The American Psychiatric Association (APA) has updated its Privacy Policy and Terms of Use, including with new information specifically addressed to individuals in the European Economic Area. As described in the Privacy Policy and Terms of Use, this website utilizes cookies, including for the purpose of offering an optimal online experience and services tailored to your preferences.

Please read the entire Privacy Policy and Terms of Use. By closing this message, browsing this website, continuing the navigation, or otherwise continuing to use the APA's websites, you confirm that you understand and accept the terms of the Privacy Policy and Terms of Use, including the utilization of cookies.

×
Clinical SynthesisFull Access

Novel Formulations of ADHD Medications: Stimulant Selection and Management

Abstract

Attention-deficit hyperactivity disorder (ADHD) is the most commonly diagnosed psychiatric disorder in children and adolescents in the United States. In 2016, approximately 3.8 million U.S. children ages 2 to 17 years with ADHD were being treated with medication. There are approximately 30 different amphetamine (AMPH) and methylphenidate (MPH) formulations on the market. These include immediate-release and extended-release compounds. The extended-release formulations contain various ratios of immediate-release and extended-release components, which determine the pharmacokinetic (PK) profile. For stimulants, the PK and pharmacodynamic (PD) profiles are tightly linked, and the immediate-release and extended-release percentages influence onset and duration of drug effects. Choosing the right stimulant medication for a patient depends on an understanding of the PK/PD profile, the time of day that symptoms are most impairing, the need for morning and evening symptom control and individual patient preferences.

Attention-deficit hyperactivity disorder (ADHD) is the most commonly diagnosed psychiatric disorder in children, and symptoms often continue into adulthood (13). Among U.S. children ages 2 to 17, approximately 6.1 million (9.4%) were estimated to have been diagnosed as having ADHD on the basis of the 2016 National Survey of Children’s Health (4). Of these, 62.0% (approximately 3.8 million) with current ADHD symptoms were taking medication. The estimated prevalence of ADHD among U.S. adults is 4.4% (3). The prevalence of adult ADHD medication use in the total U.S. population was 1.48% in 2010—the highest in the world (5).

Several professional societies in the United States, including the American Academy of Pediatrics (AAP), the American Academy of Child and Adolescent Psychiatry (AACAP), and the Society for Developmental and Behavioral Pediatrics (SDBP), have released guidelines for the treatment of ADHD in children and adolescents. Although organizations in other countries, such as the National Institute for Health Care and Excellence, have also published ADHD treatment guidelines, this article focuses on recommendations for treatment and medications available in the United States (6).

Key action statement 5b in the recent AAP “Clinical Practice Guideline for the Diagnosis, Evaluation, and Treatment of ADHD in Children and Adolescents” states that elementary school– and middle school–age children should be treated with a U.S. Food and Drug Administration (FDA)–approved medication, along with parent training in behavior management (7). Key action statement 5c states that adolescents should be prescribed FDA-approved medications for ADHD, with the adolescent’s assent, and encourages the primary care provider to also prescribe evidence-based training or behavioral interventions along with appropriate educational interventions. For preschool-age children (ages 4 to <6), key action statement 5a recommends evidence-based parent training in behavior management. If parent training in behavior management is not available or is available and not effective, treatment with methylphenidate is recommended.

The SDBP issued a practice guideline for the assessment and treatment of complex ADHD in children and adolescents in February 2020 (8). It recommends evidence-based treatments for ADHD and coexisting conditions.

Although the AACAP “Practice Parameter for the Assessment and Treatment of Children and Adolescents With Attention-Deficit/Hyperactivity Disorder ” was published more than a decade ago, it is still relevant (9). It recommends initial pharmacological treatment with an FDA-approved medication.

FDA-approved medications include stimulants and nonstimulants. Stimulants include amphetamine (AMPH) and methylphenidate (MPH). Nonstimulants include atomoxetine, guanfacine extended release, and clonidine extended release (1014).

Drug Review

Stimulants are by far the most widely prescribed medications for the treatment of ADHD in the United States (15). Thus this article focuses on the use of stimulants to treat ADHD, specifically how to target specific patient needs. Although osmotic-release oral system (OROS) MPH (Concerta), mixed amphetamine salts (Adderall), mixed amphetamine salts extended release (Adderall XR), and lisdexamfetamine (Vyvanse) have the largest market share in the United States, stimulant formulations prescribed less frequently should also be considered when treating individual patients (15).

Approximately 30 stimulants are FDA-approved for the treatment of ADHD, and 12 of these have been approved since 2010. New stimulant formulations approved since 2010 include MPH extended-release oral suspension (Quillivant XR), MPH extended-release chewable tablets (QuilliChew ER), racemic AMPH (Evekeo), racemic AMPH orally disintegrating tablets (Evekeo ODT), multilayer-release MPH (Aptensio XR), AMPH extended-release oral suspension 2.5 mg/mL (Dyanavel XR), MPH extended-release orally disintegrating tablet (Cotempla XR-ODT), AMPH extended-release orally disintegrating tablets (Adzenys XR-ODT), AMPH extended-release oral suspension 1.25 mg/mL (Adzenys ER), mixed amphetamine salts 16 hour (Mydayis), MPH delayed-release and extended-release (Jornay PM), and MPH multilayer-release 16 hour (Adhansia XR) (1626). AMPH immediate release (Zenzedi) is not a new formulation but includes multiple dextroamphetamine doses not previously available (27). Medications approved since 2000 are listed in Tables 1 and 2.

