Title of Invention

ABUSE RESISTANT AMPHETAMINE COMPOUNDS

Abstract The invention describes compounds compositions and methods or using the some comprising a chemical moiety covalently attached to amphetamine. These compounds and compositions are useful for reducing or preventing abuse and overdose of amphetamine. These compounds and compositions find particular use in providing an abuse-resistant alternative treatment for certain disorders, such as attention deficit hyperactivity disorder (ADHD), ADD, narcolepsy, and obesity. Oral bioavailability of amphetamine is maintained at therapeutically useful doses. At higher doses bioavailability is substantially reduced, thereby providing a method of reducing oral abuse liability. Further, compounds and compositions of the invention decrease the bioavailability of amphetamine by parenteral routes, such as intravenous or intranasal administration, further limiting their abuse liability.
Full Text WO 2005/000334 PCT/US2004/017204
ABUSE RESISTANT AMPHETAMINE COMPOUNDS
CROSS REFERENCE RELATED APPLICATIONS
[001] This application claims benefit under 35 U.S.C. 119(e) to U.S. provisional
application No. 60/473,929 filed May 29, 2003 and provisional application No.
60/567,801 filed May 5, 2004, both of which are hereby incorporated by reference in
their entirety.
BACKGROUND OF THE INVENTION
(i) Field of the Invention
[002] The invention relates to amphetamine compounds, compositions and
methods of delivery and use comprising amphetamine covalently bound to a
chemical moiety.
[003] The invention relates to compounds comprised of amphetamine covalently
bound to a chemical moiety in a manner that diminishes or eliminates
pharmacological activity of amphetamine until released. The conjugates are stable
in tests that simulate procedures likely to be used by illicit chemists in attempts to
release amphetamine. The invention further provides for methods of therapeutic
delivery of amphetamine compositions by oral administration. Additionally, release
of amphetamine following oral administration occurs gradually over an extended
period of time thereby eliminating spiking of drug levels. When taken at doses
above the intended prescription, the bioavailability of amphetamine, including peak
levels and total amount of drug absorbed, is substantially decreased. This decreases
the potential for amphetamine abuse which often entails the use of extreme doses (1
g or more a day). The compositions are also resistant to abuse by parenteral routes
of administration, such as intravenous "shooting", intranasal "snorting", or
inhalation "smoking", that are often employed in illicit use. The invention thus
provides a stimulant based treatment for certain disorders, such as attention deficit
hyperactivity disorder (ADHD), which is commonly treated with amphetamine.
Treatment of ADHD with compositions of the invention results in substantially
decreased abuse liability as compared to existing stimulant treatments.

WO 2005/000334 PCT/US2004/017204
2
(ii) Background of the Invention
[004] The invention is directed to amphetamine conjugate compounds,
compositions, and methods of manufacture and use thereof. In particular, the
invention is directed to an anti-abuse/sustained release formulation which maintains
its therapeutic effectiveness when administered orally. The invention further relates
to formulations which diminish or reduce the euphoric effect while maintaining
therapeutically effective blood concentrations following oral administration.
[005] Amphetamine is prescribed for the treatment of various disorders, including
attention deficit hyperactivity disorder (ADHD), obesity and narcolepsy.
Amphetamine and methamphetamine stimulate the central nervous system and have
been used medicinally to treat ADHD, narcolepsy and obesity. Because of its
stimulating effects amphetamine and its derivatives (e.g., amphetamine analogues)
are often abused. Similarly, p-methoxyamphetamine, methylenedioxyamphetaminc,
2,5-dimethoxy-4-methylamphetamine, 2,4,5-trimethoxyamphetamine and 3,4-
methylenedioxymethamphetamine are also often abused.
[006] In children with attention deficit hyperactivity disorder (ADHD), potent CNS
stimulants have been used for several decades as a drug treatment given either alone
or as an adjunct to behavioral therapy. While methylphenidate (Ritalin) has been the
most frequently prescribed stimulant, the prototype of the class, amphetamine
(alpha-methyl phenethvlamine) has been used all along and increasingly so in recent
years. (Bradley C, Bowen M, "Amphetamine (Benzedrine) therapy of children's
behavior disorders." American Journal of Orthopsvchiatry 11: 92) (1941).
[007] The potential for abuse of amphetamines is a major drawback to its use. The
high abuse potential has earned it Schedule II status according to the Controlled
Substances Act (CSA). Schedule II classification is reserved for those drugs that
have accepted medical use but have the highest potential for abuse. The abuse
potential of amphetamine has been known for many years and the FDA requires the
following black box warning in the package inserts of products:

WO 2005/000334 PCT/US2004/017204
3
AMPHETAMINES HAVE A HIGH POTENTIAL FOR ABUSE. ADMINISTRATION OF
AMPHETAMINES FOR PROLONGED PERIODS OF TIME MAY LEAD TO DRUG
DEPENDENCE AND MUST BE AVOIDED. PARTICULAR ATTENTION SHOULD BE PAID
TO THE POSSIBILITY OF SUBJECTS OBTAINING AMPHETAMINES FOR
NONTHERAPEUTIC USE OR DISTRIBUTION TO OTHERS, AND THE DRUGS SHOULD BE
PRESCRIBED OR DISPENSED SPARINGLY.
[008] Furthermore, recent developments in the abuse of prescription drug products
increasingly raise concerns about the abuse of amphetamine prescribed for ADHD.
Similar to OxyContin, a sustained release formulation of a potent narcotic analgesic,
Adderall XR® represents a product with increased abuse liability relative to the
single dose tablets. The source of this relates to the higher concentration of
amphetamine in each tablet and the potential for release of the full amount of active
pharmaceutical ingredient upon crushing. Therefore, like OxyContin, it may be
possible for substance abusers to obtain a high dose of the pharmaceutical with rapid
onset by snorting the powder or dissolving it in water and injecting it. (Cone, E. J.,
R. V. Fant, et al., "Oxycodone involvement in drug abuse deaths: a DAWN-based
classification scheme applied to an oxycodone postmortem database containing over
1000 cases." J Anal Toxicol 27(2); 57-67; discussion 67) (2003).
[009] It has been noted recently that "53 percent of children not taking medication
for ADHD knew of students with the disorder either giving away or selling their
medications. And 34 percent of those being treated for the disorder acknowledged
they had been approached to sell or trade them." (Dartmouth-Hitchcock, 2003)
"Understanding ADHD Stimulant Abuse." http://12.42.224.168/healthyliving/
familyhome/jan03familyhomestimulantabuse.htm). In addition, it was reported that
students at one prep school obtained Dexedrine and Adderall to either swallow
tablets whole or crush and sniff them. (Dartmouth-Hitchcock (2003).
[010] According to the drug enforcement administration (DEA, 2003):
Methylphenidate and amphetamine can be abused orally or the tablets can be
crushed and snorted or dissolved in water and injected. The pattern of abuse
is characterized by escalation in dose, frequent episodes of binge use
followed by severe depression and an overpowering desire to continue the
use of these drugs despite serious adverse medical and social consequences.

WO 2005/000334 PCT/US2004/017204
4
Rendering this potent stimulant resistant to abuse, particularly by parenteral
routes such as snorting or injecting, would provide considerable value to this
otherwise effective and beneficial prescription medication.
(DEA (2003). "Stimulant Abuse By School Age Children: A Guide for
School
Officials."http://www.deadiversion.usdoj.gov/pubs/brochures/stimulant/stim
ulant_abuse.htm).
[011] Typically, sustained release formulations contain drug particles mixed with
or covered by a polymer material, or blend of materials, which are resistant to
degradation or disintegration in the stomach and/or in the intestine for a selected
period of time. Release of the drug may occur by leeching, erosion, rupture,
diffusion or similar actions depending upon the nature of the polymer material or
polymer blend used. Additionally, these formulations are subject to breakdown
following relatively simple protocols which allows for abuse of the active
ingredient.
[012] Conventionally, pharmaceutical manufacturers have used hydrophilic
hydrocolloid gelling polymers such as hydroxypropyl methylcellulose,
hydroxypropyl cellulose or Pullulan to formulate sustained release tablets or
capsules. These polymers first form a gel when exposed to an aqueous environment
of low pH thereby slowly diffusing the active medicament which is contained within
the polymer matrix. When the gel enters a higher pH environment such as that
found in the intestines, however, it dissolves resulting in a less controlled drug
release. To provide better sustained release properties in higher pH environments,
some pharmaceutical manufacturers use polymers which dissolve only at higher
pHs, such as acrylic resins, acrylic latex dispersions, cellulose acetate phthalate, and
hydroxypropyl methylcellulose phthalate, either alone or in combination with
hydrophilic polymers.
[013] These formulations are prepared by combining the medicament with a finely
divided powder of the hydrophilic polymer, or the hydrophilic and water-insoluble
polymers. These ingredients are mixed and granulated with water or an organic
solvent and the granulation is dried. The dry granulation is then usually further
blended with various pharmaceutical additives and compressed into tablets.

WO 2005/000334 PCT/US2004/017204
5
[014] Although these types of formulations have been successfully used to
manufacture dosage forms which demonstrate sustained release properties, these
formulations are subject to several shortcomings including uneven release and are
subject to abuse.
[015] The need exists for an abuse resistant dosage form of amphetamine which is
therapeutically effective. Further the need exists for an amphetamine dosage form
which provides sustained release and sustained therapeutic effect.
SUMMARY OF INVENTION
[016] The invention provides covalent attachment of amphetamine and derivatives
or analogs thereof to a variety of chemical moieties. The chemical moieties may
include any substance which results in a prodrug form, i.e., a molecule which is
converted into its active form in the body by normal metabolic processes. The
chemical moieties may be for instance, amino acids, peptides, glycopeptides,
carbohydrates, nucleosides, or vitamins.
[017] The chemical moiety is covalently attached either directly or indirectly
through a linker to the amphetamine. The site of attachment is typically determined
by the functional group(s) available on the amphetamine.
[018] In one embodiment of the invention, the chemical moiety is a carrier peptide
as defined herein. The carrier peptide may be attached to amphetamine through the
carrier's N-terminus, C-terminus or side chain of an amino acid which may be either
a single amino acid or part of a longer chain sequence (i.e. a dipeptide, tripeptide, an
oligopcptide or a polypeptide). Preferably, the carrier peptide is (i) an amino acid,
(ii) a dipeptide, (iii) a tripeptide, (iv) an oligopeptide, or (v) polypeptide. The carrier
peptide may also be (i) a homopolymer of a naturally occurring amino acid, (ii) a
heteropolymer of two or more naturally occurring amino acids, (iii) a homopolymer
of a synthetic amino acid, (iv) a heteropolymer of two or more synthetic amino
acids, or (v) a heteropolymer of one or more naturally occurring amino acids and
one or more synthetic amino acids, A further embodiment of the carrier and/or
conjugate is that the unattached portion of the carrier/conjugate may be in a free and

WO 2005/000334 PCT/US2004/017204
6
unprotected state. Preferably, synthetic amino acids with alkyl side chains are
selected from alkyls of C1-C17 in length and more preferably from C1-C6 in length.
[019] Covalent attachment of a chemical moiety to amphetamine can decrease its
pharmacological activity when administered through injection or intranasally.
Compositions of the invention, however, provide amphetamine covalently attached
to a chemical moiety which remains orally bioavailable. The bioavailability is a
result of the hydrolysis of the covalent linkage following oral administration.
Hydrolysis is time-dependent, thereby allowing amphetamine to become available in
its active form over an extended period of time. In one embodiment, the
composition provides oral bioavailability which resembles the pharmacokinetics
observed for extended release formulations. In another embodiment, release of
amphetamine is diminished or eliminated when delivered by parenteral routes.
[020] In one embodiment, the compositions maintain their effectiveness and abuse
resistance following the crushing of the tablet, capsule or other oral dosage form. In
contrast, conventional extended release formulations used to control the release of
amphetamine through incorporation into matrices are subject to release of up to the
entire amphetamine content immediately following crushing. When the content of
the crushed tablet is injected or snorted, the large dose of amphetamine produces the
"rush" effect sought by addicts.
[021] In one embodiment, the amphetamine is attached to a single amino acid
which is either naturally occurring or a synthetic amino acid. In another
embodiment, the amphetamine is attached to a dipeptide or tripeptide, which could
be any combination of the naturally occurring amino acids and synthetic amino
acids. In another embodiment, the amino acids are selected from L-amino acids for
digestion by proteases.
[022] In another embodiment, the side chain attachment of amphetamine to the
polypeptide or amino acid are selected from homopolymers or heteropolymers of
glutamic acid, aspartic acid, serine, lysine, cysteine, threonine, asparagine, arginine,
tyrosine, and glutamine. Examples of peptides include, Lys, Ser, Phe, Gly-Gly-Gly,
Leu-Ser, Leu-Glu, homopolymers of Glu and Leu, and heteropolymers of (Glu)n-

WO 2005/000334 PCT/US2004/017204
7
Leu-Ser. In a preferred embodiment, the composition is selected from Lys-Amp,
Ser-Amp, Phe-Amp, and Gly-Gly-GIy-Amp.
[023] In another embodiment, the invention provides a carrier and amphetamine
which are bound to each other but otherwise unmodified in structure. This
embodiment may further be described as the carrier having a free carboxy and/or
amine terminal and/or side chain groups other than at the location of attachment for
the amphetamine. In a preferred embodiment, the carrier, whether a single amino
acid, dipeptide, tripeptide, oligopeptide or polypeptide, comprises only naturally
occurring amino acids.
[024] Another embodiment of the invention provides a method for delivering
amphetamine dosage which prevents euphoria, comprising administering to a patient
in need a composition formulated for oral dosage comprising amphetamine
covalently attached to a chemical moiety wherein said blood levels of amphetamine
maintain a therapeutically effect level but do not result in a euphoric effect.
[025] In another embodiment, the covalent attachment of a chemical moiety
substantially decreases the potential for overdose by decreasing the toxicity of
amphetamine at doses above those considered therapeutic, while maintaining its
pharmaceutical activity within a normal dose range. Covalent attachment of the
chemical moiety may decrease or eliminate the pharmacological activity of
amphetamine. Therefore, restoring activity requires release of the amphetamine
from the chemical moiety. At higher doses partial or complete saturation of
processes responsible for amphetamine release may be reached thus diminishing or
eliminating the release of harmful levels of active amphetamine. For example,
aspects of pharmacological activity, release, saturation are further depicted in figures
1-55.
[026] In another embodiment of the invention, the covalent attachment of a
chemical moiety substantially decreases the potential for overdose by decreasing the
rate or overall amount of absorption of the amphetamine when given at doses above
those considered therapeutic.

