Therapeutic Doses of Methylphenidate

| categories: mph, dose

Contents

1 Therapeutic Doses of Methylphenidate

Though methylphenidate (MPH) is very effective for the treatment of attention-deficit/hyperactivity disorder (ADHD) the doses required to achieve clinical responses vary significantly across individuals (0.1 mg/kg to 1 mg/kg) [18]. Similarly the responses to MPH when given in a laboratory setting for research purposes are also quite variable. Other than differences in metabolism of MPH, part of the variability in the behavioral responses to MPH are due to differences in dopamine (DA) cell activity and in the levels of DA D2 receptors between subjects [2021].

For a typical 25 kg child the maximum serum concentration is expected to be about [17]: 5 ng/ml for a low dose (5 mg; 0.2 mg/kg), 10 ng/ml for a medium dose (10 mg; 0.4 mg/kg), and 20 ng/ml for a high dose (20 mg; 0.8 mg/kg). Regardless of dose, time to maximum (Tmax) is expected to be about 1.5 h to 2 h [17], and half-life (T12) is expected to be 2 h to 3 h [17]. The maximum behavioral effects (reduction in overactivity, impulsivity, and inattention) occur about 1 h to 2 h after oral doses [17], behavioral effects dissipate significantly by 2 h after max behavioral effect (3 h to 4 h after each dose) [17]. Oral MPH at doses used therapeutically induce greater than 50 % dopamine transporter (DAT) blockade with an estimated median effective dose (ED50) dose of 0.25 mg/kg

1.1 Experiments in mice

Oral dose of 0.75 mg/kg MPH can produce plasma levels of d-MPH in the 6 ng/ml to 10 ng/ml range in mice [2]. 2 ng/ml to 9 ng/ml represents the lower limit of the clinical range, whereas a 3 mg/kg dose was predicted to produce peak plasma concentrations between 30 ng/ml to 60 ng/ml, corresponding to upper limits of the clinical range [7]. Brain concentrations were 4- and 5-fold higher than the plasma concentration going up to 6–8 fold higher than plasma at 30 min with a low dose of MPH [2]. Higher doses had less brain loading by proportion, with a brain/plasma ratio below 1 at 2 mg/kg to 5 mg/kg dose for early time periods, and increasing once plasma levels drop [2].

2 Selective metabolism of MPH isomers

Therapeutic effects of oral dl-MPH appear to be limited to the d-MPH isomer [14916], as consistent with a pure d-MPH formulation (dexmethylphenidate; Focalin®;) becoming available in 2002. Any potential benefit of removing the l-MPH isomer from an oral dl-MPH formulation is not obvious [15], due to the very extensive oral pre-systemic metabolism of l-MPH: typically only low pg/mL concentrations of the l-isomer reach the systemic circulation [4101222]. This trace l-MPH plasma concentration represents only about 1 % of the d-MPH plasma concentration [13]. In effect, oral dl-MPH in humans is subject to in vivo biocatalytic resolution by carboxylesterase 1 [23], analogous to the enzymatic approach that has been used industrially to eliminate l-MPH from dl-MPH and yield enantiopure d-MPH [13].

3 Variability of Response to MPH treatment

Imaging studies have documented large variability in the magnitude of the changes in extracellular DA induced by MPH. After oral administration of 60 mg MPH the range of changes in extracellular DA was 3 % to 48 % [20]. While this variability is in part related to age, a large percentage of the variability still remains unaccounted by age. The levels of DAT blockade in these subjects did not predict the changes in extracellular DA as assessed by decreases in DA D2 receptor availability. Volkow et al. [19] suggest this lack of a correlation as an indication that the DA increases were due not just to DAT blockade by MPH but to the individual variability in the amount of DA released by the DA cells. This is consistent with the findings that homovanillic acid (HVA) levels in cerebrospinal fluid, which serve as a marker of DA turnover in the central nervous system, predicted response to MPH in children with ADHD; the higher the levels the better the responses [3].

3.1 Response Variability based on Impulsivity

MPH treatment reverses attentional impairments induced with NMDA glutamate antagonist, muscarinic acetylcholine receptor (mAChR) antagonist, nicotinic acetylcholine receptor (nAChR) antagonist [14], and also eliminated impairments of attention and memory in the ‘spontaneous hypertensive rat’ model [6]. This is consistent with the suggestion that psychostimulants only improve impulse control under conditions in which baseline levels of impulsivity are high, as is seen in patients with ADHD [11]. Dose-effect functions need to be carefully assessed in testing drug effects on attentional function in animal models because all drugs which improve attention have an inverted U-shaped dose-effect function in which either lower or higher than optimal doses are less effective and even impairing [8]. The combination of this characteristic non-monotonic dose-effect function with the fact that there is often considerable individual variability in drug sensitivity can complicate detection of beneficial responses to attention-improving drugs.