TABLE 1. Extended-release methylphenidate formulations approved since 2000 for the treatment of attention-deficit hyperactivity disorder

FormulationDose formOnset of effectaMaximum duration of effectaInitial doseDosing
Methylphenidate multilayer release, extended release (Adhansia XR)25-, 35-, 45-, 55-, 70-, 85-mg capsules1.0 hour16 hours25 mg25–85 mg; increase 10–15 mg every 5 days
Methylphenidate multilayer release (Aptensio XR)10-, 15-, 20-, 30-, 40-, 50-, 60-mg capsules1.0 hour12 hours10 mg10–60 mg; increase by 10 mg every 7 days
Osmotic-release oral system methylphenidate (Concerta)18-, 27-, 36-, 54-mg tablets1.0 hour12.5 hours18 mg18–54 mg for children; 18–72 mg for adolescents and adults (not to exceed 2 mg/kg/day)
Methylphenidate extended release, orally disintegrating tablet (Cotempla XR-ODT)8.6-, 17.3-, 25.9-mg tablets1.0 hour12 hours17.3 mg17.3–51.8 mg; increase 8.6–17.3 mg/day every 7 days
Methylphenidate transdermal system (Daytrana)10-, 15-, 20-, 30-mg patches2.0 hours12 hours (when worn for 9 hours)10 mg10–30 mg
Dexmethylphenidate extended release (Focalin XR)5-, 10-, 15-, 20-, 25-, 30-, 35-, 40-mg capsules0.5 hour12 hours5 mg for children; 10 mg for adults5–30 mg for children; 10–40 mg for adults
Methylphenidate delayed release/extended release (Jornay PM)20-, 40-, 60-, 80-, 100-mg capsules10 hours after dosing23 hours after dosing20 mg20–100 mg; increase 20 mg weekly
Methylphenidate controlled delivery (Metadate CD)10-, 20-, 30-, 40-, 50-, 60-mg capsules1.5 hours7.5–12 hours (depending on dose)20 mg10–60 mg
Methylphenidate extended release, oral suspension (Quillivant XR)Extended-release suspension 25 mg/5 mL0.75 hour12 hours20 mg20–60 mg; increase 10–20 mg every 7 days
Methylphenidate extended release, chewable tablet (QuilliChew ER)20 mg-, 30 mg-, 40-mg tablets2.0 hours8 hours20 mg20–60 mg; increase 10–20 mg every 7 days
Long-acting methylphenidate (Ritalin)10-, 20-, 30-, 40-mg capsules30 minutes12 hours (depending on dose)10–20 mg10–60 mg

aData for onset and duration of effect may not be in the Food and Drug Administration–approved label.

TABLE 1. Extended-release methylphenidate formulations approved since 2000 for the treatment of attention-deficit hyperactivity disorder

Enlarge table

TABLE 2. Intermediate- and extended-release amphetamine formulations approved since 2000 for the treatment of attention-deficit hyperactivity disorder

FormulationDose formOnset of effectaMaximum duration of effectaInitial doseDosing
Mixed amphetamine salts, extended release (Adderall XR)5-, 10-, 15-, 20-, 25-, 30-mg capsules1.5 hours12 hours (depending on dose)10 mg for ages 6–17; 20 mg for adults5–30 mg for ages 6–12; 5–20 mg for ages 13–17; 20 mg for adults
Extended-release amphetamine, orally disintegrating tablets (Adzenys XR ODT)3.1-, 6.3-, 9.4-, 12.5-, 15.7-, 18.8-mg tabletsBioequivalent to Adderall XRBioequivalent to Adderall XR6.3 mg for ages 6–17; 12.5 mg for adults6.3–18.8 mg for ages 6–12; 6.3–12.5 mg for ages 13–17; 12.5 mg for adults
Extended-release amphetamine, suspension (Adzenys ER)1.25-mg/mL extended-release suspensionBioequivalent to Adderall XRBioequivalent to Adderall XR6.3 mg for ages 6–17; 12.5 mg for adults6.3–18.8 mg for ages 6–12; 6.3–12.5 mg for ages 13–17; 12.5 mg for adults
Amphetamine, extended release, oral suspension (Dyanavel XR)2.5-mg/mL extended-release suspension30 minutes13 hours2.5–5 mg for ages 6 and older2.5–20 mg for ages 6 and older; increase 2.5–5 mg every 4–7 days
Racemic amphetamine (Evekeo)5-, 10-mg tablets45 minutes10 hours2.5 mg for ages 3 and older; 5 mg for ages 6 and older5–40 mg divided daily or twice daily for ages 6 and older
Racemic amphetamine, orally disintegrating tablets (Evekeo ODT)5-, 10-, 15-, 20-mg tabletsBioequivalent to EvekeoBioequivalent to Evekeo5 mg daily or twice dailyIncrease 5 mg weekly; maximum dose not listed in label
Mixed amphetamine salts, extra-long extended release (Mydayis)12.5-, 25-, 37.5-, 50-mg capsules2.0 hours16 hours12.5 mg for ages 13 and older12.5–25 mg for ages 13–17; 12.5–50 mg for adults
Lisdexamfetamine (Vyvanse)10-, 20-, 30-, 40-, 50-, 60-, 70-mg capsules; 10-, 20-, 30-, 40-, 50-, 60-mg chewable tablets1.5 hours14 hours in adults; 13 hours in children30 mg30–70 mg; increase 10–20 mg weekly

aData for onset and duration of effect may not be in the Food and Drug Administration–approved label.

TABLE 2. Intermediate- and extended-release amphetamine formulations approved since 2000 for the treatment of attention-deficit hyperactivity disorder

Enlarge table

Stimulant formulations not mentioned above include racemic MPH controlled delivery (Metadate CD), MPH long acting (Ritalin LA), Methylin, and Methylin ER (2830). Methylin is an oral suspension of MPH immediate-release formulation, and Methylin ER is bioequivalent to Ritalin SR. Ritalin SR has been shown to have efficacy from approximately 1 hour to 9 hours after dosing (31). MPH transdermal system (Daytrana), a patch placed on the hip that delivers MPH through the skin, is also marketed (32). Dexmethylphenidate, the d-threo enantiomer of racemic MPH, is marketed as Focalin, dexmethylphenidate extended release (Focalin XR), and multiple generics (33, 34).