WO 2005/000334 PCT/US2004/017204
8
[027] In another embodiment of the invention, the covalent attachment of a
chemical moiety substantially decreases the potential for overdose by increasing the
rate or overall amount of clearance of amphetamine when given at doses above those
considered therapeutic.
[028] Another embodiment provides a method of treating a patient suffering from
attention deficit hyperactivity disorder, narcolepsy or obesity comprising providing,
administering, prescribing, etc. compositions of the invention.
[029] Another embodiment of the invention provides a method for delivering
amphetamine, comprising providing a patient with a therapeutically effective
amount of amphetamine covalently attached to a chemical moiety which provides a
therapeutically bioequivalent AUC when compared to amphetamine alone but does
not provide a Cmax which results in euphoria when taken orally.
[030] Other objects, advantages and embodiments of the invention are described
below and will be obvious from this description and practice of the invention.
BRIEF DESCRIPTION OF DRAWINGS
[031] Fig. 1. Synthesis of amino acid amphetamine conjugates.
[032] Fig. 2. Synthesis of lysine amphetamine conjugate.
[033] Fig. 3. Synthesis of serine amphetamine conjugate.
[034] Fig. 4. Synthesis of phenylalanine amphetamine conjugate.
[035] Fig. 5. Synthesis of triglycine amphetamine conjugate.
[036] Fig. 6. Plasma concentrations of J-amphetamine from individual animals
orally administered ^-amphetamine or L-lysine-d-amphetamine.
[037] Fig. 7. Plasma concentrations of d-amphetamine following oral
administration of d-amphetamine sulfate or L-lysine-d-amphetamine (1.5mg/kg d-
amphetamine base) to rats (ELISA analysis).
[038] Fig. 8. Plasma concentrations of d-amphetamine following oral
administration of d-amphetamine sulfate or L-lysine-d-amphetamine (3 mg/kg d-
amphetamine base) to rats (ELISA analysis).

WO 2005/000334 PCT/US2004/017204
9
[039] Fig. 9. Plasma concentrations of d-amphetamine following oral
administration of (/-amphetamine sulfate or L-lysine-d-amphetamine (6 mg/kg d-
amphetamine base) to rats (ELISA analysis).
[040] Fig. 10. Plasma concentrations of d-amphetamine at 30-minutes post-dose
for escalating doses of L-lysine-d-amphe famine or d-amphetamine sulfate (ELISA
analysis).
[041] Fig. 11. Plasma concentrations of d-amphetamine following oral
administration of L-lysine-d-amphetamine or d-amphetamine sulfate (60 mg/kg d-
amphetamine base) to rats (ELISA analysis).
[042] Fig. 12. Plasma concentrations of d-amphetamine following intranasal
administration of L-lysine-d-amphetamine or d-amphetamine sulfate (3 mg/kg d-
amphetamine base) to rats (ELISA analysis).
[043] Fig. 13. Plasma concentrations of d-amphetamine following bolus
intravenous administration of L-Iysine-d-amphetamine or d-amphetamine sulfate
(1.5mg/kg d-amphetamine base) to rats (ELISA analysis).
[044] Fig. 14. Plasma concentrations of d-amphetamine levels following oral
administration of Dexadrine Spansule capsules, crushed Dexadrine Spansule
capsules, or L-lysine-d-amphetamine (3 mg/kg d-amphetamine base) to rats (ELISA
analysis).
[045] Figs. 15A-B. Plasma concentrations of d-amphetamine in ng/mL (Fig. 15A),
and in uM (Fig. 15B), following oral administration of L-lysine-d-amphetamine or
d-amphetamine sulfate (1.5mg/kg d-amphetamine base) to rats (LC/MS/MS
analysis).
[046] Figs. 16A-B. Plasma concentrations of d-amphetamine in ng/mL (Fig. 16A),
and in uM (Fig. 16B), following oral administration of L-lysine-d-amphetamine or
d-amphetamine sulfate (3 mg/kg d-amphetamine base) to rats (LC/MS/MS analysis).
[047] Figs. 17A-B. Plasma concentrations of d-amphetamine in ng/mL (Fig. 17A),
and in uM (Fig. 17B), following oral administration of L-lysine-d-amphetamine or
d-amphetamine sulfate (6 mg/kg d-amphetamine base) to rats (LC/MS/MS analysis).

WO 2005/000334 PCT/US2004/017204
10
[048] Figs. 18A-B. Plasma concentrations of d-amphetamine in ng/mL (Fig. 18A),
and in uM (Fig. 18B), following oral administration of L-lysine-d-amphetamine or
d-amphetamine sulfate (12 mg/kg d-amphetamine base) to rats (LC/MS/MS
analysis).
[049] Figs. 19A-B. Plasma concentrations of d-amphetamine in ng/mL (Fig. 19A),
and in uM (Fig. 19B), following oral administration of or d-amphetamine sulfate (60
mg/kg d-amphetamine base) to rats (LC/MS/MS analysis).
[050] Fig. 20. Comparative bioavailability (Cmax) of L-lysine-d-amphetamine and
d-amphetamine in proportion to escalating human equivalent doses in rats (mg/kg d-
amphetamine base).
[051] Fig. 21. Comparative bioavailability (AUCinf) of L-lysine-d-amphetamine
and d-amphetamine in proportion to escalating doses in rats (mg/kg d-amphetamine
base).
[052] Fig. 22. Comparative Bioavailability (AUCinf) of L-lysine-d-amphetamine
and d-amphetamine in proportion to escalating human equivalent doses in rats
(mg/kg d-amphetamine base).
[053] Fig. 23. Plasma concentrations of d-amphetamine following intranasal
administration of L-lysine-d-amphetamine or d-amphetamine sulfate (3 mg/kg d-
amphetamine base) to rats (LC/MS/MS analysis).
[054] Fig. 24. Plasma concentrations of d-amphetamine and L-lysine-d-
amphetamine in ng/mL (Fig. 24A), and in M (Fig. 24B), following intranasal
administration of L-lysine-d-amphetamine or d-amphetamine sulfate (3 mg/kg d-
amphetamine base) to rats (LC/MS/MS analysis).
[055] Fig. 25. Plasma concentrations of d-amphetamine following bolus
intravenous administration of L-lysine-d-amphetamine or d-amphetamine sulfate
(1.5 mg/kg d-amphetamine base) to rats (LC/MS/MS analysis).
[056] Figs. 26A-B. Plasma concentrations of d-amphetamine in ng/mL (Fig. 26A),
and in M (Fig. 26B), following intranasal administration of L-lysine-d-

WO 2005/000334 PCT/US2004/017204
11
amphetamine or d-amphetamine sulfate (3 mg/kg d-amphetamine base) to rats
(LC/MS/MS analysis).
[057] Fig. 27. Mean plasma concentration time profile of L-lysine-d-amphetamine
following 30-min intravenous infusion (2 mg/kg) or oral administration of L-lysine-
d-amphetamine (2 mg/kg) in conscious male beagle dogs (n=3).
[058] Fig. 28. Plasma concentration time profile of d-amphetamine following 30-
min intravenous infusion or oral administration of L-lysine-d-amphetamine (2
mg/kg) in conscious male beagle dogs (n=3).
[059] Figs. 29A-B. Mean plasma concentration time profile of L-Iysine-d-
amphetamine and d-amphetamine levels in ng/ml (Fig. 29A), and in uM (Fig. 29B),
following 30-min intravenous infusion (2 mg/kg) in conscious male beagle dogs
(n=3).
[060] Figs. 30A-B. Mean plasma concentration time profile of L-lysine-d-
amphetamine and d-amphetamine levels in ng/ml (Fig. 30A), and in nM (Fig, 30B),
following oral administration of L-Iysine-d-amphetamine (2 mg/kg) in conscious
male beagle dogs (n=3).
[061] Figs. 31A-B. Individual plasma concentration time profile of L-Iysine-d-
amphetamine following intravenous administration (Fig. 31 A) or oral administration
(Fig. 31B) of L-lysine-d-amphetamine in conscious male beagle dogs. The oral
formulation used comprises solution and 0.2 mg/mL in water.
[062] Figs. 32A-B. Individual plasma concentration time profile of d-amphetamine
following intravenous administration (Fig. 32A) or oral administration (Fig. 32B) of
L-lysine-d-amphetamine in conscious male beagle dogs.
[063] Fig. 33. Plasma concentrations of d-amphetamine following oral
administration of L-lysine-d-amphetamine or d-amphetamine sulfate (1.8 mg/kg d-
amphetamine base) to male dogs.
[064] Fig. 34. Plasma concentrations of d-amphetamine following oral
administration of L-lysine-d-amphetamine or d-amphetamine sulfate (1.8 mg/kg d-
amphetamine base) to female dogs.

WO 2005/000334 PCT/US2004/017204
12
[065] Fig. 35. Mean blood pressure following intravenous bolus injection of
increasing amounts of L-lysine-d-amphetamine or d-amphetamine in male and
female dogs.
[066] Fig. 36. Left ventricular blood pressure following intravenous bolus injection
of increasing amounts of L-lysine-d-amphetamine or d-amphetamine in male and
female dogs.
[067] Fig. 37. Locomotor activity of rats following oral administration of L-lysine-
d-amphetamine or d-amphetamine (5 hour time-course).
[068] Fig. 38. Locomotor activity of rats following oral administration of L-lysine-
d-amphetamine or d-amphetamine (12 hour time-course).
[069] Fig. 39. Locomotor activity of rats following intranasal administration of L-
lysine-d-amphetamine or d-amphetamine (I hour time-course).
[070] Fig. 40. Locomotor activity of rats following intranasal administration (with
carboxymethylcellulose) of L-lysine-d-amphetamine or d-amphetamine (2 hour
time-course).
[071] Fig. 41. Locomotor activity of rats following intravenous administration of
L-lysine-d-arnphetamine or d-amphetamine (3 hour time-course).
[072] Fig. 42. Intranasal bioavailability of abuse-resistant amphetamine amino
acid-, di-, and tri-peptide conjugates (ELISA analysis).
[073] Fig. 43. Oral bioavailability of abuse-resistant amphetamine amino acid-, di-,
and tri-peptidc conjugates (ELISA analysis).
[074] Fig. 44. Intravenous bioavailability of an abuse-resistant amphetamine tri-
peptide conjugate (ELISA analysis),
[075] Fig. 45. Intranasal bioavailability of an abuse-resistant amphetamine amino
acid conjugate (ELISA analysis).
[076] Fig, 46. Oral bioavailability of an abuse-resistant amphetamine amino acid

WO 2005/000334 PCT/US2004/017204
13
[077] Fig. 47. Intravenous bioavailability of abuse-resistant amphetamine amino
acid-, di-, and tri-peptide conjugates (ELISA analysis).
[078] Fig. 48. Intranasal bioavailability of an abuse-resistant amphetamine amino
tri-peptide conjugate (ELISA analysis).
[079] Fig. 49. Intranasal bioavailability of abuse-resistant amphetamine amino
acid-, and di-peptide conjugates (ELISA analysis).
[080] Fig. 50. Intranasal bioavailability of an abuse-resistant amphetamine di-
peptide conjugate containing D- and L- amino acid isomers (ELISA analysis).
[081] Figs. 51A-B. Plasma concentrations of d-amphetamine and L-lysine-d-
amphetamine in ng/mL for the serum levels (Fig. 51A), and in ng/g for brain tissue
(Fig. 51B), following oral administration of L-lysine-d-amphetamine or d-
amphetamine sulfate (5mg/kg d-amphetamine base) to rats. Serum and brain tissue
d-amphetamine and L-lysine-d-amphetamine concentrations were measured by
LC/MS/MS (compound indicated in parenthesis).
[082] Figs. 52A-B. Plasma d-amphetamine and L-lysine-d-amphetamine levels
(52A, ng/mL; 52B, M) over a 72 hour period following oral administration of L-
lysinc-d-amphetamine (25 mg L-lysine-d-amphetamine rnesylate containing 7.37 mg
d-amphetamine base) to humans (LC/MS/MS analysis).
[083] Figs. 53A-B. Plasma d-amphetamine and L-lysine-d-amphetamine levels
(53A, ng/mL; 53B, M) over a 72 hour period following oral administration of L-
lysine-d-amphetamine (25 mg L-lysine-d-amphetamine mesylate containing 22.1 mg
d-amphetamine base) to humans (LC/MS/MS analysis).
[084] Figs. 54A-B. Plasma d-amphetamine levels (54A, 0-12 hours; 54B, 0-72
hours) following oral administration of L-lysine-d-amphetarnine (75 mg L-lysine-d-
amphetamine mesylate containing 22.1 mg d-amphetamine base) or Adderall XR®
(35 mg containing 21.9 mg amphetamine base to humans (LC/MS/MS analysis).
[085] Figs. 55A-B. Plasma d-amphetamine levels (55A, 0-12 hours; 55B, 0-72
hours) following oral administration of L-lysine-d-amphetamine (75 mg L-lysine-d-
amphetamine mesylate containing 22.1 mg d-amphetamine base) or Dexadrine

WO 2005/000334 PCT/US2004/017204
14
Spansule® (30 mg containing 22.1 mg amphetamine base) to humans (LC/MS/MS
analysis).
DETAILED DESCRIPTION OF THE INVENTION
[086] In accordance with the present invention and as used herein, the following
terms are defined with the following meanings, unless explicitly stated otherwise.
For additional methods of attaching amphetamine to carriers, see application number
U.S. 10/156,527, and/or PCT/US03/05524 and/or PCT/US03/05525 each of which
is hereby incorporated by reference in its entirety.
[087] The invention utilizes covalent modification of amphetamine to decrease its
potential for causing overdose or abuse. The amphetamine is covalently modified in
a manner that decreases its pharmacological activity, as compared to the unmodified
amphetamine, at doses above those considered therapeutic. When given at lower
doses, such as those intended for therapy, the covalently modified amphetamine
retains pharmacological activity similar to that of the unmodified amphetamine. The
covalent modification of amphetamine may comprise the attachment of any
chemical moiety through conventional chemistry.
[088] Compounds, compositions and methods of the invention provide reduced
potential for overdose, reduced potential for abuse or addiction, and/or improve
amphetamine's characteristics with regard to high toxicities or suboptimal release
profiles. Without wishing to be limited to the below theory, we believe that
overdose protection results from a natural gating mechanism at the site of hydrolysis
that limits the release of the active amphetamine from the prodrug at greater than
therapeutically prescribed amounts. Therefore, abuse resistance is provided by
limiting the "rush" or "high" available from the active amphetamine released by the
prodrug and limiting the effectiveness of alternative routes of administration.
Further, it is believed that the prodrug itself does not cross the blood brain barrier
and is thus substantially absent from the central nervous system.
[089] Throughout this application the use of "peptide" is meant to include a single
amino acid, a dipeptide, a tripeptide, an oligopeptide, a polypeptide, or the carrier
peptide. Oligopeptide is meant to include from 2 amino acids to 70 amino acids.