3.2 Non-responders

As yet no explanation has been found for the fact that about one third of children with ADHD do not clinically respond to MPH [5]. ‘Non-responders’ to MPH treatment had higher levels of both d-MPH and l-MPH enantiomers in plasma at all time points taken than ‘responders’, leading them to think that d-MPH may be getting into the brain more in ‘responders’ than in ‘non-responders’, leaving more MPH in blood plasma in ‘non-responders’ [5]. MPH’s effects are dependent on the rate of DA release. This, in turn, provides an explanation for the variability in the responses to MPH: subjects with high DA tone will be more sensitive to MPH’s therapeutic effects than those with a low DA tone [19]. It also provides an explanation for non-responsiveness, which could reflect very low endogenous DA activity.

Acronyms

ADHD
attention-deficit/hyperactivity disorder
DAT
dopamine transporter
DA
dopamine
ED50
median effective dose
HVA
homovanillic acid
mAChR
muscarinic acetylcholine receptor
MPH
methylphenidate
nAChR
nicotinic acetylcholine receptor

References

[1]    T. Aoyama, H. Kotaki, Y. Sawada, and T. Iga. Stereospecific distribution of methylphenidate enantiomers in rat brain: specific binding to dopamine reuptake sites. Pharm Res, 11(3):407–11, Mar 1994.

[2]    A. Balcioglu, J.-Q. Ren, D. McCarthy, T. J. Spencer, J. Biederman, and P. G. Bhide. Plasma and brain concentrations of oral therapeutic doses of methylphenidate and their impact on brain monoamine content in mice. Neuropharmacology, 57(7-8):687–93, Dec 2009. doi: 10.1016/j.neuropharm. 2009.07.025.

[3]    F. X. Castellanos, J. Elia, M. J. Kruesi, W. L. Marsh, C. S. Gulotta, W. Z. Potter, G. F. Ritchie, S. D. Hamburger, and J. L. Rapoport. Cerebrospinal fluid homovanillic acid predicts behavioral response to stimulants in 45 boys with attention deficit/hyperactivity disorder. Neuropsychopharmacology, 14(2):125–37, Feb 1996. doi: 10.1016/0893-_ 133X(95)00077-_Q.

[4]    Y.-S. Ding, S. J. Gatley, P. K. Thanos, C. Shea, V. Garza, Y. Xu, P. Carter, P. King, D. Warner, N. B. Taintor, D. J. Park, B. Pyatt, J. S. Fowler, and N. D. Volkow. Brain kinetics of methylphenidate (ritalin) enantiomers after oral administration. Synapse, 53(3):168–75, Sep 2004. doi: 10.1002/syn.20046.

[5]    L. M. Jonkman, M. N. Verbaten, D. de Boer, R. A. Maes, J. K. Buitelaar, C. Kemner, H. van Engeland, and H. S. Koelega. Differences in plasma concentrations of the d- and l-threo methylphenidate enantiomers in responding and non-responding children with attention-deficit hyperactivity disorder. Psychiatry Res, 78(1-2):115–8, Mar 1998.

[6]    K. M. Kantak, T. Singh, K. A. Kerstetter, K. A. Dembro, M. M. Mutebi, R. C. Harvey, C. F. Deschepper, and L. P. Dwoskin. Advancing the spontaneous hypertensive rat model of attention deficit/hyperactivity disorder. Behav Neurosci, 122(2):340–57, Apr 2008. doi: 10.1037/0735-_ 7044.122.2.340.

[7]    R. Kuczenski and D. S. Segal. Exposure of adolescent rats to oral methylphenidate: preferential effects on extracellular norepinephrine and absence of sensitization and cross-sensitization to methamphetamine. J Neurosci, 22(16):7264–7271, Aug 2002. doi: 20026690. URL http://dx.doi.org/20026690.

[8]    E. D. Levin, P. J. Bushnell, and A. H. Rezvani. Attention-modulating effects of cognitive enhancers. Pharmacol Biochem Behav, 99(2):146–54, Aug 2011. doi: 10.1016/j.pbb.2011.02.008.