Stimulants exhibit a strong concentration-response relationship for both efficacy and safety (3537). For this reason, different formulations with different pharmacokinetic profiles developed from the same active component (AMPH or MPH) may have differing onset and duration of effect.

How to Choose a Stimulant Formulation

With the vast number of medications available to treat ADHD, selecting an initial pharmacological treatment may seem complex. However, the array of available medications allows the clinician to tailor treatment to individual patient needs. When choosing an initial therapy for ADHD, one should first consider that stimulants are in general more effective than nonstimulants (38). Other factors, such as the time of day when a patient is most symptomatic, the ability to swallow a tablet or capsule, concern about abuse or diversion either with the patient or a household member, and patient or parent preference, should also be considered. Daytime functioning or evening commitments should also influence the formulation choice. In general, when choosing a stimulant, an extended-release formulation should be chosen in lieu of an immediate-release formulation to improve adherence and decrease the risk of misuse (39, 40).

A recent meta-analysis of efficacy and tolerability of ADHD medications in children, adolescents, and adults favored the use of MPH in children and adolescents and AMPH in adults as initial pharmacological treatment for ADHD (41). FDA-approved drugs evaluated also included guanfacine extended release and atomoxetine.

Is the Patient Able to Swallow Tablets or Capsules?

Although the extended-release stimulant capsule formulations can be opened and sprinkled on applesauce for patients who cannot or prefer not to swallow a capsule whole, this maneuver is more difficult than it sounds. Capsules must be opened carefully to avoid spilling the contents, and it is important that the patient not chew the contents to avoid premature release of the drug (dose dumping). For patients who cannot or will not swallow an intact tablet or capsule, it may be more practical to choose an extended-release suspension in a chewable or orally disintegrating tablet formulation. It is important to consider patient and parent preference, including taste, when choosing a preparation. For example, teens may prefer not to take a suspension. Suspensions may also be less transportable than tablets or capsules, and MPH extended-release oral suspension is dispensed in a glass bottle that can break if dropped.

Oral suspensions can be easily titrated with just one prescription. The directions contained in the package inserts allow quick titration to optimal dose: MPH extended-release oral suspension (10–20 mg weekly), AMPH extended-release oral suspension 1.25 mg/mL (3.1–6.3 mg weekly), and AMPH extended-release oral suspension 2.5 mg/mL (2.5–10 mg every 4 to 7 days), so that patients can escalate to an effective dose within 1 month of starting medication. In contrast, for patients who have difficulty tolerating stimulant increases with tablets or capsules, the medication can be titrated very slowly with an oral suspension. Dosage can easily be adjusted by less than a mL to allow time for attenuation of adverse effects.

How to Achieve Optimal Response

Dose optimization of medication is important when treating ADHD. In double-blind, placebo-controlled clinical studies using one stimulant, 25%−35% of subjects were considered nonresponders (9). However, in crossover studies comparing AMPH with MPH, 68%−97% of subjects responded to at least one class of stimulants and 12%−71% responded to both (42).

How, then, does a clinician maximize the opportunity for response? A standardized scale should be used to obtain baseline and follow-up ADHD symptom ratings. For children, adolescents, and adults, several scales are available. A clinician should choose one for children and adolescents and another for adults to use regularly. Most laboratory classroom studies used an open-label, dose-optimization design prior to the double-blind, placebo-controlled classroom period. For several studies, a 30% improvement on an ADHD rating scale and a Clinical Global Impression improvement score of 1 (very much improved) or 2 (much improved) were considered minimal criteria for optimal response. Many protocols allowed further dose increases if tolerability was acceptable and subjects could benefit from an increased dose. If subjects had difficulty tolerating the higher dose, a dose reduction was allowed. Additionally, if subjects could not achieve the above criteria, they were discontinued from the trial. These criteria should be considered when treating patients in clinical practice. If a patient is unable to reach or tolerate an optimal dose in practice, the drug should be discontinued and another formulation prescribed.

Target Specific Times of Day

Early-morning efficacy.

The time of day when symptoms are most impairing can be targeted with stimulant formulations. If a child or adult has severe problems in the morning that cause difficulty preparing for school or work, a medication that is effective in the morning can be chosen. For example, delayed-release/extended-release MPH is formulated to have delayed onset of effect for approximately 10 hours after ingestion and is designed for patients to take in the evening and have onset of effect upon awakening the next morning. Data from two studies enrolling subjects ages 6 to 12 years who had morning impairment showed significant improvement in completing the morning routine. A laboratory classroom study demonstrated that the drug was effective, compared with placebo, during the school day and into the evening. An important consideration with delayed-release/extended-release MPH is dosing. The relative bioavailability of delayed-release/extended-release MPH is 73.9%, compared with immediate-release MPH (43). At the end of a 6-week, open-label, dose-optimization period in one study, the mean daily optimized dose was 66.2 mg (SD=19.56) (44). Although the FDA-recommended starting dose is 20 mg, most patients will not achieve optimal symptom control until higher doses are prescribed. For example, a patient taking 72 mg of OROS-MPH may need to take 100 mg of delayed-release/extended-release MPH.