WO 2005/000334 PCT/US2004/017204
15
Further, at times the invention is described as being an active agent attached to an
amino acid, a dipeptide, a tripeptide, an oligopeptide, polypeptide or carrier peptide
to illustrate specific embodiments for the active agent conjugate. Preferred lengths
of the conjugates and other preferred embodiments are described herein.
[090] Throughout this application the use of "chemical moiety" is meant to include
at least amino acid(s), peptide(s), glycopeptide(s), carbohydrate(s), lipid(s),
nucleoside(s), or vitamin(s).
[091] Carbohydrates include sugars, starches, cellulose, and related compounds.
e.g., (CH2O)n, wherein n is an integer larger than 2 or Cn(H2O)n_1, with n larger than
5. More specific examples include, for instance, fructose, glucose, lactose, maltose,
sucrose, glyceraldehyde, dihydroxyacetone, erythrose, ribose, ribulose, xylulose,
galactose, mannose, sedoheptulose, neuraminic acid, dextrin, and glycogen.
[092] A glycoprotein is a carbohydrate (or glycan) covalently linked to protein.
The carbohydrate may be in the form of a monosaccharide, disaccharide(s),
oligosaccharide(s), polysaccharide(s), or their derivatives (e.g. sulfo- or phospho-
substituted).
[093] A glycopeptide is a carbohydrate linked to an oligopeptide composed of L-
and/or D-amino acids. A glyco-amino-acid is a saccharide attached to a single
amino acid by any kind of covalent bond. A glycosyl-amino- acid is a compound
consisting of saccharide linked through a glycosyl linkage (O-, N- or S-) to an amino
acid.
[094] A "composition" as used herein refers broadly to any composition containing
a described molecule conjugate(s). The composition may comprise a dry
formulation, an aqueous solution, or a sterile composition. Compositions
comprising the molecules described herein may be stored in freeze-dried form and
may be associated with a stabilizing agent such as a carbohydrate. In use, the
composition may be deployed in an aqueous solution containing salts, e.g., NaCl,
detergents, e.g., sodium dodecyl sulfate (SDS), and other components.
[095] "Amphetamine" shall mean any of the sympatho mimetic phenethylamine
derivatives which have central nervous system stimulant activity, such as but not

WO 2005/000334 PCT/US2004/017204
16
limited to, amphetamine, methamphetamine, p-methoxyamphetamine,
methylenedioxyamphetamine, 2,5-dimethoxy-4-methylamphetamine, 2,4,5-
trimethoxyamphetamine and 3,4-methylenedioxymethamphetamine.

Amphetamine Methylphenidate
[100] Other embodiments are described according to the following abbreviations.
Lys-Amp = L-lysine-d-amphetamine, Lys-Amph, Lysine-Amphetamine,
KAMP, K-amphetamine, or 2,6-diaminohexanoic acid-(l-methyl-2-
phenylethyl)-amide
Phe-Amp = Phenylalanine-Amphetamine, FAMP, or
2-amino-3-phenylpropanoicacid-(l-methyl-2-phenylethyl)-amide,
Ser-Amp = Serine-Amphetamine, SAMP, or
2-amino-3-hydroxylpropanoic acid-(l-methyl-2-phenylethyI)-amide,
Gly3-Amp = GGG-Amphetamine, GGGAMP, or
2-Amino-N-({ [(1 -methyl-2-phenyl-ethylcarbomyl)-rnethyl]-carbornyl} -
methyl) - acetamide
[101] This patent is meant to cover all compounds discussed regardless of absolute
configurations. Thus, natural, L-amino acids are discussed but the use of D-amino
acids are also included. Similarly, references to amphetamine should be interpreted
as inclusive of dextro- and levo-isomers.
[102] Furthermore, the following abbreviations may be used throughout the patent.
BOC = t-butyloxycarbonyl
CMC = carboxymethylcellulose
DIPEA = di-isopropyl ethyl amine
mp = melting point
NMR = nuclear magnetic resonance
OSu = hydroxysuccinimido ester

WO 2005/000334 PCT/US2004/017204
17
[103] "In a manner inconsistent with the manufacturer's instructions" is meant to
include but is not limited to consuming amounts greater than amounts described on
the label or ordered by a licensed physician, and/or altering by any means (e.g.
crushing, breaking, melting, separating etc.) the dosage formulation such that the
composition maybe injected, inhaled or smoked.
[104] Use of the phrases such as "decreased", "reduced", "diminished" or
"lowered" is meant to include at least a 10% change in pharmacological activity
with greater percentage changes being preferred for reduction in abuse potential and
overdose potential. For instance, the change may also be greater than 25%, 35%,
45%, 55%, 65%, 75%, 85%, 95%, 96%, 97%, 98%, 99%, or increments therein.
[105] For each of the recited embodiments, the amphetamine may be any of the
above discussed stimulants. In one embodiment, the amphetamine is
dextroamphetamine or methylphenidate.
[106] The attached chemical moiety may be any chemical substance that decreases
the pharmacological activity until amphetamine is released. Preferably the chemical
moiety is a single amino acid, dipeptide or tripeptide. Amphetamine binds to
specific sites to produce various effects (Hoebel, et al, 1989). The attachment of
certain chemical moieties can therefore diminish or prevent binding to these
biological target sites. Further, the covalent modification may prevent stimulant
activity by preventing the drug from crossing the blood-brain barrier. Preferably,
absorption of the composition into the brain is prevented or substantially diminished
and/or delayed when delivered by routes other than oral administration.
[107] The attached chemical moiety may further comprise naturally occurring or
synthetic substances. This includes, but is not limited to, the attachment of
amphetamine to amino acids, peptides, lipids, carbohydrates, glycopeptides, nucleic
acids or vitamins. These chemical moieties could be expected to affect delayed
release in the gastrointestinal tract and prevent rapid onset of the desired activity,
particularly when delivered by parenteral routes. (Hoebel, B. G., L. Hernandez, et
al., "Microdialysis studies of brain norepinephrine, serotonin, and dopamine release

WO 2005/000334 PCT/US2004/017204
18
during ingestive behavior. Theoretical and clinical implications." Ann N Y Acad Sci
575: 171-91) (1989).
[108] For each of the recited embodiments, the amino acid or peptide may
comprise of one or more of the naturally occurring (L-) amino acids: alanine,
arginine, asparagine, aspartic acid, cysteine, glycine, glutamic acid, glutamine,
histidine, isoleucine, leucine, lysine, methionine, proline, phenylalanine, serine,
tryptophan, threonine, tyrosine, and valine. In another embodiment, the amino acid
or peptide is comprised of one or more of the naturally occurring (D) amino acids:
alanine, arginine, asparagine, aspartic acid, cysteine, glycine, glutamic acid,
glutamine, histidine, isoleucine, leucine, lysine, methionine, proline, phenylalanine,
serine, tryptophan, threonine, tyrosine, and valine. In another embodiment, the
amino acid or peptide is comprised of one or more unnatural, non-standard or
synthetic amino acids such as, aminohexanoic acid, biphenylalanine,
cyclohexylalanine, cyclohexyl glycine, diethylglycine, dipropylglycine, 2,3-
diaminoproprionic acid, homophenylalanine, homoserine, homotyrosine,
naphthylalanine, norleucine, ornithine, pheylalanine(4-fluoro),
phenylalanine(2,3,4,5,6 pentafluoro), phenylalanine(4-nitro), phenylglycine,
pipecolic acid, sarcosine, tetrahydroisoquinoline-3-carboxylic acid, and tert-leucine.
In another embodiment, the amino acid or peptide comprises of one or more amino
acid alcohols, for example, serine and threonine. In another embodiment the amino
acid or peptide comprises of one or more N-methyl amino acids, for example, N-
methyl aspartic acid.
[109] In another embodiment, the specific carriers are utilized as a base short chain
amino acid sequence and additional amino acids are added to the terminus or side
chain. In another embodiment, the above amino acid sequence may have one more
of the amino acids substituted with one of the 20 naturally occurring amino acids. It
is preferred that the substitution be with an amino acid which is similar in structure
or charge compared to the amino acid in the sequence. For instance, isoleucine
(IIe)[I] is structurally very similar to leucine (Leu)[L], whereas, tyrosine (Tyr)[Y] is
similar to phenylalanine (Phe)[F], whereas serine (Ser)[S] is similar to threonine
(Thr)[T], whereas cysteine (Cys)[C] is similar to methionine (Met)[M], whereas

WO 2005/000334 PCT/US2004/017204
19
atanine (Ala)[A] is similar to valine (Val)[V], whereas lysine (Lys)[K] is similar to
arginine (Arg)[R], whereas asparagine (Asn)[N] is similar to glutamine (Gln)[Q],
whereas aspartic acid (Asp)[D] is similar to glutamic acid (Glu)[E], whereas
histidine (His)[H] is similar to proline (Pro)[P], and glycine (Gly)[G] is similar to
tryptophan (Trp)[W]. In the alternative, the preferred amino acid substitutions may
be selected according to hydrophilic properties (i.e., polarity) or other common
characteristics associated with the 20 essential amino acids. While preferred
embodiments utilize the 20 natural amino acids for their GRAS characteristics, it is
recognized that minor substitutions along the amino acid chain which do not effect
the essential characteristics of the amino acid chain are also contemplated.
[110] In one embodiment, the carrier range is between one to 12 chemical moieties
with one to 8 moieties being preferred. In another embodiment, the number of
chemical moieties is selected from 1, 2, 3, 4, 5, 6, or 7. In another embodiment, the
molecular weight of the carrier portion of the conjugate is below about 2,500, more
preferably below about 1,000, and most preferably below about 500 kD. In one
embodiment, the chemical moiety is a single lysine. In another embodiment, the
chemical moiety is a lysine bound to an additional chemical moiety.
[1ll] Another embodiment of the invention is a composition for preventing
overdose comprising amphetamine which has been covalently bound to a chemical
moiety.
[112] Another embodiment of the invention is a composition for safely delivering
amphetamine comprising a therapeutically effective amount of said amphetamine
which has been covalently bound to a chemical moiety wherein said chemical
moiety reduces the rate of absorption of the amphetamine as compared to delivering
the unbound amphetamine.
[113] Another embodiment of the invention is a composition for reducing
amphetamine toxicity comprising amphetamine which has been covalently bound to
a chemical moiety wherein said chemical moiety increases the rate of clearance
when given at doses exceeding those within the therapeutic range of said
amphetamine.