[9]    J. S. Markowitz and K. S. Patrick. Differential pharmacokinetics and pharmacodynamics of methylphenidate enantiomers: does chirality matter? J Clin Psychopharmacol, 28(3 Suppl 2):S54–61, Jun 2008. doi: 10.1097/JCP.0b013e3181733560.

[10]    J. S. Markowitz, A. B. Straughn, and K. S. Patrick. Advances in the pharmacotherapy of attention-deficit-hyperactivity disorder: focus on methylphenidate formulations. Pharmacotherapy, 23(10):1281–99, Oct 2003.

[11]    T. A. Paine, H. C. Tomasiewicz, K. Zhang, and W. A. Carlezon, Jr. Sensitivity of the five-choice serial reaction time task to the effects of various psychotropic drugs in sprague-dawley rats. Biol Psychiatry, 62(6):687–93, Sep 2007. doi: 10.1016/j.biopsych.2006.11.017.

[12]    K. S. Patrick, M. A. González, A. B. Straughn, and J. S. Markowitz. New methylphenidate formulations for the treatment of attention-deficit/hyperactivity disorder. Expert Opin Drug Deliv, 2(1):121–43, Jan 2005. doi: 10.1517/17425247.2.1.121.

[13]    K. S. Patrick, A. B. Straughn, J. S. Perkins, and M. A. González. Evolution of stimulants to treat adhd: transdermal methylphenidate. Hum Psychopharmacol, 24(1):1–17, Jan 2009. doi: 10.1002/hup.992.

[14]    A. H. Rezvani, E. Kholdebarin, M. C. Cauley, E. Dawson, and E. D. Levin. Attenuation of pharmacologically-induced attentional impairment by methylphenidate in rats. Pharmacol Biochem Behav, 92(1):141–6, Mar 2009. doi: 10.1016/j.pbb.2008.11.005.

[15]    N. R. Srinivas, J. W. Hubbard, D. Quinn, E. D. Korchinski, and K. K. Midha. Extensive and enantioselective presystemic metabolism of dl-threo-methylphenidate in humans. Prog Neuropsychopharmacol Biol Psychiatry, 15(2):213–20, 1991.

[16]    N. R. Srinivas, J. W. Hubbard, D. Quinn, and K. K. Midha. Enantioselective pharmacokinetics and pharmacodynamics of dl-threo-methylphenidate in children with attention deficit hyperactivity disorder. Clin Pharmacol Ther, 52(5):561–8, Nov 1992.

[17]    J. M. Swanson and N. D. Volkow. Serum and brain concentrations of methylphenidate: implications for use and abuse. Neurosci Biobehav Rev, 27(7):615–621, Nov 2003.

[18]     J. M. Swanson, D. Cantwell, M. Lerner, K. McBurnett, and G. Hanna. Effects of stimulant medication on learning in children with adhd. J Learn Disabil, 24(4):219–30, 255, Apr 1991.

[19]    N. D. Volkow, J. S. Fowler, G.-J. Wang, Y. S. Ding, and S. J. Gatley. Role of dopamine in the therapeutic and reinforcing effects of methylphenidate in humans: results from imaging studies. European Neuropsychopharmacology, 12:557–566, 2002.

[20]    N. D. Volkow, G.-J. Wang, J. S. Fowler, J. Logan, D. Franceschi, L. Maynard, Y.-S. Ding, S. J. Gatley, A. Gifford, W. Zhu, and J. M. Swanson. Relationship between blockade of dopamine transporters by oral methylphenidate and the increases in extracellular dopamine: therapeutic implications. Synapse, 43(3):181–7, Mar 2002. doi: 10.1002/syn.10038.

[21]    N. D. Volkow, G.-J. Wang, J. S. Fowler, P. P. K. Thanos, J. Logan, S. J. Gatley, A. Gifford, Y.-S. Ding, C. Wong, N. Pappas, and P. Thanos. Brain da d2 receptors predict reinforcing effects of stimulants in humans: replication study. Synapse, 46(2):79–82, Nov 2002. doi: 10.1002/syn.10137.

[22]    W. Wargin, K. Patrick, C. Kilts, C. T. Gualtieri, K. Ellington, R. A. Mueller, G. Kraemer, and G. R. Breese. Pharmacokinetics of methylphenidate in man, rat and monkey. J Pharmacol Exp Ther, 226(2): 382–6, Aug 1983.

[23]    J. Zhu and M. E. A. Reith. Role of the dopamine transporter in the action of psychostimulants, nicotine, and other drugs of abuse. CNS Neurol Disord Drug Targets, 7(5):393–409, Nov 2008.