaccione P, Chitayat D, Viguera AC, Cohen LS (2023): Risk of Major Malformations in Infants After First-Trimester Exposure to Stimulants: Results From the Massachusetts General Hospital National Pregnancy Registry for Psychiatric Medications. J Clin Psychopharmacol. 2023 Jul-Aug 01;43(4):326-332. doi: 10.1097/JCP.0000000000001702. PMID: 37235505.

  149. Russell DJ, Wyrwoll CS, Preen DB, Kelty E (2024): Investigating maternal and neonatal health outcomes associated with continuing or ceasing dexamphetamine treatment for women with attention-deficit hyperactivity disorder during pregnancy: a retrospective cohort study. Arch Womens Ment Health. 2024 Mar 1. doi: 10.1007/s00737-024-01450-4. PMID: 38424254.

  150. Moran, Ongur, Hsu, Castro, Perlis, Schneeweiss (2019): Psychosis with Methylphenidate or Amphetamine in Patients with ADHD. N Engl J Med. 2019 Mar 21;380(12):1128-1138. doi: 10.1056/NEJMoa1813751. n = 221.846

  151. Jongsma, Gayer-Anderson, Lasalvia, Quattrone, Mulè, Szöke, Selten, Turner, Arango, Tarricone, Berardi, Tortelli, Llorca, de Haan, Bobes, Bernardo, Sanjuán, Santos, Arrojo, Del-Ben, Menezes, Velthorst, Murray, Rutten, Jones, van Os, Morgan, Kirkbride; for the European Network of National Schizophrenia Networks Studying Gene-Environment Interactions Work Package 2 (EU-GEI WP2) Group(2018): Treated Incidence of Psychotic Disorders in the Multinational EU-GEI Study. JAMA Psychiatry. 2018;75(1):36–46. doi:10.1001/jamapsychiatry.2017.3554; n = 12,9 Millionen Personenjahre

  152. Gelbe Liste: Dexamfetamin. Abgerufen 12.02.23

  153. Cosgrove K, Johnston CS (2017): Examining the Impact of Adherence to a Vegan Diet on Acid-Base Balance in Healthy Adults. Plant Foods Hum Nutr. 2017 Sep;72(3):308-313. doi: 10.1007/s11130-017-0620-7. PMID: 28677099.

  154. Osuna-Padilla IA, Leal-Escobar G, Garza-García CA, Rodríguez-Castellanos FE (2019): Dietary Acid Load: mechanisms and evidence of its health repercussions. Nefrologia (Engl Ed). 2019 Jul-Aug;39(4):343-354. English, Spanish. doi: 10.1016/j.nefro.2018.10.005. PMID: 30737117. REVIEW

  155. Passey C (2017): Reducing the Dietary Acid Load: How a More Alkaline Diet Benefits Patients With Chronic Kidney Disease. J Ren Nutr. 2017 May;27(3):151-160. doi: 10.1053/j.jrn.2016.11.006. PMID: 28117137 REVIEW

  156. Terrillion CE, Dao DT, Cachope R, Lobo MK, Puche AC, Cheer JF, Gould TD (2017): Reduced levels of Cacna1c attenuate mesolimbic dopamine system function. Genes Brain Behav. 2017 Jun;16(5):495-505. doi: 10.1111/gbb.12371. PMID: 28186690; PMCID: PMC5457318.

  157. Shire (2018): Fachinformation Elvanse

  158. Schoretsanitis, de Leon, Eap, Kane, Paulzen (2019): Clinically Significant Drug-Drug Interactions with Agents for Attention-Deficit/Hyperactivity Disorder. CNS Drugs. 2019 Dec;33(12):1201-1222. doi: 10.1007/s40263-019-00683-7.

  159. Pharmazeutische Zeitung: Lisdexamfetamin; abgerufen 10.02.21

  160. Castells, Ramon, Cunill, Olivé, Serrano (2020): Relationship Between Treatment Duration and Efficacy of Pharmacological Treatment for ADHD: A Meta-Analysis and Meta-Regression of 87 Randomized Controlled Clinical Trials. J Atten Disord. 2020 Feb 20:1087054720903372. doi: 10.1177/1087054720903372. PMID: 32075485.