WO 2005/000334 PCT/US2004/017204
20
[114] Another embodiment of the invention is a composition for reducing
amphetamine toxicity comprising amphetamine which has been covalently bound to
a chemical moiety wherein said chemical moiety provides a serum release curve
which does not increase above amphetamine's toxicity level when given at doses
exceeding those within the therapeutic range of amphetamine.
[115] Another embodiment of the invention is a composition for reducing
bioavailability of amphetamine comprising amphetamine covalently bound to a
chemical moiety wherein said bound amphetamine maintains a steady-state serum
release curve which provides a therapeutically effective bioavailability but prevents
spiking or increased blood serum concentrations compared to unbound amphetamine
when given at doses exceeding those within the therapeutic range of amphetamine.
[116] Another embodiment of the invention is a composition for preventing a Cmax
spike for amphetamine when taken by mean other than orally while still providing a
therapeutically effective bioavailability curve if taken orally comprising an
amphetamine which has been covalently bound to a chemical moiety.
[117] Another embodiment of the invention is a composition for preventing a toxic
release profile in a patient comprising amphetamine covalently bound to a chemical
moiety wherein said bound amphetamine maintains a steady-state serum release
curve which provides a therapeutically effective bioavailability but prevents spiking
or increase blood serum concentrations compared to unbound amphetamine.
[118] Another embodiment of the invention is a compound of Formula I:
A-Xn-Zm
wherein A is an amphetamine as defined herein; X is a chemical moiety as defined
herein and n is between 1 and 50 and increments thereof; and Z is a further chemical
moiety different from X which acts as an adjuvant and m is between 1 and 50 and
increments thereof. In another embodiment, n is between 1 and 50, more preferably
between 1 and 10, and m is 0.
[119] Embodiments of the invention provide amphetamine compositions which
allow the amphetamine to be therapeutically effective when delivered at the proper

WO 2005/000334 PCT/US2004/017204
21
dosage but reduces the rate of absorption or extent of bioavailability of the
amphetamine when given at doses exceeding those within the therapeutic range of
amphetamine. Embodiments of the invention also provide amphetamine
compositions wherein the covalently bound chemical moiety increases the rate of
clearance of amphetamine when given at doses exceeding those within the
therapeutic range of the amphetamine.
[120] In another embodiment, the amphetamine compositions have substantially
lower toxicity compared to unbound amphetamine. In another embodiment, the
amphetamine compositions reduce or eliminate the possibility of overdose by oral
administration. In another embodiment, the amphetamine compositions reduce or
eliminate the possibility of overdose by intranasal administration. In another
embodiment, the amphetamine compositions reduce or eliminate the possibility of
overdose by injection. In another embodiment, the amphetamine compositions
reduce or eliminate the possibility of overdose by inhalation.
[121] In another embodiment, the amphetamine conjugates of the invention may
further comprise a polymer blend which comprises a hydrophilic polymer and/or a
water-insoluble polymer. The polymers may be used according to industry
standards to further enhance the sustained release/abuse resistant properties of the
amphetamine conjugate without reducing the abuse resistance. For instance, a
composition might include: about 70% to about 100% amphetamine conjugate by
weight, from about 0.01% to about 10% of a hydrophilic polymer (e.g.
hydroxypropyl methylcellulose), from about 0.01% to about 2.5% of a water-
insoluble polymer (e.g. acrylic resin), from about 0,01% to about 1.5% of additives
(e.g. magnesium stearate), and from about 0.01% to about 1% colorant by weight.
[122] Hydrophilic polymers suitable for use in the sustained release formulations
include one or more natural or partially or totally synthetic hydrophilic gums such as
acacia, gum tragacanth, locust bean gum, guar gum, or karaya gum, modified
cellulosic substances such as methylcellulose, hydroxomethylcellulose,
hydroxypropyl methylcellulose, hydroxypropyl cellulose, hydroxyethylcellulose,
carboxymethylcellulose; proteinaceous substances such as agar, pectin, carrageen,

WO 2005/000334 PCT/US2004/017204
22
and alginates; and other hydrophilic polymers such as carboxypolymethylene,
gelatin, casein, zein, bentonite, magnesium aluminum silicate, polysaccharides,
modified starch derivatives, and other hydrophilic polymers known to those of skill
in the art, or a combination of such polymers. These hydrophilic polymers gel and
would dissolve slowly in aqueous acidic media thereby allowing the amphetamine
conjugate to diffuse from the gel in the stomach. When the gel reaches the intestines
it would dissolve in controlled quantities in the higher pH medium to allow further
sustained release. Preferred hydrophilic polymers are the hydroxypropyl
methylcelluloses such as those manufactured by The Dow Chemical Company and
known as Methocel ethers, such as Methocel E10M.
[123] Other formulations may further comprise pharmaceutical additives including,
but not limited to: lubricants such as magnesium stearate, calcium stearate, zinc
stearate, powdered stearic acid, hydrogenated vegetable oils, talc, polyethylene
glycol, and mineral oil; colorants such as Emerald Green Lake, FD&C Red No. 40,
FD&C Yellow No. 6, D&C Yellow No. 10, or FD&C Blue No. 1 and other various
certified color additives (See 21 CFR, Part 74); binders such as sucrose, lactose,
gelatin, starch paste, acacia, tragacanth, povidone polyethylene glycol, Pullulan and
corn syrup; glidants such as colloidal silicon dioxide and talc; surface active agents
such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate, triethanolamine,
polyoxyethylene sorbitan, poloxalkol, and quarternary ammonium salts;
preservatives and stabilizers; excipients such as lactose, mannitol, glucose, fructose,
xylose, galactose, sucrose, maltose, xylitol, sorbitol, chloride, sulfate and phosphate
salts of potassium, sodium, and magnesium; and/or any other pharmaceutical
additives known to those of skill in the art. In one preferred embodiment, a
sustained release formulation further comprises magnesium stearate and Emerald
Green Lake.
[124] An amphetamine conjugate, which is further formulated with excipients, may
be manufactured according to any appropriate method known to those of skill in the
art of pharmaceutical manufacture. For instance, the amphetamine-conjugate and a
hydrophilic polymer may be mixed in a mixer with an aliquot of water to form a wet
granulation. The granulation may be dried to obtain hydrophilic polymer

WO 2005/000334 PCT/US2004/017204
23
encapsulated granules of amphetamine-conjugate. The resulting granulation may be
milled, screened, then blended with various pharmaceutical additives such as, water
insoluble polymers, and/or additional hydrophilic polymers. The formulation may
then tableted and may further be film coated with a protective coating which rapidly
dissolves or disperses in gastric juices.
[125] However, it should be noted that the amphetamine conjugate controls the
release of amphetamine into the digestive tract over an extended period of time
resulting in an improved profile when compared to immediate release combinations
and prevention of abuse without the addition of the above additives. In a preferred
embodiment, no further sustained release additives are required to achieve a blunted
or reduced pharmacokinetic curve (e.g., reduced euphoric effect) while achieving
therapeutically effective amounts of amphetamine release when taken orally.
[126] The compounds of the invention can be administered by a variety of dosage
forms. Any biologically-acceptable dosage form known to persons of ordinary skill
in the art, and combinations thereof, are contemplated. Examples of preferred
dosage forms include, without limitation, chewable tablets, quick dissolve tablets,
effervescent tablets, reconstitutable powders, elixirs, liquids, solutions, suspensions,
emulsions, tablets, multi-layer tablets, bi-layer tablets, capsules, soft gelatin
capsules, hard gelatin capsules, caplets, lozenges, chewable lozenges, beads,
powders, granules, particles, microparticles, dispersible granules, cachets and
combinations thereof.
[127] The most effective means for delivering the abuse-resistant compounds of
the invention is orally, to permit maximum release of the amphetamine, and provide
therapeutic effectiveness and/or sustained release while maintaining abuse
resistance. When delivered by oral route the amphetamine is released into
circulation, preferably over an extended period of time as compared to amphetamine
alone.
[128] Formulations of the invention suitable for oral administration can be
presented as discrete units, such as capsules, caplets or tablets. These oral
formulations also can comprise a solution or a suspension in an aqueous liquid or a

WO 2005/000334 PCT/US2004/017204
24
non-aqueous liquid. The formulation can be an emulsion, such as an oil-in-water
liquid emulsion or a water-in-oil liquid emulsion. The oils can be administered by
adding the purified and sterilized liquids to a prepared enteral formula, which is then
placed in the feeding tube of a patient who is unable to swallow.
[129] Soft gel or soft gelatin capsules may be prepared, for example by dispersing
the formulation in an appropriate vehicle (vegetable oils are commonly used) to
form a high viscosity mixture. This mixture is then encapsulated with a gelatin
based film using technology and machinery known to those in the soft gel industry.
The industrial units so formed are then dried to constant weight.
[130] Chewable tablets, for example may be prepared by mixing the formulations
with excipients designed to form a relatively soft, flavored, tablet dosage form that is
intended to be chewed rather than swallowed. Conventional tablet machinery and
procedures, that is both direct compression and granulation, i.e., or slugging, before
compression, can be utilized. Those individuals involved in pharmaceutical solid
dosage form production are versed in the processes and the machinery used as the
chewable dosage form is a very common dosage form in the pharmaceutical
industry.
[131] Film-coated tablets, for example may be prepared by coating tablets using
techniques such as rotating pan coating methods or air suspension methods to
deposit a contiguous film layer on a tablet.
[132] Compressed tablets, for example may be prepared by mixing the formulation
with excipients intended to add binding qualities to disintegration qualities. The
mixture is either directly compressed or granulated then compressed using methods
and machinery known to those in the industry. The resultant compressed tablet
dosage units are then packaged according to market need, i.e., unit dose, rolls, bulk
bottles, blister packs, etc.
[133] The invention also contemplates the use of biologically-acceptable carriers
which may be prepared from a wide range of materials. Without being limited
thereto, such materials include diluents, binders and adhesives, lubricants,
plasticizers, disintegrants, colorants, bulking substances, flavorings, sweeteners and

WO 2005/000334 PCT/US2004/017204
25
miscellaneous materials such as buffers and adsorbents in order to prepare a
particular medicated composition.
[134] Binders may be selected from a wide range of materials such as
hydroxypropylmethylcellulose, ethylcellulose, or other suitable cellulose derivatives,
povidone, acrylic and methacrylic acid co-polymers, pharmaceutical glaze, gums,
milk derivatives, such as whey, starches, and derivatives, as well as other
conventional binders known to persons skilled in the art. Exemplary non-limiting
solvents are water, ethanol, isopropyl alcohol, methylene chloride or mixtures and
combinations thereof. Exemplary non-limiting bulking substances include sugar,
lactose, gelatin, starch, and silicon dioxide.
[135] Preferred plasticizers may be selected from the group consisting of diethyl
phthalate, diethyl sebacate, triethyl citrate, cronotic acid, propylene glycol, butyl
phthalate, dibutyl sebacate, castor oil and mixtures thereof, without limitation. As is
evident, the plasticizers may be hydrophobic as well as hydrophilic in nature.
Water-insoluble hydrophobic substances, such as diethyl phthalate, diethyl sebacate
and castor oil are used to delay the release of water-soluble vitamins, such as
vitamin B6 and vitamin C. In contrast, hydrophilic plasticizers are used when water-
insoluble vitamins are employed which aid in dissolving the encapsulated film,
making channels in the surface, which aid in nutritional composition release.
[136] It should be understood that in addition to the ingredients particularly
mentioned above, the formulations of this invention can include other suitable
agents such as flavoring agents, preservatives and antioxidants. Such antioxidants
would be food acceptable and could include vitamin E, carotene, BHT or other
antioxidants known to those of skill in the art.
[137] Other compounds which may be included by admixture are, for example,
medically inert ingredients, e.g., solid and liquid diluent, such as lactose, dextrose,
saccharose, cellulose, starch or calcium phosphate for tablets or capsules, olive oil or
ethyl oleate for soft capsules and water or vegetable oil for suspensions or
emulsions; lubricating agents such as silica, talc, stearic acid, magnesium or calcium
stearate and/or polyethylene glycols; gelling agents such as colloidal days;

WO 2005/000334 PCT/US2004/017204
26
thickening agents such as gum tragacanth or sodium alginate, binding agents such as
starches, arabic gums, gelatin, methylcellulose, carboxymethylcellulose or
polyvinylpyrrolidone; disintegrating agents such as starch, alginic acid, alginates or
sodium starch glycolate; effervescing mixtures; dyestuff; sweeteners; wetting agents
such as lecithin, polysorbates or laurylsulphates; and other therapeutically
acceptable accessory ingredients, such as humectants, preservatives, buffers and
antioxidants, which are known additives for such formulations.
[138] For oral administration, fine powders or granules containing diluting,
dispersing and/or surface-active agents may be presented in a draught, in water or a
syrup, in capsules or sachets in the dry state, in a non-aqueous suspension wherein
suspending agents may be included, or in a suspension in water or a syrup. Where
desirable or necessary, flavoring, preserving, suspending, thickening or emulsifying
agents can be included.
[139] Liquid dispersions for oral administration may be syrups, emulsions or
suspensions. The syrups may contain as carrier, for example, saccharose or
saccharose with glycerol and/or mannitol and/or sorbitol. The suspensions and the
emulsions may contain a carrier, for example a natural gum, agar, sodium alginate,
pectin, methylcellulose, carboxymethylcellulose or polyvinyl alcohol.
[140] The dose range for adult human beings will depend on a number of factors
including the age, weight and condition of the patient. Tablets and other forms of
presentation provided in discrete units conveniently contain a daily dose, or an
appropriate fraction thereof, of one or more of the compounds of the invention. For
example, units may contain from 5 mg to 500 mg, but more usually from 10 mg to
250 mg, of one or more of the compounds of the invention.
[141] It is also possible for the dosage form to combine any forms of release
known to persons of ordinary skill in the art. These include immediate release,
extended release, pulse release, variable release, controlled release, timed release,
sustained release, delayed release, long acting, and combinations thereof. The
ability to obtain immediate release, extended release, pulse release, variable release,

WO 2005/000334 PCT/US2004/017204
27
controlled release, timed release, sustained release, delayed release, long acting
characteristics and combinations thereof is known in the art.
[142] Compositions of the invention may be administered in a partial, i.e.,
fractional dose, one or more times during a 24 hour period, a single dose during a 24
hour period of time, a double dose during a 24 hour period of time, or more than a
double dose during a 24 hour period of time. Fractional, double or other multiple
doses may be taken simultaneously or at different times during the 24 hour period.
The doses may be uneven doses with regard to one another or with regard to the
individual components at different administration times.
[143] Likewise, the compositions of the invention may be provided in a blister
pack or other such pharmaceutical package. Further, the compositions of the present
inventive subject matter may further include or be accompanied by indicia allowing
individuals to identify the compositions as products for a prescribed treatment. The
indicia may additionally include an indication of the above specified time periods
for administering the compositions. For example, the indicia may be time indicia
indicating a specific or general time of day for administration of the composition, or
the indicia may be a day indicia indicating a day of the week for administration of
the composition. The blister pack or other combination package may also include a
second pharmaceutical product.
[144] It will be appreciated that the pharmacological activity of the compositions
of the invention can be demonstrated using standard pharmacological models that
are known in the art. Furthermore, it will be appreciated that the inventive
compositions can be incorporated or encapsulated in a suitable polymer matrix or
membrane for site-specific delivery, or can be functionalized with specific targeting
agents capable of effecting site specific delivery. These techniques, as well as other
drug delivery techniques, are well known in the art.
[145] In another embodiment of the invention, the solubility and dissolution rate of
the composition is substantially changed under physiological conditions encountered
in the intestine, at mucosal surfaces, or in the bloodstream. In another embodiment
the solubility and dissolution rate substantially decrease the bioavailability of the