For patients with early-morning impairment who prefer not to take medication in the evening, two extended-release formulations have demonstrated onset of efficacy at 30 minutes after dosing. These include dexmethylphenidate extended-release and AMPH extended-release oral suspension 2.5 mg/mL (45, 46). Additionally, MPH extended-release oral suspension has onset of effect at 45 minutes after dosing (47). Onset of effect is influenced by the percentage of immediate-release compound in different formulations. For example, OROS-MPH is composed of 22% immediate-release MPH, whereas MPH controlled delivery contains 30% immediate-release MPH and MPH long acting and dexmethylphenidate extended release have a 50% immediate-release component. Although MPH extended-release oral suspension contains approximately 20% MPH immediate release and 80% MPH extended release, there are thousands of MPH particles in each dose. The extended-release particles are covered by a proprietary coating of various thicknesses to achieve early onset and extended duration of effect (47).

OROS-MPH has 22% MPH immediate-release coating on the tablet, and the label states that OROS-MPH did not have significant change from placebo in a laboratory classroom trial until 2 hours after dosing (13). A head-to-head trial between OROS-MPH and MPH controlled delivery demonstrated that OROS-MPH has an earlier onset of effect, with an effect size of 0.52 at 1.5 hours postdose (48). A later trial, measuring time points from 1 hour to 12.5 hours after dosing, demonstrated onset of efficacy at 1 hour and duration of effect to 12.5 hours, compared with placebo (49).

Another option is to apply the MPH transdermal system to the hip before the patient awakens; however, this approach requires a caregiver to be up early to apply the patch because it takes about 2 hours to onset of effect (50). Because it is recommended to wear the patch for no more than 9 hours, application early in the morning would likely require removal and proper disposal by the child at school. For example, if the MPH transdermal system is applied at 0400, it should be removed at 1300. Because more than 50% of MPH drug content remains in the patch after removal at 9 hours, the potential for misuse exists for discarded patches (51).

Evening efficacy.

Multiple marketed extended-release formulations have been shown to be effective 12 hours postdose, as measured in a laboratory classroom, after the drug has been optimized over several weeks. It is important to remember that drug effects are compared with placebo effects in the laboratory classroom trials. In an examination of one such effect—dexmethylphenidate 20 mg at 12 hours—ratings were similar to predose but significantly better than placebo ratings. This is an example of a statistically significant but not clinically relevant outcome. Consequently, many patients may require the addition of an afternoon immediate-release formulation to achieve noticeable efficacy at 12 hours and beyond.

To avoid use of multiple medications for patients who require a longer duration of effect, one MPH extended-release and three AMPH extended-release formulations are available. Laboratory classroom studies in adults demonstrated duration of effect of up to 16 hours for dose-optimized MPH extended-release and mixed amphetamine salts extended-release formulations (52, 53). The duration of effect for lisdexamfetamine in laboratory classroom trials was shown to be 13 hours in children ages 6 to 12 and 14 hours in adults (54, 55). In another laboratory classroom trial, AMPH extended-release oral suspension 2.5 mg/mL also showed efficacy to 13 hours (56).

Because lisdexamfetamine is a prodrug and the l-lysine has to be cleaved from the dextro-amphetamine for the drug to become active, its onset of effect is delayed. In a laboratory classroom study in children ages 6 to 12, onset of effect occurred approximately 1.5 hours after ingestion (55). In the mixed amphetamine salts 16-hour trials, the first onset of effect was measured at 2 hours after dosing in an adolescent laboratory classroom (57).

In addition to differences in onset and duration of effect, formulations differ in the time of peak efficacy and the amount of interpatient variability. Many of the extended-release properties depend on pH in different segments of the gastrointestinal (GI) tract. GI transit time can influence release of the extended-release portion of the drug and shorten or lengthen the duration of efficacy. Food can delay onset of effects for many formulations and shorten it for others (37). For example, administration with a high-fat meal delayed time to maximum concentration (Tmax) by 1 hour for OROS-MPH and by 2.5 hours for mixed amphetamine salts extended release and shortened Tmax by 30 minutes for MPH extended-release orally disintegrating tablet (37). Maximum concentration of the active moiety may also be decreased when administered with a high-fat meal.

Few head-to-head studies have evaluated extended-release stimulants. In two studies comparing dexmethylphenidate extended release with OROS-MPH, the dexmethylphenidate extended-release formulation had a faster onset of effect, whereas OROS-MPH showed greater effect at 10–12 hours (58, 59). When controlled-delivery MPH was compared with OROS-MPH, both were significantly better statistically, compared with placebo, from 1.5 to 7.5 hours (48). The MPH controlled-delivery effect was statistically better, compared with OROS-MPH, at 1.5–4.5 hours, whereas the OROS-MPH effect was statistically better, compared with MPH controlled delivery, at 12 hours (48). Direct comparisons of other MPH products have not been conducted.

Extend Duration of Effect

When prescribing a stimulant, it is important to understand that increasing the dose can prolong duration of efficacy. This is illustrated by the trial comparing 20 mg dexmethylphenidate extended release with 30 mg dexmethylphenidate extended release. Attempted and correct scores on a math test were higher for the patients on the 30 mg dose at 10–12 hours (60). Mixed amphetamine salts extended release is another example. In the mixed amphetamine salts extended-release laboratory classroom trials, onset of effect was seen by 1.5 hours after dosing (61). However, only the 20- and 30-mg doses were effective at 10.5 and 12 hours after dosing. Although extended duration of effect is seen in trials, it may not be seen clinically. If a patient is tolerating a specific dose of medication but the dose is not fully optimized, increasing the dose may be a consideration. Adverse effects should be queried before and after the dose increase to ensure that an attempt at extending the efficacy does not make the drug side effects intolerable.