WO 2005/000334 PCT/US2004/017204
28
amphetamine, particularly at doses above those intended for therapy. In another
embodiment, the decrease in bioavailability occurs upon intranasal administration.
In another embodiment, the decrease in bioavailability occurs upon intravenous
administration.
[146] For each of the described embodiments, one or more of the following
characteristics may be realized: The toxicity of the amphetamine conjugate is
substantially lower than that of the unbound amphetamine. The covalently bound
chemical moiety reduces or eliminates the possibility of overdose by oral
administration. The covalently bound chemical moiety reduces or eliminates the
possibility of overdose or abuse by intranasal administration. The covalently bound
chemical moiety reduces or eliminates the possibility of overdose or abuse by
injection.
[147] The invention further provides methods for altering amphetamines in a
manner that decreases their potential for abuse. Methods of the invention provide
various ways to regulate pharmaceutical dosage through covalent attachment of
amphetamine to different chemical moieties. One embodiment provides a method of
preventing overdose comprising administering to an individual amphetamine which
has been covalently bound to a chemical moiety.
[148] Another embodiment provides a method of safely delivering amphetamine
comprising providing a therapeutically effective amount of a amphetamine which
has been covalently bound to a chemical moiety wherein the chemical moiety
reduces the rate of absorption of amphetamine as compared to delivering the
unbound amphetamine.
[149] Another embodiment provides a method of reducing amphetamine toxicity
comprising providing a patient with amphetamine which has been covalently bound
to a chemical moiety, wherein the chemical moiety increases the rate of clearance of
pharmacologically active amphetamine (i.e., released amphetamine) when given at
doses exceeding those within the therapeutic range of amphetamine.
[150] Another embodiment provides a method of reducing amphetamine toxicity
comprising providing a patient with amphetamine which has been covalently bound

WO 2005/000334 PCT/US2004/017204
29
to a chemical moiety, wherein the chemical moiety provides a serum release curve
which does not increase above the amphetamine's toxicity level when given at doses
exceeding those within the therapeutic range for the unbound amphetamine.
[151] Another embodiment provides a method of reducing bioavailability of
amphetamine comprising providing amphetamine covalently bound to a chemical
moiety, wherein the bound amphetamine maintains a steady-state serum release
curve which provides a therapeutically effective bioavailability but prevents spiking
or increase blood serum concentrations compared to unbound amphetamine when
given at doses exceeding those within the therapeutic range for the unbound
amphetamine.
[152] Another embodiment provides a method of preventing a Cmax spike for
amphetamine while still providing a therapeutically effective bioavailability curve
comprising providing amphetamine which has been covalently bound to a chemical
moiety.
[153] In another embodiment, methods of the invention provide bioavailability
curves similar to those of Figures 6-55.
[154] Another embodiment provides a method for preventing a toxic release profile
in a patient comprising administering to a patient amphetamine covalently bound to
a chemical moiety, wherein said bound amphetamine maintains a steady-state serum
release curve which provides a therapeutically effective bioavailability but prevents
spiking or increase blood serum concentrations compared to unbound amphetamine,
particularly when taken at doses above prescribed amounts.
[155] Another embodiment of the invention is a method for reducing or preventing
abuse of amphetamine comprising providing, administering, or prescribing said
composition to a human in need thereof, wherein said composition comprises a
chemical moiety covalently attached to amphetamine such that the pharmacological
activity of amphetamine is decreased when the composition is used in a manner
inconsistent with the manufacturer's instructions.

WO 2005/000334 PCT/US2004/017204
30
[156] Another embodiment of the invention is a method for reducing or preventing
abuse of amphetamine comprising consuming an amphetamine conjugate of the
invention, wherein said conjugate comprises a chemical moicty covalently attached
to amphetamine such that the pharmacological activity of amphetamine is
substantially decreased when the composition is used in a manner inconsistent with
the manufacturer's instructions.
[157] Another embodiment of the invention is a method of preventing overdose of
amphetamine comprising providing, administering, or prescribing an amphetamine
composition of the invention to a human in need thereof, wherein said composition
comprises a chemical moiety covalently attached to amphetamine in a manner that
decreases the potential of overdose from amphetamine.
[158] Another embodiment of the invention is a method of preventing overdose of
amphetamine, comprising consuming an amphetamine composition of the invention,
wherein said composition comprises a chemical moiety covalently attached to
amphetamine in a manner that decreases the potential of overdose from
amphetamine.
[159] Another embodiment of the invention is a method for reducing or preventing
the euphoric effect of amphetamine comprising providing, administering, or
prescribing said to a human in need thereof, a composition comprising a chemical
moiety covalently attached to amphetamine such that the pharmacological activity of
amphetamine is decreased when the composition is used in a manner inconsistent
with the manufacturer's instructions.
[160] Another embodiment of the invention is a method for reducing or preventing
the euphoric effect of amphetamine, comprising consuming a said composition
comprising a chemical moiety covalently attached to amphetamine such that the
pharmacological activity of amphetamine is decreased when the composition is used
in a manner inconsistent with the manufacturer's instructions.
[161] Another embodiment of the invention is any of the preceding methods
wherein said amphetamine composition is adapted for oral administration, and
wherein said amphetamine is resistant to release from said chemical moiety when

WO 2005/000334 PCT/US2004/017204
31
the composition is administered parenterally, such as intranasally or intravenously.
Preferably, said amphetamine may be released from said chemical moiety in the
presence of acid and/or enzymes present in the stomach, intestinal tract, or blood
serum. Optionally, said composition may be in the form of a tablet, capsule, oral
solution, oral suspension, or other oral dosage form discussed herein,
[162] For each of the recited methods, the chemical moiety may be one or more
amino acid(s), oligopeptide(s), polypeptide(s), carbohydrate(s), glycopeptide(s),
nucleic acid(s), or vitamin(s). Preferably, said chemical moiety is an amino acid,
oligopeptide, or polypeptide or carbohydrate. Where the chemical moiety is a
polypeptide, preferably said polypeptide comprises fewer than 70 amino acids,
fewer than 50 amino acids, fewer than 10 amino acids, or fewer than 4 amino acids.
Where the chemical moiety is an amino acid, preferably said amino acid is lysine,
serine, phenyialanine or glycine. Most preferably, said amino acid is lysine.
[163] For each of the recited embodiments, covalent attachment may comprise an
ester or carbonate bond.
[164] For each of the recited methods, the composition may yield a therapeutic
effect without substantial euphoria. Preferably, said amphetamine composition
provides a therapeutic ally bioequivalent AUC when compared to amphetamine
alone but does provide a Cmax which results in euphoria.
[165] Another embodiment of the invention is a method for reducing or preventing
abuse of amphetamine comprising orally administering an amphetamine
composition of the invention to a human in need thereof, wherein said composition
comprises an amino acid or peptide (e.g., lysine) covalently attached to
amphetamine such that the pharmacological activity of amphetamine is decreased
when the composition is used in a manner inconsistent with the manufacturer's
instructions.
[166] Another embodiment is a method of preventing overdose of a amphetamine
comprising orally administering an amphetamine composition to a human in need
thereof, wherein said composition comprises an amino acid or peptide (e.g., lysine)

WO 2005/000334 PCT/US2004/017204
32
covalently attached to amphetamine in a manner that decreases the potential of
amphetamine to result in overdose.
[167] Another embodiment is a method for reducing or preventing the euphoric
effect of amphetamine comprising orally administering an amphetamine
composition to a human in need thereof, wherein said composition comprises an
amino acid or peptide (e.g., Iysine) covalently attached to amphetamine such that the
pharmacological activity of amphetamine is decreased when the composition is used
in a manner inconsistent with the manufacturer's instructions.
[168] For each of the recited methods of the invention the following properties
may be achieved through bonding amphetamine to the chemical moiety. In one
embodiment, the toxicity of the compound may be lower than that of the
amphetamine when amphetamine is delivered in its unbound state or as a salt
thereof. In another embodiment, the possibility of overdose by oral administration is
reduced or eliminated. In another embodiment, the possibility of overdose by
intranasal administration is reduced or eliminated. In another embodiment, the
possibility of overdose by injection administration is reduced or eliminated.
[169] Another embodiment of the invention provides methods of treating various
diseases or conditions comprising administering compounds or compositions of the
invention which further comprise commonly prescribed active agents for the
respective illness or diseases wherein the amphetamine is covalently attached to a
chemical moiety. For instance, one embodiment of the invention comprises a
method of treating attention deficit hyperactivity disorder (ADHD) comprising
administering to a patient amphetamine covalently bound to a chemical moiety.
Another embodiment provides a method of treating attention deficit disorder (ADD)
comprising administering to a patient compounds or compositions of the invention,
amphetamine covalently bound to a chemical moiety.
[170] Another embodiment of the invention provides a method of treating
narcolepsy comprising administering to a patient compounds or compositions of the
invention.

WO 2005/000334 PCT/US2004/017204
33
[171] In order to facilitate a more complete understanding of the invention,
Examples are provided below. However, the scope of the invention is not limited to
specific embodiments disclosed in these Examples, which are for purposes of
illustration only.
Examples
Example 1. General synthesis of amino acid-amphetamine conjugates,
[172] Amino acid conjugates were synthesized by the general method described in
Figs. 1-5.
Example 2. Synthesis of L-lysine-J-amphetamine.
[173] L-lysine-^-amphetamine was synthesized (see Fig. 2) by the following
method:
a. Coupling

Reagents MW Weight mmoles Molar Equivalents
d-amphetamine freebase 135.2 4-75 g 35.13 1
Boc-Lys(Boc)-OSu 443.5 15.58 g 35.13 1
Di-iPr-Et-Amine 129 906 mg 7.03 0.2, d=0.74, 1.22 mL
1,4-Dioxane 100 mL - -
[174] To a solution of Boc-Lys(Boc)-OSu (15.58 g, 35.13 mmol) in dioxane (100
mL) under an inert atmosphere was added d-amphetamine freebase (4.75 g, 35.13
mmol) and DiPEA (0.9 g, 1.22 mL, 7.03 mmol). The resulting mixture was allowed
to stir at room temperature overnight. Solvent and excess base were then removed
using reduced pressure evaporation. The crude product was dissolved in ethyl
acetate and loaded on to a flash column (7 cm wide, filled to 24 cm with silica) and
eluted with ethyl acetate. The product was isolated; the solvent reduced by rotary
evaporation and the purified protected amide was dried by high-vac to obtain a white
solid. 'H NMR (DMSO-d6)  1.02-1.11 (m, 2H, Lys 7-CH2),  1.04 (d, 3H, Amp
0C-CH3),  1.22-1.43 (m, 4H, Lys- and -CH2),  1.37 (18H, Boc, 6x CH3),  2.60-
2.72 (2H, Amp CH2),  3.75-3.83, (m, 1H, Lys -H)  3.9-4.1 (m, 1H, Amp -H), 

WO 2005/000334 PCT/US2004/017204
34
6.54-6.61 (d, 1H, amide NH),  6.7-6.77 (m, 1H, amide NH),  7.12-7.29 (m, 5H,
ArH),  7.65-7.71 (m, 1, amide NH); mp = 86-88 °C.

b. Deprotection
Reagents MW Weight mmoles Molar Equivalents
4M HC1 in dioxane 4 mmoI/mL 50 mL 200 6.25
B oc-Lys(B oc)-Amp 463.6 14.84 g 32 1
1,4-Dioxane - 50 mL - -
[175] The protected amide was dissolved in 50 mL of anhydrous dioxane and
stirred while 50 mL (200 mmol) of 4M HCl/dioxane was added and stirred at room
temperature overnight. The solvents were then reduced by rotary evaporation to
afford a viscous oil. Addition of 100 mL MeOH followed by rotary evaporation
resulted in a golden colored solid material that was further dried by storage at room
temperature under high vacuum. !H NMR (DMSO-d6)  0.86-1.16 (m, 2H, Lys -
CH2),  1.1 (d, 3H, Amp -CH3),  1.404.56 (m, 4H, Lys- and -CH2),  2.54-
2.78 (m, 2H, Amp CH2, 2H, Lys e-CH2), 3.63-3.74 (m, 1H, Lys -H),  4.00-4.08
(m, 1H, Amp -H),  7.12-7.31 (m, 5H, Amp ArH),  8.13-8.33 (d, 3H, Lys amine)
 8.70-8.78 (d, IH, amide NH); mp = 120-122 °C.
Example 3. Synthesis of Ser-Amp.
[176] Ser-Amp was synthesized by a similar method (see Fig. 3) except the amino
acid starting material was Boc-Ser(0-tBu)-OSu and the deprotection was done using
a solution of trifluoroacetic acid instead of HC1.
Example 4. Synthesis of Phe-Amp,
[177] Phe-Amp was synthesized by a similar method (see Fig. 4) except the amino
acid starting material was Boc-Phe-OSu.
Example 5. Synthesis of Gly3-Amp.
[178] Gly3-Amp was synthesized by a similar method (see Fig. 5) except the amino
acid starting material was Boc-GGG-OSu.

WO 2005/000334 PCT/US2004/017204
35
Example 6. Pharmacokinetics of L-lvsine-d-amphetamine compared to d-
amphetamine sulfate (ELISA Analysis)
[179] Male Sprague-Dawley rats were provided water ad libitum, fasted overnight
and dosed by oral gavage L-lysine-d-amphetamine or d-amphetamine sulfate. In all
studies doses contained equivalent amounts of d-amphetamine base. Plasma d-
amphetamine concentrations were measured by ELISA (Amphetamine Ultra,
109319, Neogen, Corporation, Lexington, KY). The assay is specific for d-
amphetamine with only minimal reactivity (0.6%) of the major d-amphetamine
metabolite (para-hydroxy-d-amphetamine) occurring. L-Iysine-d-amphetamine was
also determined to be essentially unreactive in the ELISA ( [180] Mean (n=4) plasma concentration curves of d-amphetarnine or L-lysine-d-
amphetamine are shown in Fig. 6. Extended release was observed in all four L-
lysine-d-amphetamine dosed animals and Cmax was substantially decreased as
compared to animals dosed with d-amphetamine sulfate. Plasma d-amphetamine
concentrations of individual animals for d-amphetamine or L-lysine-d-amphetamine
are shown in Table 1, The mean plasma d-amphetamine concentrations are shown
in Table 2. The time to peak concentration for L-lysine-d-amphetamine was similar
to that of d-amphetamine. Pharmacokinetic parameters for oral administration of d-
amphetamine or L-lysine-d-amphetamine are summarized in Table 3.
Table 1. Plasma Concentrations of d-amphetamine from Individual Animals Orally
Administered d-amphetamine or L-lysine-d-amphetamine (3 mg/kg d-amphetamine
base).

Time damphetamine (ng/ml) L-lysine-damphetamine (ng/ml)
(hours) Rat#1 Rat #2 Rat #3 Rat #4 Rat#1 Rat #2 Rat #3 Rat #4
0.5 144 157 101 . 115 52 62 74 44
1 152 78 115 78 48 72 79 57
1.5 85 97 117 95 42 62 76 53
3 34 45 72 38 61 60 71 43
5 20 14 12 15 49 33 44 22
8 3 3 2 2 15 14 12 8

WO 2005/000334 PCT/US2004/017204
36
Table 2. Mean Plasma Concentrations of d-amphetamine Following Oral
Administration of d-amphetamine or L-Iysine-d-amphetamine.

Hours Plasma damphetamine Concentrations (ng/m!)

damphetamine L-lysine- damphetamine

Mean +/-SD CV Mean +/-SD CV
0.5 129 25 20 58 13 22
1 106 35 33 64 14 22
1.5 99 13 14 58 14 25
3 47 17 36 59 11 19
5 15 4 24 37 12 32
8 2 1 35 12 3 24
Table 3. Pharmacokinetic Parameters of d-amphetamine Following Oral
Administration of d-amphetamine or L-lysine-d-amphetamine.

Drug AUC (0-8 h)
ng/ml h Percent
Amphetamine Cmax
(ng/ml) Percent
Amphetamine Mean Peak
(ng/ml) Percent
Amphetamine
Amphetamine 341 +/- 35 100 111+/- 27 100 129 100
Lys-Amp 333 +/- 66 98 61 +/-13 55 64 50
[181] Example 6 illustrates that when lysine is conjugated to the active agent
amphetamine the peak levels of amphetamine are decreased while bioavailability is
maintained approximately equal to amphetamine. The bioavailability of
amphetamine released from L-lysine-d-amphetamine is similar to that of
amphetamine sulfate at the equivalent dose, thus L-lysine-d-amphetamine maintains
its therapeutic value. The gradual release of amphetamine from L-lysine-d-
amphetamine and decrease in peak levels reduce the possibility of overdose.
Example 7: Oral bioavailability of L-lysine-d-amphetamine at various doses
approximating a range of therapeutic human doses
[182] Mean (n=4) plasma concentration curves of d-amphetamine vs. L-lysine-d-
amphetamine are shown for rats orally administered 1.5, 3, and 6 mg/kg in Figs. 7, 8
and 9, respectively. Extended release was observed at all three doses for L-lysine-d-
amphetamine dosed animals. The mean plasma concentrations for 1.5, 3, and 6

WO 2005/000334 PCT/US2004/017204
37
mg/kg are shown in Tables 4, 5 and 6, respectively. Pharmacokinetic parameters for
oral administration of d-amphetamine vs. L-lysine-d-amphetamine at the various
doses are summarized in Table 7.
Table 4. Mean Plasma Concentrations of d-amphetamine vs. L-lysine-d-
amphetamine Following Oral Admistration (1.5 mg/kg)

Hours Plasma Amphetamine Concentrations (ng/ml)

damphetamine L-lysine-d-amphetamine

Mean +/-SD CV Mean +/-SD CV
0 0 0 0 0 0 0
0.25 103 22 21 31 11 37
0.5 126 20 16 51 23 45
1 101 27 27 68 23 34
1.5 116 28 24 72 10 14
3 66 13 20 91 5 5
5 40 7 18 75 16 22
8 17 2 15 39 13 34
Table 5. Mean Plasma Concentrations of d-amphetamine vs. L-lysine-d-
amphetamine Following Oral Admistration (3 mg/kg)

Hours Plasma Amphetamine Concentrations (ng/ml)

damphetamine L-lysine-d-amphetamine

Mean +/-SD CV Mean +/-SD CV
0 0 0
0.25 96 41 43 51 49 97
0.5 107 49 46 36 35 96
1 121 17 14 81 44 54
1.5 120 33 27 97 32 33
3 91 30 33 88 13 15
5 62 22 36 91 21 23
8 19 6 33 46 16 34

WO 2005/000334 PCT/US2004/017204
38
Table 6. Mean Plasma Concentrations of d-amphetamine vs. L-lysine-d-
amphetamine Following Oral Admistration (6 mg/kg).

Hours Plasma Amphetamine Concentrations (ng/ml)

damphetamine L-lysine-d-amphetamine

Mean +/-SD CV Mean +/-SD CV
0 0 0
0.25 204 14 7 74 38 51
0.5 186 9 5 106 39 37
1 167 12 7 133 33 24
1.5 161 24 15 152 22 15
3 111 29 26 157 15 10
5 78 9 11 134 18 13
8 35 5 15 79 12 15

Table 7. Pharmacokinetic Parameters of d-amphetamine Following Oral Administration of d-amphetamine or L-lysine-^-amphetamine.

Parameter 1.5 mg/kg 3 mg/kg 6 mg/kg

damphetamine L-lysine-d
amphetamine damphetamine L-lysine-d
amphetamine damphetamine L-lysine-d
amphetamine
AUC (ng/ml
h) 481 538 587 614 807 1005
Percent 100 112 100 105 100 125
Q/nax
(ng/ml) 133 93 587 614 807 1005
Percent 100 70 100 105 100 125
(hours) 0.938 3.5 1 1.56 0.563 2.625
Percent 100 373 100 156 100 466

WO 2005/000334 PCT/US2004/017204

WO 2005/000334 PCT/US2004/017204
40
Example 8: Oral bioavailability of L-lysine-d-amphetamine at various doses
approximating a range of therapeutic human doses compared to a
suprapharmacological dose
[183] Male Sprague-Dawley rats were provided water ad libitum, fasted overnight
and dosed by oral gavage with 1.5, 3,6, 12, and 60 mg/kg of amphetamine sulfate or
L-lysine-d-amphetamine containing the equivalent amounts of d-amphetamine.
Concentrations of d-amphetamine were measured by ELISA.
[184] It has been demonstrated that when lysine is conjugated to the active agent d-
amphetamine the levels of d-amphetamine at 30 minutes post-administration are
decreased by approximately 50% over a dose range of 1.5 to 12 mg/kg. However,
when a suprapharmcological dose (60 mg/kg) is given the levels of d-amphetamine
from L-lysine-d-amphetamine only reached 8% of those seen for d-amphetamine
sulfate (Tables 8 and 9, Fig. 10). The substantial decrease in oral bioavailability at a
high dose greatly reduces the abuse potential of L-lysine-d-amphetamine.
Table 8. Levels of d-amphetamine vs. Dosage at 0.5 h Post Dosing with d-
amphetamine Sulfate.

Dose mg/kg 1.5 3 6 12 60
ng/ml 0.5 h 109+/-59 196 +/- 72 294 +/- 202 344 +/- 126 3239 +/- 73
Percent 100 100 100 100 100
Table 9. Levels of d-amphetamine vs. Dosage at 0.5 h Post Dosing with L-lysine-d-
amphetamine.

Dose mg/kg 1.5 3 6 12 60
ng/ml 0.5 h 45 +A 10 86 +/- 26 129+/- 46 172+/- 113 266 +/- 18
Percent 41 44 44 50 8
Example 9: Decreased oral bioavailability of L-lvsine-d-amphetamine at a high dose
[185] An additional oral PK study illustrated in Fig. 11 shows the d-amphetamine
blood levels of a 60 mg/kg dose over an 8 h time course. In the case of d-
amphetamine blood levels quickly reached a very high level and 8 of 12 animals
either died or were sacrificed due to acute symptoms of toxicity. Blood levels

WO 2005/000334 PCT/US2004/017204
41
(Tables 10-11) of animals administered L-lysine-d-amphetamine, on the other hand,
did not peak until 5 hours and reached only a fraction of the levels of the animals
receiving amphetamine (note: valid data past 3 h for d-amphetamine could not be
determined due to death and sacrifice of animals).
Table 10. Mean Plasma Concentrations of d-amphetamine vs. L-lysine-d-
amphetamine Following Oral Administration of a High Dose (60 mg/kg).

Hours Plasma Amphetamine Concentrations (ng/ml)

damphetamine L-lysine-d-amphetamine

Mean +/-SD CV Mean +/-SD CV
0 NA NA NA NA NA NA
0.25 2174 907 42 35 17 48
0.5 2643 578 22 81 33 41
1 2828 1319 47 212 30 14
1.5 2973 863 29 200 79 40
3 2944 95 3 440 133 30
5 NA NA NA 565 100 18
8 NA NA NA 410 206 50
Table 11. Pharmacokinetic Parameters of d amphetamine vs. L-lysine-d-
amphetamine

Drug AUC
ng/ml h Percent
damphetamine Cmax
(ng/ml) Percent
damphetamine Mean Peak
(ng/ml) Percer
dampheta
Amphetamine 8,130 100 3623 100 2973 100
L-lysine-d-
amphetamine 3,143 39 582 16 565 19
Example 10. Oral Bioavailability of d-amphetamine following administration of an
extended release formulation (intact or crushed) or L-lysine-d-amphetamine
[186] Doses of an extended release formulation of d-amphetamine sulfate
(Dexadrine Spansule capsules) were orally administered to rats as intact capsules or
as crushed capsules and compared to a dose of L-lysine-d-amphetamine containing
an equivalent amount of d-amphetamine base (Fig. 14). The crushed capsules
showed an increase in CmaX and AUCinf of 84 and 13 percent, respectively, as
compared to intact capsules (Tables 12-13). In contrast, Cmax and AUCinf of d-

WO 2005/000334 PCT/US2004/017204
42
amphetamine following administration of L-lysine-d-amphetamine were similar to
that of the intact capsule illustrating that extended release is inherent to the
compound itself and can not be circumvented by simple manipulation.
Table 12. Time-course Concentrations of d-amphetamine Following Oral
Administration of Extended Release Dexadrine Spansule Capsules or Crushed
Extended Release Dexadrine Spansule Capsules or L-lysine-d-amphetamine at
Doses Containing 3 mg/kg d-Amphetamine Base.

Hours Plasma Concentration (ng/ml)

Intact Spansule Capsule Crushed Spansule Capsule L-lysine-damphetamine
0 0 0 0
0.25 32 46 3
0.5 33 85 5
1 SO 147 34
1.5 61 101 60
3 64 66 76
5 46 39 66
8 34 12 38
Table 13. Time-course Concentrations of d-amphetamine Following Oral
Administration of Extended Release Dexadrine Spansule Capsules or Crushed
Extended Release Dexadrine Spansule Capsules or L-lysine-d-amphetamine at
Doses Containing 3 me/kg d-Amphetamine Base.

Parameter Intact Spansule Capsule Crushed Spansule Capsule L-lysine-damphetamin
AUC0.8h(ng.h/ml) 399 449 434
Percent 100 113 109
Cmax (ng/ml) 80 147 76
Percent 100 184 95
Tmax (hours) 1 1 3
Percent 100 100 300
[187] Example 10 illustrates the advantage of the invention over conventional
controlled release formulations of d-amphetamine.
Example 11: Decreased intranasal bioavailability of L-lysine-d-amphetamine vs.
amphetamine

WO 2005/000334 PCT/US2004/017204
43
[188] Male Sprague-Dawley rats were dosed by intranasal administration with 3
mg/kg of amphetamine sulfate or L-lysine-d-amphetamine hydrochloride containing
the equivalent amounts of d-amphetamine. L-lysine-d-amphetamine did not release
any significant amount of d-amphetamine into circulation by IN administration.
Mean (n=4) plasma amphetamine concentration curves of amphetamine vs. L-
lysine-d-amphetamine are shown in Fig. 12. Pharmacokinetic parameters for IN
administration of L-lysine-d-amphetamine are summarized in Table 14.
Table 14. Pharmacokinetic Parameters of Amphetamine vs. L-Iysine-d-amphetarrune
by IN Administration.