Single-Isomer Versus Racemic Formulation

Another consideration in treatment is whether to use a single isomer or a racemic formulation. With MPH, the d-threo-enantiomer preferentially crosses the blood-brain barrier. Plasma concentrations are ten to 40 times higher than those of the l-enantiomer (62). When dexmethylphenidate is used, the dose used is half of the racemic drug. Theoretically, one might expect fewer adverse effects with the single d-enantiomer; however, no data available support this notion—adverse effects are similar for dexmethylphenidate and racemic MPH in clinical trials.

In contrast, for AMPH, both the both the d- and l- isomers are effective in treating ADHD (63). Both isomers increase the extracellular concentrations of dopamine and norepinephrine in the brain. Currently, only d-AMPH and d,l-AMPH preparations are on the market. The d,l-AMPH preparations contain d- and l-isomers in a ratio of 50:50 (racemic AMPH and racemic AMPH orally disintegrating tablets) or in a ratio of 3:1 (mixed amphetamine salts, AMPH extended-release oral suspension, AMPH extended-release orally disintegrating tablet, and AMPH extended-release oral suspension) (14, 6466). AMPH extended-release orally disintegrating tablets and AMPH extended-release oral suspension 1.25 mg/mL are bioequivalent to mixed amphetamine salts extended release; however, AMPH extended-release orally disintegrating tablets and AMPH extended-release oral suspension do not contain salt molecules, and their doses are described only as AMPH base.

In 2013, the FDA began to require all newly approved stimulant formulations to express dose of the active moiety rather than the salt form (67). Although the directive was meant to decrease confusion, the goal has not been accomplished. For example, mixed amphetamine salts extended-release 30 mg is equivalent to 18.8-mg AMPH extended-release orally disintegrating tablets or AMPH extended-release oral suspension 1.25 mg/mL, and MPH hydrochloride 30 mg is equivalent to 25.9-mg MPH extended-release orally disintegrating tablets (68, 69). Multiplying the dose of MPH hydrochloride extended-release products by 0.8647 will result in the appropriate dose of MPH extended-release orally disintegrating tablets. Because the conversion factor for AMPH extended-release orally disintegrating tablets depends on the molecular weight of the AMPH salt and because marketed AMPH products vary in their salt composition, the reader is referred to the table in Engelking et al. (67) for the appropriate conversion factors.

Regarding the AMPH orally disintegrating tablets and AMPH extended-release suspension 1.25 mg/mL, 50% of AMPH microparticles are coated for delayed release and 50% are left uncoated for the immediate-release component (69). Although bioequivalent to mixed amphetamine salts extended release, the properties of the orally disintegrating tablets and suspension dosage forms may make these formulations more tolerable for individual patients. With mixed amphetamine salts extended release, half of the drug is released immediately and half approximately 4 hours later (64). AMPH extended-release oral suspension 2.5 mg/mL is bioequivalent to two doses of mixed amphetamine salts extended release dosed 4 hours apart but is also formulated with immediate-release and extended-release microparticles and no salt—and doses are described in mg of AMPH base.

Racemic AMPH is intermediate acting. A single dose measured in a laboratory classroom demonstrated onset and duration of effect at 45 minutes and 10 hours, respectively (70). Racemic AMPH orally disintegrating tablets are bioequivalent to racemic AMPH and are expected to have similar onset and duration, but this formulation has not been evaluated in a laboratory classroom (71). These drugs are often dosed twice daily, 4 to 6 hours apart (67, 72).

Dextro-AMPH is marketed in multiple immediate-release preparations and also as the extended-release prodrug lisdexamfetamine. The immediate-release formulation of dextro-AMPH is dosed two to three times daily (73, 74).

Conclusions

Multiple MPH and AMPH drugs are available for the treatment of ADHD, and the pharmacokinetic and pharmacodynamic properties of various formulations are tightly linked. Choosing the most appropriate medication depends on time of day when symptom control is needed, tolerability, and individual preferences. Small differences in the ratio of immediate-release to extended-release drug may translate to significant differences in onset and duration of effect. It is important have a thorough understanding of the entire ADHD armamentarium to provide the best care for patients.

Center for Psychiatry and Behavioral Medicine, Inc., Las Vegas.
Send correspondence to Dr. Childress ().

Dr. Childress reports receipt of research or writing support, participation on advisory boards, and service as a consultant or speaker for Adlon Therapeutics, Aevi Genomic Medicine, Akili Interactive, Allergan, Arbor Pharmaceuticals, Cingulate Therapeutics, Emalex Biosciences, Ironshore Pharmaceuticals, Jazz Pharmaceuticals, KemPharm, Lundbeck, Neos Therapeutics, Neurovance, NLS Pharma, Otsuka, Pearson, Pfizer, Purdue Pharma, Rhodes Pharmaceuticals, Sunovion, Supernus Pharmaceuticals, Takeda, and Tris Pharma.

References

1 Data and Statistics on Children's Mental Health. Atlanta, Centers for Disease Control and Prevention, 2020. cdc.gov/childrensmentalhealth/data.htmlGoogle Scholar

2 Mannuzza S, Klein RG, Bessler A, et al.: Adult outcome of hyperactive boys: educational achievement, occupational rank, and psychiatric status. Arch Gen Psychiatry 1993; 50:565–576CrossrefGoogle Scholar

3 Kessler RC, Adler L, Barkley R, et al.: The prevalence and correlates of adult ADHD in the United States: results from the National Comorbidity Survey Replication. Am J Psychiatry 2006; 163:716–723CrossrefGoogle Scholar

4 Danielson ML, Bitsko RH, Ghandour RM, et al.: Prevalence of parent-reported ADHD diagnosis and associated treatment among US children and adolescents, 2016. J Clin Child Adolesc Psychol 2018; 47:199–212CrossrefGoogle Scholar

5 Raman SR, Man KKC, Bahmanyar S, et al.: Trends in attention-deficit hyperactivity disorder medication use: a retrospective observational study using population-based databases. Lancet Psychiatry 2018; 5:824–835CrossrefGoogle Scholar

6 Attention Deficit Hyperactivity Disorder: Diagnosis and Management. London, National Institute for Health Care and Excellence, 2019. https://www.nice.org.uk/guidance/ng87Google Scholar

7 Wolraich ML, Hagan JF Jr, Allan C, et al.: Clinical practice guideline for the diagnosis, evaluation, and treatment of attention-deficit/hyperactivity disorder in children and adolescents. Pediatrics 2019; 144:e20192528CrossrefGoogle Scholar

8 Barbaresi WJ, Campbell L, Diekroger EA, et al.: Society for Developmental and Behavioral Pediatrics clinical practice guideline for the assessment and treatment of children and adolescents with complex attention-deficit/hyperactivity disorder. J Dev Behav Pediatr 2020; 41(suppl 2S):S35–S57CrossrefGoogle Scholar

9 Pliszka S: Practice parameter for the assessment and treatment of children and adolescents with attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry 2007; 46:894–921CrossrefGoogle Scholar

10 Highlights of Prescribing Information: Kapvay (Clonidine Hydrochloride) Extended-Release Tablets, for Oral Use. Package insert. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/022331s021lbl.pdfGoogle Scholar

11 Highlights of Prescribing Information: Strattera (Atomoxetine) Capsules for Oral Use. Package insert. Silver Spring, MD, US Food and Drug Administration, 2020. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/021411s049lbl.pdfGoogle Scholar

12 Highlights of Prescribing Information: Intuniv (Guanfacine) Extended-Release Tablets, for Oral Use. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/022037s019lbl.pdfGoogle Scholar

13 Highlights of Prescribing Information: Concerta (Methylphenidate HCl) Extended-Release Tablets, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2013. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021121s038lbl.pdfGoogle Scholar

14 Highlights of Prescribing Information: Dyanavel XR (Amphetamine) Extended-Release Oral Suspension, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/208147s005lbl.pdfGoogle Scholar

15 Top 25 Psychiatric Medications for 2018. Blog. Newburyport, MA, PsychCentral, 2019. https://psychcentral.com/blog/top-25-psychiatric-medications-for-2018Google Scholar

16 New Drug Application Approval (NDA 207690): QuilliChew ER (Methylphenidate Hydrochloride) Extended-Release Chewable Tablets. Silver Spring, MD, US Food and Drug Administration, 2015. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2015/207960Orig1s000ltr.pdfGoogle Scholar

17 FDA-Approved Drugs: Abbreviated New Drug Application (ANDA 200166): Evekeo Oral Tablet. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=200166Google Scholar

18 FDA-Approved Drugs: New Drug Application (NDA 209905): Evekeo Orally Disintegrating Tablet. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=209905Google Scholar

19 New Drug Application Approval (NDA 205831): Aptensio XR (Methylphenidate Hydrochloride Extended-Release) Capsules. Silver Spring, MD, US Food and Drug Administration, 2015. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2015/205831Orig1s000ltr.pdfGoogle Scholar

20 New Drug Application Approval (NDA 208147): Dyanavel XR (Amphetamine) Extended-Release Oral Suspension. Silver Spring, MD, US Food and Drug Administration, 2015. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2015/208147Orig1s000ltr.pdfGoogle Scholar

21 FDA-Approved Drugs: New Drug Application (NDA 204325): Adzenys (Amphetamine) Oral Suspension, Extended Release. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=204325Google Scholar

22 New Drug Application Approval (NDA 204326): Adzenys XR-ODT (Amphetamine) Extended-Release Orally Disintegrating Tablets. Silver Spring, MD, US Food and Drug Administration, 2016. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2016/204326Orig1s000ltr.pdfGoogle Scholar

23 New Drug Application Approval (NDA 022063): Mydayis (Mixed Salts of a Single-Entity Amphetamine Product) Extended-Release Capsules, For Oral Use, CII. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm?event=overview.process&ApplNo=022063Google Scholar

24 New Drug Application Approval (NDA 209311): Jornay PM (Methylphenidate Hydrochloride) Extended-Release. Silver Spring, MD, US Food and Drug Administration, 2018. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2018/209311Orig1s000ltr.pdfGoogle Scholar

25 New Drug Application Approval (NDA 212038): Adhansia XR (Methylphenidate Hydrochloride) Extended-Release Capsules. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2019/212038Orig1s000ltr.pdfGoogle Scholar

26 New Drug Application Approval (NDA 202100): Quillivant XR (Methylphenidate Hydrochloride) for Extended-Release Oral Suspension. Silver Spring, MD, US Food and Drug Administration, 2012. https://www.accessdata.fda.gov/drugsatfda_docs/appletter/2012/202100Orig1s000ltr.pdfGoogle Scholar

27 Zenzedi New Drug Application CII (Dextroamphetamine Sulfate) [cited July 12, 2020]. https://zenzedi.com/index.phpGoogle Scholar

28 Highlights of Prescribing Information: Ritalin LA (Methylphenidate Hydrochloride) Extended-Release Capsules for Oral Use. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021284s041lbl.pdfGoogle Scholar

29 Once Daily Metadate CD: (Methylphenidate Hcl, USP) Extended-Release Capsules. Package insert. Silver Spring, MD, US Food and Drug Administration, 2014. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021259s030lbl.pdfGoogle Scholar

30 Methylin Oral Solution (Methylphenidate HCl). Package insert. Silver Spring, MD, US Food and Drug Administration, 2013. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021419s014lbl.pdfGoogle Scholar