Drug AUC (0-1.5 h)
ng/ml h Percent
(/■amphetamine Zmax
(ng/ml) Percent
^amphetamine
Amphetamine 727 100 1,377 100
L-lysine-J-
amphetamine 4 0.5 7 0.5
[096] Example 11 illustrates that when lysine is conjugated to the active agent d-
amphetamine the bioavailability by the intranasal route is substantially decreased
thereby diminishing the ability to abuse the drug by this route.
Example 12: Intravenous bioavailability of amphetamine vs. L-lysine-d-
amphetamine
[189] Male Sprague-Dawley rats were dosed by intravenous tail vein injection with
1.5 mg/kg of d-amphetamine or L-lysine-d-amphetamine containing the equivalent
amount of amphetamine. As observed with IN dosing, the conjugate did not release
a significant amount of d-amphetamine. Mean (n=4) plasma concentration curves of
amphetamine vs. L-Iysine-d-amphetamine are shown in Fig. 13. Pharmacokinetic
parameters for IV administration of L-Iysine-d-amphetamine are summarized in
Table 15.

WO 2005/000334 PCT/US2004/017204
44
Table 15. Pharmacokinetic Parameters of d-amphetamine vs. L-lysine-d-
amphetamine by IV Administration.

Drug AUC (0-1
ng/ml .5
h h) %
Amphetamine Cmax (ng/m!) %
Amphetamine
Amphetamine 190 100 169 100
K-amphetamine 6 3 5 3
[190] Example 12 illustrates that when lysine is conjugated to the active agent
amphetamine the bioavailability of amphetamine by the intravenous route is
substantially decreased, thereby diminishing the ability to abuse the drug by this
route.
Example 13. Oral Bioavaialabilitv of L-lvsine-d-amphetamine compared to d-
amphetamine at escalating doses.
[191] As shown in Figs. 15-19, the fraction of intact L-lysine-d-amphetamine
absorbed following oral administration in rats increased non-linearly in proportion to
escalating doses from 1.5 to 12 mg/kg (d-amphetamine base). The fraction absorbed
at 1.5 mg/kg was only 2.6 percent whereas it increased to 24.6 percent by 12 mg/kg.
The fraction absorbed fell to 9.3 percent at the high dose of 60 mg/kg. Tmax ranged
from 0.25 to 3 hours and peak concentrations occurred earlier than for d-
amphetamine in L-lysine-d-amphetamine dosed rats. L-lysine-d-amphetamine was
cleared more rapidly than d-amphetamine with nearly undetectable concentrations
by 8 hours at the lowest dose.
[192] Tmax for d-amphetamine from L-lysine-d-amphetamine ranged from 1.5 to 5
hours as compared to 0.5 to 1.5 following administration of d-amphetamine sulfate.
The difference in time to reach maximum concentration was greater at higher doses.
Cmax of d-amphetamine following oral delivery of L-lysine-d-amphetamine was
reduced by approximately half as compared to Cmax following i-amphetamine
sulfate administration at doses of 1.5 to 6 mg/kg, approximating human equivalent
doses (HEDs) in the therapeutic range (HED d-amphetamine sulfate; 19.9 to 39.9
mg). HEDs are defined as the equivalent dose for a 60 kg person in accordance to
the body surface area of the animal model. The adjustment factor for rats is 6.2. The
HED for a rat dose of 1.5 mg/kg of d-amphetamine, for example, is equivalent to

WO 2005/000334 PCT/US2004/017204
45
1.5/6.2 x 60 = 14.52 d-amphetamine base; which is equivalent to 14.52/.7284 = 19.9
mg d-amphetamine sulfate, when adjusted for the salt content.
[193] At doses above HEDs in the targeted therapeutic range (12 and 60 mg/kg;
HED d-amphetamine sulfate 79.8 and 399 mg), Cmax was reduced by 73 and 84
percent, respectively, as compared to d-amphetamine sulfate. AUCs of d-
amphetamine following oral administration of L-lysine-d-amphetamine were similar
to those of d-amphetamine sulfate at lower doses. As observed with Cmax, however,
the AUCs for d-amphetamine from L-lysine-d-arnphetamine were substantially
decreased compared to those of d-amphetamine sulfate at higher doses with the
AUCinf reduced by 76% at the highest dose (60 mg/kg; HED 399 mg d-amphetamine
sulfate.
[194] In summary, oral bio availability of d-amphetamine from L-lysine-d-
amphetamine decreased to some degree at higher doses in rats. However,
pharmacokinetics with respect to dose were nearly linear for L-lysine-d-
amphetamine at doses from 1.5 to 60 mg/kg (HED d-amphetamine sulfate; 19.9 to
797.2 mg) with the fraction absorbed ranging from 52 to 81 percent (extrapolated
form 1.5 mg/kg dose). Pharmacokinetics of d-amphetamine sulfate was also nearly
linear at lower doses of 1.5 to 6 mg/kg (HED; 19.9 to 79.7) with the fraction
absorbed ranging form 62 to 84. In contrast to L-lysine-d-amphetamine, however,
parameters were disproportionately increased at higher doses for d-amphetamine
sulfate with the fraction absorbed calculated as 101 and 223 percent (extrapolated
form 1.5 mg/kg dose), respectively, for the suprapharmacological doses of 12 and 60
mg/kg (HED d-amphetamine sulfate; 159.4 and 797.2 mg).
[195] The results suggest that the capacity for clearance of d-amphetamine when
delivered as the sulfate salt becomes saturated at the higher doses whereas the
gradual hydrolysis of L-lysine-d-amphetamine precludes saturation of d-
amphetamine elimination at higher doses. The difference in proportionality of dose
to bioavailability (Cmax and AUC) for d-amphetamine and L-lysine-d-amphetamine
is illustrated in Figs. 20-22. The pharmacokinetic properties of L-lysine-d-
amphetamine as compared to d-amphetamine at the higher doses decrease the ability

WO 2005/000334 PCT/US2004/017204
46
to escalate doses. This improves the safety and reduces the abuse liability of L-
Iysine-d-amphetamine as a method of delivering d-amphetamine for the treatment of
ADHD or other indicated conditions.
Example 14. Intranasal Bioavailability of L-lvsine-d-amphetamine compared to d-
amphetamine.
[196] As shown in Figs. 23-24, bioavailability of d-amphetamine following bolus
intranasal administration of L-lysine-d-amphetamine was approximately 5 percent of
that of the equivalent d-amphetamine sulfate dose with AUCinf values of 56 and
1032, respectively. Cmax of d-amphetamine following L-lysine-d-amphetamine
administration by the intranasal route was also about 5 percent of that of the
equivalent amount of d-amphetamine sulfate with values of 78.6 ng/mL and 1962.9
ng/mL, respectively. As with intravenous administration, Tmax of d-amphetamine
concentration was delayed substantially for L-lysine-d-amphetamine (60 minutes) as
compared to Tmax of d-amphetamine sulfate (5 minutes), again reflecting the gradual
hydrolysis of L-lysine-d-amphetamine. A high concentration of intact L-lysine-d-
amphetamine was detected following intranasal dosing suggesting that the large
decrease in bioavailability of d-amphetamine was due to minimal hydrolysis of L-
lysine-d-amphetamine when delivered by this route. It appears that only minimal
amounts of d-amphetamine can be delivered by intranasal administration of L-
lysine-d-amphetamine.
Example 15. Intravenous Bioavaialabilitv of L-lysine-d-amphetamine compared to
d-amphetamine.
[197] As shown in Figs. 25-26, bioavailability of d-amphetamine following bolus
intravenous administration of L-lysine-d-amphetamine was approximately one-half
that of the equivalent d-amphetamine sulfate dose with AUCinf values of 237.8 and
420.2, respectively. Cmax of d-amphetamine following L-lysine-d-amphetamine
administration was only about one-fourth that of the equivalent amount of d-
amphetamine with values of 99.5 and 420.2, respectively. Tmax of d-amphetamine
concentration was delayed substantially for L-lysine-d-amphetamine (30 minutes) as
compared to Tmax of d-amphetamine sulfate (5 minutes), reflecting the gradual
hydrolysis of L-lysine-d-amphetamine. In conclusion, the bioavailability of d-

WO 2005/000334 PCT/US2004/017204
47
amphetamine by the intravenous route is substantially decreased and delayed when
given as L-lysine-d-amphetamine. Moreover, bioavailability is less than that
obtained by oral administration of the equivalent dose of L-lysine-d-amphetamine.
Summary of LC/MS/MS Bioavailability Data in Rats
[198] The following tables summarize the bioavailability data collected in the
experiments discussed in examples 13-15. Tables 15-17 summarize the
pharmacokinetic parameters of d-amphetamine following oral, intransal, or bolus
intravenous administration of d-amphetamine or L-lysine-d-amphetamine.
Table 15. Pharmacokinetic Parameters of d-amphetamine Following Oral
Administration of L-lysine-d-amphetamine or d-amphetamine at Escalating Doses.

Route Drug Dose
(mg/kg) Cmax
(ng/mL) Tmax
(h) AUC(0-8)
(ng»mL/h) AUC(inf)
(ng»mL/h) F
(%) AUC/Dose
(ng.h.kg/mL/mg) Cmax/Dose
ng.kg/mUmg
Oral L-lysine-
damphetamine 1.5 59.6 3 308 331 61 220.7 39.7
Oral damphetamine 1.5 142.2 0.5 446 461 84 307.3 94.8
Oral L-lysine-
damphetamine 3 126.9 1.5 721 784 72 261.3 42.3
Oral damphetamine 3 217.2 1.5 885 921 84 307.0 72.4
Oral L-lysine-
damphetamine 6 310.8 3 1,680 1,797 82 299.5 51.8
Oral damphetamine 6 815.3 0.25 1,319 1,362 62 227.0 135.9
Oral L-lysine-
damphetamine 12 412.6 5 2,426 2,701 62 225.1 34.4
Oral damphetamine 12 1,533.1 0.25 4,252 4,428 101 369.0 127.8
Oral L-lysine-
damphetamine 60 2,164.3 5 9995.1 11,478 52 191.3 36.1
Oral damphetamine 60 13,735 1 32,323 48,707 223 811.8 228.9
Table 16. Pharmacokinetic Parameters of d-amphetamine Following Bolus
Intravenous Administration of L-lysine-d-amphetamine.

Route Drug Dose
(mg/kg) Cmax
(ng/mL) Tmax
(h) AUC(0-24)
(ng.mL/h) AUC(inf)
(ng-mL/h)
IV L-!ysine-
damphetamine 1.5 99.5 0.5 237.8 237.9
IV damphetamine 1.5 420.2 0.083 546.7 546.9

WO 2005/000334 PCT/US2004/017204
48
Table 17. Pharmacokinetic Parameters of d-amphetamine Following Intranasal
Administration of L-lysine-d-amphetamine.
Route Drug Dose
(mg/kg) Cmax
(ng/mL) Tmax
(h) AUC(0-1)
(ng»mL/h) AUC(inf)
(ng»mL/h)
IN L-lysine-d
amphetamine 10.16 78.6 1 56 91
IN damphetamine 4.12 1962.9 0.083 1032 7291
[199] Tables 18-20 summarize the pharmacokinetic parameters of L-lysine-d-
amphetamine following oral, bolus intravenous, or intransal administration of L-
Iysine-d-amphetamine.
Table 18. Pharmacokinetic Parameters of L-lysine-d-amphetamine Following Oral
Administration of L-lysine-d-amphetamine at Escalating Doses.

Dose Drug Dose
(mg/kg) Cmax
(ng/ml) Tmax
(ng/ml) AUC{0-8)
(ng»ml/h) AUC(inf)
(ng»ml/h) F
(%)
Oral L-iysine-
otemphetamine 1.5 36.5 0.25 59.4 60 2.6
Oral L-lysine-
damphetamine 3 135.4 1.5 329.7 332.1 7.2
Oral L-lysine-
damphetamine 6 676.8 0.25 1156.8 1170.8 12.8
Oral L-lysine-
damphetamlne 12 855.9 1 4238.6 4510.4 24.6
Oral L-lysine-
damphetamine 60 1870.3 3 8234.3 8499.9 9.3
Table 19. Pharmacokinetic Parameters of L-lysine-damphetamine Following Bolus
Intravenous Administration of L-lysine-d-amphetamine.

Route Drug Dose
(mg/kg) Cmax
(ng/mL) Tmax
(h) AUC(0-24)
(ng-mUh) AUC(inf)
(ng»mL/h)
IV L-lysine-
damphetamine 1.5 4513.1 0.083 2,282 2,293

WO 2005/000334 PCT/US2004/017204
49
Table 20. Pharmacokinetic Parameters of L-lysine-d-amphetamine Following
Intranasal Administration of L-lysine-d-amphetamine.

Route Drug Dose
(mg/kg) Cmax
(ng/mL) Tmax
(h) AUC(0-1)
(ng-mL/h) AUC(inf)
(ng-mL/h)
IN L-lysine-
damphetamine 3 3345.1 0.25 2,580 9,139
[200] Tables 21 and 22 summarize the percent bioavailability of d-amphetamine
following oral, intranasal, or intravenous administration of L-lysine-d-amphefamine
as compared to d-amphetamine sulfate.
Table 21. Percent Bioavailability (AUCinf) of d-amphetamine Following
Administration of L-lysine-d-amphetamine by Various Routes as Compared to
Bioavailability Following Administration of d-amphetamine Sulfate.

Dose {mg/kg)
damphetamine base 1.5 3 6 12 60
HED 19.9 39.9 79.7 159.4 797.2
Oral 72 85 132 61 24
IV 43 NA NA NA NA
IN NA 1 NA NA NA
Table 22. Percent Bioavailability (Cmax) of d-amphetamine Following
Administration of L-lysine-d-amphetamine by Various Routes as Compared to
Bioavailability Following Administration of d-amphetamine Sulfate.

Dose (mg/kg) d-
amphetamine base 1.5 3 6 12 60
HED 19.9 39.9 79.7 159.4 797.2
Oral 42 58 38 27 16
IV 24 NA NA NA NA
IN NA 4 NA NA NA
[201] Tables 23-28 summarize the time-course concentrations of d-amphetamine
and L-lysine-d-amphetamine following oral, intranasal or intravenous
administration of either d-amphetamine or L-lysine-d-amphetamine.