31 Pelham WE Jr, Greenslade KE, Vodde-Hamilton M, et al.: Relative efficacy of long-acting stimulants on children with attention deficit-hyperactivity disorder: a comparison of standard methylphenidate, sustained-release methylphenidate, sustained-release dextroamphetamine, and pemoline. Pediatrics 1990; 86:226–237CrossrefGoogle Scholar

32 Findling RL, Bukstein OG, Melmed RD, et al.: A randomized, double-blind, placebo-controlled, parallel-group study of methylphenidate transdermal system in pediatric patients with attention-deficit/hyperactivity disorder. J Clin Psychiatry 2008; 69:149–159CrossrefGoogle Scholar

33 Highlights of Prescribing Information: Focalin (Dexmethylphenidate Hydrochloride) Tablets, for Oral Use, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021278s026lbl.pdfGoogle Scholar

34 Highlights of Prescribing Information: Focalin XR (dexmethylphenidate hydrochloride) Extended-Release Capsules, for Oral Use, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021802s036lbl.pdfGoogle Scholar

35 Kimko H, Gibiansky E, Gibiansky L, et al.: Population pharmacodynamic modeling of various extended-release formulations of methylphenidate in children with attention deficit hyperactivity disorder via meta-analysis. J Pharmacokinet Pharmacodyn 2012; 39:161–176CrossrefGoogle Scholar

36 Li L, Wang Y, Uppoor RS, et al.: Exposure-response analyses of blood pressure and heart rate changes for methylphenidate in healthy adults. J Pharmacokinet Pharmacodyn 2017; 44:245–262CrossrefGoogle Scholar

37 Childress AC, Komolova M, Sallee FR: An update on the pharmacokinetic considerations in the treatment of ADHD with long-acting methylphenidate and amphetamine formulations. Expert Opin Drug Metab Toxicol 2019; 15:937–974CrossrefGoogle Scholar

38 Faraone SV: Using meta-analysis to compare the efficacy of medications for attention-deficit/hyperactivity disorder in youths. PT 2009; 34:678–694Google Scholar

39 Marcus SC, Wan GJ, Kemner JE, et al.: Continuity of methylphenidate treatment for attention-deficit/hyperactivity disorder. Arch Pediatr Adolesc Med 2005; 159:572–578CrossrefGoogle Scholar

40 Faraone SV, Upadhyaya HP: The effect of stimulant treatment for ADHD on later substance abuse and the potential for medication misuse, abuse, and diversion. J Clin Psychiatry 2007; 68:e28CrossrefGoogle Scholar

41 Cortese S, Adamo N, Del Giovane C, et al.: 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; 5:727–738CrossrefGoogle Scholar

42 Hodgkins P, Shaw M, Coghill D, et al.: Amfetamine and methylphenidate medications for attention-deficit/hyperactivity disorder: complementary treatment options. Eur Child Adolesc Psychiatry 2012; 21:477–492CrossrefGoogle Scholar

43 Liu T, Gobburu JVS, Po MD, et al.: Pharmacokinetics of HLD200, a delayed-release and extended-release methylphenidate: evaluation of dose proportionality, food effect, multiple-dose modeling, and comparative bioavailability with immediate-release methylphenidate in healthy adults. J Child Adolesc Psychopharmacol 2019; 29:181–191CrossrefGoogle Scholar

44 Childress AC, Cutler AJ, Marraffino A, et al.: A randomized, double-blind, placebo-controlled study of HLD200, a delayed-release and extended-release methylphenidate, in children with attention-deficit/hyperactivity disorder: an evaluation of safety and efficacy throughout the day and across settings. J Child Adolesc Psychopharmacol 2020; 30:2–14CrossrefGoogle Scholar

45 Brams M, Muniz R, Childress A, et al.: A randomized, double-blind, crossover study of once-daily dexmethylphenidate in children with attention-deficit hyperactivity disorder: rapid onset of effect. CNS Drugs 2008; 22:693–704CrossrefGoogle Scholar

46 Childress AC, Kando JC, King TR, et al.: Early-onset efficacy and safety pilot study of amphetamine extended-release oral suspension in the treatment of children with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol 2019; 29:2–8CrossrefGoogle Scholar

47 Wigal SB, Childress AC, Belden HW, et al.: NWP06, an extended-release oral suspension of methylphenidate, improved attention-deficit/hyperactivity disorder symptoms compared with placebo in a laboratory classroom study. J Child Adolesc Psychopharmacol 2013; 23:3–10CrossrefGoogle Scholar

48 Swanson JM, Wigal SB, Wigal T, et al.: A comparison of once-daily extended-release methylphenidate formulations in children with attention-deficit/hyperactivity disorder in the laboratory school (the Comacs Study). Pediatrics 2004; 113:e206–e216CrossrefGoogle Scholar

49 Armstrong RB, Damaraju CV, Ascher S, et al.: Time course of treatment effect of OROS methylphenidate in children with ADHD. J Atten Disord 2012; 16:697–705CrossrefGoogle Scholar

50 Highlights of Prescribing Information: Daytrana (Methylphenidate Transdermal System), CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021514s030lbl.pdfGoogle Scholar

51 Patrick KS, Straughn AB, Perkins JS, et al.: Evolution of stimulants to treat ADHD: transdermal methylphenidate. Hum Psychopharmacol 2009; 24:1–17CrossrefGoogle Scholar

52 Wigal SB, Wigal T, Childress A, et al.: The time course of effect of multilayer-release methylphenidate hydrochloride capsules: a randomized, double-blind study of adults with ADHD in a simulated adult workplace environment. J Atten Disord 2020; 24:373–383CrossrefGoogle Scholar

53 Wigal T, Childress A, Frick G, et al.: Effects of SHP465 mixed amphetamine salts in adults with ADHD in a simulated adult workplace environment. Postgrad Med 2018; 130:111–121CrossrefGoogle Scholar

54 Wigal T, Brams M, Gasior M, et al.: Randomized, double-blind, placebo-controlled, crossover study of the efficacy and safety of lisdexamfetamine dimesylate in adults with attention-deficit/hyperactivity disorder: novel findings using a simulated adult workplace environment design. Behav Brain Funct 2010; 6:34CrossrefGoogle Scholar

55 Wigal SB, Kollins SH, Childress AC, et al.: A 13-hour laboratory school study of lisdexamfetamine dimesylate in school-aged children with attention-deficit/hyperactivity disorder. Child Adolesc Psychiatry Ment Health 2009; 3:17CrossrefGoogle Scholar

56 Childress AC, Wigal SB, Brams MN, et al.: Efficacy and safety of amphetamine extended-release oral suspension in children with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol 2018; 28:306–313CrossrefGoogle Scholar

57 Wigal S, Lopez F, Frick G, et al.: A randomized, double-blind, 3-way crossover, analog classroom study of SHP465 mixed amphetamine salts extended-release in adolescents with ADHD. Postgrad Med 2019; 131:212–224CrossrefGoogle Scholar

58 Muniz R, Brams M, Mao A, et al.: Efficacy and safety of extended-release dexmethylphenidate compared with d,l-methylphenidate and placebo in the treatment of children with attention-deficit/hyperactivity disorder: a 12-hour laboratory classroom study. J Child Adolesc Psychopharmacol 2008; 18:248–256CrossrefGoogle Scholar

59 Silva R, Muniz R, McCague K, et al.: Treatment of children with attention-deficit/hyperactivity disorder: results of a randomized, multicenter, double-blind, crossover study of extended-release dexmethylphenidate and D,L-methylphenidate and placebo in a laboratory classroom setting. Psychopharmacol Bull 2008; 41:19–33Google Scholar

60 Silva RR, Brams M, McCague K, et al.: Extended-release dexmethylphenidate 30 mg/d versus 20 mg/d: duration of attention, behavior, and performance benefits in children with attention-deficit/hyperactivity disorder. Clin Neuropharmacol 2013; 36:117–121CrossrefGoogle Scholar

61 McCracken JT, Biederman J, Greenhill LL, et al.: Analog classroom assessment of a once-daily mixed amphetamine formulation, SLI381 (Adderall XR), in children with ADHD. J Am Acad Child Adolesc Psychiatry 2003; 42:673–683CrossrefGoogle Scholar

62 Coury D: Dexmethylphenidate (Focalin) for ADHD. Med Lett Drugs Ther 2002; 44:45–46Google Scholar

63 Heal DJ, Smith SL, Gosden J, et al.: Amphetamine, past and present—a pharmacological and clinical perspective. J Psychopharmacol 2013; 27:479–496CrossrefGoogle Scholar

64 Highlights of Prescribing Information: Adderall XR (Mixed Salts of a Single-Entity Amphetamine Product) Extended Release Capsules, for Oral Use, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/021303s034lbl.pdfGoogle Scholar

65 Highlights of Prescribing Information: Adzenys XR-ODT (Amphetamine) Extended-Release Orally Disintegrating Tablets, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/204326s002lbl.pdfGoogle Scholar

66 Highlights of Prescribing Information: Adzenys ER (Amphetamine) Extended-Release Oral Suspension, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2017. https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/204325s000lbl.pdfGoogle Scholar

67 Engelking D, Childress AC, McMahen R, et al.: How to dose attention-deficit/hyperactivity disorder medications without a grain of salt. J Child Adolesc Psychopharmacol 2018; 28:576–577CrossrefGoogle Scholar

68 Sikes CR, McMahen RL, Stark JG, et al.: Pharmacokinetics of a new amphetamine extended-release oral suspension in children with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol 2018; 28:29–35CrossrefGoogle Scholar

69 Stark JG, Engelking D, McMahen R, et al.: Pharmacokinetics of a novel amphetamine extended-release orally disintegrating tablet in children with attention-deficit/hyperactivity disorder. J Child Adolesc Psychopharmacol 2017; 27:216–222CrossrefGoogle Scholar

70 Childress AC, Brams M, Cutler AJ, et al.: The efficacy and safety of Evekeo, racemic amphetamine sulfate, for treatment of attention-deficit/hyperactivity disorder symptoms: a multicenter, dose-optimized, double-blind, randomized, placebo-controlled crossover laboratory classroom study. J Child Adolesc Psychopharmacol 2015; 25:402–414CrossrefGoogle Scholar

71 Caras S, Sharpe T: Pharmacokinetics of AR19, an immediate-release amphetamine sulfate formulation designed to deter manipulation for administration via nonoral routes: bioequivalence to reference racemic amphetamine sulfate, dose proportionality, and food effect. J Child Adolesc Psychopharmacol 2020; 30:69–80CrossrefGoogle Scholar

72 Highlights of Prescribing Information: Evekeo ODT (Amphetamine Sulfate) Orally Disintegrating Tablets, CII. Package insert. Silver Spring, MD, US Food and Drug Administration, 2019. https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/209905s000lbl.pdfGoogle Scholar

73 Dextroamphetamine Sulfate. Package insert. Silver Spring, MD, US Food and Drug Administration, 2013. https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/040361Orig1s017lbl.pdfGoogle Scholar

74 Zenzedi (Dextroamphetamine Sulfate, USP) Tablets. Package insert. Atlanta, Arbor Pharmaceuticals, 2017. https://zenzedi.com/docs/PIandMedicationGuide.pdfGoogle Scholar