WO 2005/000334 PCT/US2004/017204
50
Table 23. Time-course Concentrations of d-amphetamine Following Bolus
Intravenous Administration of L-lysine-d-amphetamine or d-amphetamine Sulfate at
Doses Containing 1.5 mg/kg d-amphetamine Base.

Time Concentration (ng/ml)
(hours) L-lysine-
damphetamine damphetamine
sulfate
0 0 0
0.083 52.8 420.2
0.5 99.5 249.5
1.5 47.1 97.9
3 21.0 38.3
5 9.0 13.2
8 3.7 4.3
24 0.1 0.2
Table 24. Time-course Concentrations of L-lysine-d-amphetamine Following Bolus
Intravenous Administration of L-lysine-d-amphetamine at a Dose Containing 1.5
mg/kg d-amphetamine Base.

Time Concentration
(ng/ml)
(hours) L-lysine-
i/amphetamine
0 0
0.083 4513.1
0.5 1038.7
1.5 131.4
3 19.3
5 17.9
8 8.7
24 11.5

WO 2005/000334 PCT/US2004/017204
51
Table 25. Time-course Concentrations of d-amphetamine Following Oral
Administration of L-lysine-d-amphetamine at Various Doses (mg/kg d-amphetamine
base).

Time Concentration (ng/ml)
(hours) 1,5 mg/kg 3 mg/kg 6 mg/kg 12 mg/kg 60 mg/kg
0 0 0 0 0 0
0.25 20.5 25.3 96 54.3 90.9
0.5 34 40.9 140.2 96 175.1
1 46.7 95.1 225.9 233.3 418.8
1.5 40.7 126.9 268.4 266 440.7
3 59.6 105 310.8 356.8 1145.5
5 38.6 107.6 219.5 412.6 2164.3
8 17.1 48 86 225.1 1227.5
Table 26. Time-course Concentrations of d-amphetamine Following Oral
Administration of d-amphetamine Sulfate at Various Doses (mg/kg d-amphetamine
Base).

Time Concentration ng/ml)
(hours) 1.5 mg/kg 3 mg/kg 6 mg/kg 12 mg/kg 60 mg/kg
0 0 0 0 0 0
0.25 107.1 152.6 815.3 1533.1 6243.6
0.5 142.2 198.4 462.7 1216 7931.6
1 105.7 191.3 301.3 828.8 13735.2
1.5 129.5 217.2 314 904,8 11514.9
3 52.6 135.3 134.6 519.9 NA
5 29.5 73.5 77.4 404.3 NA
8 11.5 25.7 31.8 115.4 NA

WO 2005/000334 PCT/US2004/017204
52
Table 27. Time-course Concentrations of d-amphetamine Following Intranasal
Administration of L-lysine-d-amphetamine or d-amphetamine Sulfate at Doses
Containing 3 mg/kg d-amphetamine Base.
Time Concentration (ng/ml)
(hours) L-lysine- damphetamine
damphetamine sulfate
0.083 31.2 1962.9
0.25 45.3 1497.3
0.5 61.3 996.2
1 78.6 404.6
AUC 56 1032.3
Table 28. Time-course Concentrations of L-Iysine-d-amphetamine Following
Intranasal Administration of L-lysine-d-amphetamine at a Dose Containing 3 mg/kg
d-amphetamine Base.

Time (h) Cone, (ng/ml)
L-lysine-^
amphetamine
0 0
0.083 3345.1
0.25 3369.7
0.5 2985.8
1 1359.3
Example 19. LC/MS/MS analysis of Bioavailability in Dogs
Example Experimental Design:
[202] This was a non-randomized, two-treatment crossover study. All animals
were maintained on their normal diet and were fasted overnight prior to each dose
administration. L-lysine-d-amphetamine dose was based on the body weight
measured on the morning of each dosing day. The actual dose delivered was based
on syringe weight before and after dosing. Serial blood samples were obtained from
each animal by direct venipuncture of a jugular vein using vacutainer tubes
containing sodium heparin as the anticoagulant. Derived plasma samples were

WO 2005/000334 PCT/US2004/017204
53
stored frozen until shipment to the Quest Pharmaceutical Services, Inc. (Newark,
DE). Pharmacokinetic analysis of the plasma assay results was conducted by
Calvert. Animals were treated as follows:

# of Dog/Sex Route of
Administration Treatment Dose Cone.
(mg/mL) Dose Vol.
(mL/kg) Dose Level
(mg/kg)
3M PO 1 0.2 10 2
3M IV 2 1 2 2
The mg units in the dose concentration and dose level refer to the free base form of
test article.
Administration of the Test Article:
[203] Oral: The test article was administered to each animal via a single oral
gavagc. On Day 1, animals received the oral dose by gavage using an esophageal
tube attached to a syringe. Dosing tubes were flushed with approximately 20 mL tap
water to ensure the required dosing solution was delivered.
[204] Intravenous: On Day 8, animals received L-lysine-d-amphetamine as a
single 30-minute intravenous infusion into a cephalic vein.
Sample Collection:
[205] Dosing Formulations: Post-dosing, remaining dosing formulation was saved
and stored frozen.
[206] Blood: Serial blood samples (2 mL) were collected using venipuncture tubes
containing sodium heparin. Blood samples were taken at 0, 0.25, 0.5, 1, 2, 4, 8, 12,
24, 48, and 72 hours post-oral dosing. Blood samples were collected at 0, 0.167,
0.33", 0.49 (prior to stop of infusion), 0.583, 0.667, 0.75, 1, 2, 3, 4, 8, 12, and 23
hours post-intravenous infusion start. Collected blood samples were chilled
immediately.
[207] Plasma: Plasma samples were obtained by centrifugation of blood samples.
Duplicate plasma samples (about 0.2 mL each) were transferred into prelabeled
plastic vials and stored frozen at approximately -70°C.

WO 2005/000334 PCT/US2004/017204
54
Sample Assay:
[208] Plasma samples were analyzed for L-lysine-d-amphetamine and d-
amphetamine using a validated LC-MS/MS method with an LLOQ of 1 ng/mL for
both analytes.
[209] Microsoft Excel (Version 6, Microsoft Corp., Redmond, WA) was used for
calculation of mean plasma concentration and graphing of the plasma concentration-
time data. Pharmacokinetic analysis (non-compartmental) was performed using the
WinNonlin® software program (Version 4.1, Pharsight, Inc. Mountain View, CA).
The maximum concentration, CmaX, and the time to Cmax, Tmax, were observed values.
The area under the plasma concentration-time curve (AUC) was determined using
linear-log trapezoidal rules. The apparent terminal rate constant (z) was derived
using linear least-squares regression with visual inspection of the data to determine
the appropriate number of points (minimum of 3 data points) for calculating z. The
AUC(O-inf) was calculated as the sum of AUC(0-t) and Cpred/z, where Cpred was
the predicted concentration at the time of the last quantifiable concentration. The
plasma clearance (CL/F) was determined as the ratio of Dose/AUC (0-inf). The
mean residence time (MRT) was calculated as the ratio of AUMC(0-inf)/AUC (0-
inf), where AUMC(O-inf) was the area under the first moment curve from the time
zero to infinity. The volume of distribution at steady state (Vss) was estimated as
CL*MRT. Half-life was calculated as In2/z. The oral bioavailability (F) was
calculated as the ratio of AUC(O-inf) following oral dosing to AUC(O-inf) following
intravenous dosing. Descriptive statistics (mean and standard deviation) of the
pharmacokinetic parameters were calculated using Microsoft Excel.
[210] The objectives of this study were to characterize the pharmacokinetics of L-
lysine-d-amphetamine and d-amphetamine following administration of L-lysine-d-
amphetamine in male beagle dogs. As shown in Fig. 27, in a cross-over design, L-
lysine-d-amphetamine was administered to 3 male beagle dogs orally (2 mg/kg) and
intravenously (2 mg/kg, 30-minute infusion). Blood samples were collected up to
24 and 72 hour after the intravenous and oral does, respectively. Plasma samples

WO 2005/000334 PCT/US2004/017204
55
were analyzed using a LC-MS/MS assay which provided an LLOQ of 1 ng/mL for
both analytes.
[211] The mean L-lysine-d-amphetamine and d-amphetamine plasma
concentration-time profiles following an intravenous or oral dose of L-Iysine-d-
amphetamine are presented in Figs. 29 and 30, respectively. Comparative profiles of
L-lysine-d-amphetamine to d-amphetamine following both routes are depicted in
Figs. 27-28. Individual plots are depicted in Figs. 31-32. The pharmacokinetic
parameters are summarized in Tables 29-37.
[212] Following a 30-minute intravenous infusion of L-lysine-d-amphetamine, the
plasma concentration reached a peak at the end of the infusion. Post-infusion L-
lysine-d-amphetamine concentration declined very rapidly in a biexponential
manner, and fell below the quantifiable limit (1 ng/mL) by approximately 8 hours
post-dose. Results of non-compartmental pharmacokinetic analysis indicate that L-
lysine-d-amphetamine is a high clearance compound with a moderate volume of
distribution (Vss) approximating total body water (0.7 L/kg). The mean clearance
value was 2087 ml7h•kg (34.8 mL/min•kg) and was similar to the hepatic blood
flow in the dog (40 mL/min•kg). Consequently, L-lysine-d-amphetamine is a
moderate to high hepatic extraction compound with significant first pass effects
(including the conversion to d-amphetamine) following oral administration.
[213] L-lysine-d-amphetamine was rapidly absorbed after oral administration with
Tmax at 0.5 hours in all three dogs. Mean absolute oral bioavailablity was 33%.
Since significant first pass effects are expected for L-lysine-d-amphetamine, a 33%
bioavailability suggests that L-lysine-d-amphetamine is very well absorbed in the
dog. The apparent terminal half-life was 0.39 hours, indicating rapid elimination, as
observed following intravneous administration.
[214] Plasma concentration-time profiles of d-amphetamine following intravenous
or oral administration of L-lysine-d-amphetamine were very similar, with Cmax, Tmax
and AUC values for both routes essentially the same. At a 2 mg/kg oral dose of L-
lysine-d-amphetamine, the mean Cmax of d-amphetamine was 104.3 ng/mL. The

WO 2005/000334 PCT/US2004/017204
56
half-life of d-amphetamine was 3.1 to 3.5 hours, much longer when compared to L-
lysine-d- amphetamine.
[215] In this study, L-lysine-d-amphetamine was infused over a 30 minute time
period. Due to rapid clearance of L-lysine-d-amphetamine it is likely that
bioavailability of d-amphetamine from L-lysine-d-amphetamine would decrease if a
similar dose were given by intravenous bolus injection. Even when given as an
infusion the bioavailability of d-amphetamine from L-lysine-d-amphetamine did not
exceed that of a similar dose given orally and the time to peak concentration was
substantially delayed. This data further supports that L-lysine-d-amphetamine
affords a decrease in the abuse liability of d-amphetamine by intravenous injection.
Table 29. Pharmacokinetic Parameters of L-ly sine-d-amphetamine in Male Beagle
Dogs Following Oral or Intravenous Administration of L-lysine-d-amphetamine (1
mg/kg d-amphetamine base),

Route Dose
(mg/kg) (ng/mL) Tmax
(h) AUC(inf)
(ng'h/ml) t,/2
00 MRT
(h) CL/F
(mL/h'kg) (mL/kg) F
(%)
IV 1 1650 0.49 964 0.88 0.33 2087 689 NA
(0.00) (178) (0.49-
0.49) (97.1) (0.2) (0.03) (199) (105.9)
Oral I 328.2 0.5 319 0.39 0.81 6351 NA 33
(0.00) (91.9) (0.5-0.5) (46.3) (0.1) (0.19) (898.3) (1.9)
": median (range)
Table 30. Pharmacokinetic Parameters of d-amphetamine in Male Beagle Dogs
Following Oral or Intravenous Administration of L-lysine-d-amphetamine (1 mg/kg
d-amphetamine base).

Route Dose
(mg/kg) '-■max
(ng/mL) * max
(h) AUC(inf)
(ng»h/mL) tl/2
(h)
IV 2 113.2 1.0 672.5 3.14
(0.00) (3.2) (0.67 - 2.0) (85.7) (0.4)
Oral 2 104.3 2.0 728.0 3.48
(0.00) (21.8) (2-2) (204.9) (0.4)
u: median (range)

WO 2005/000334 PCT/US2004/017204
57
Table 31. Pharmacokinetics of L-lysine-d-amphetamine in Male Beagle Dogs
Following Intravenous Administration of L-lysine-d-amphetamine (1 mg/kg d-
amphetamine base).
Dose Route : 30-min iv Infusion Dose : 2 mg/kg/h (free form)

Dog ID (ng/mL) T a
1 max
(h) AUC(0-t)
(ng'h/mL) AUC(inf)
(ng»h/mL) tin
(h) CL
(mL/h/kg) Vss
(mL/kg) MRT
(h)
1 1470.3 0.49 898.2 900.2 0.72 2222 807.4 0.36
2 1826.4 0.49 1072.3 1076.1 NDb 1859 603.4 0.32
3 1654.2 0.49 914.1 916.9 1.05 21S1 656.0 0.30

Mean 1650 0.49 961.5 964.4 0.88 2087 689.0 0.33
SD 178 0.49-0.49 96.0 97.1 0.2 199 105.9 0.03
a: median (range); b: not determined
Abbreviations of pharmacokinetic parameters are as follows : Cmax, maximum observed plasma
concentration; AUC(O-t), total area under the plasma concentration versus time curve from 0 to
the last data point; AUC(O-inf), total area under the plasma concentration versus time curve; t1/2,
apparent terminal half-life; CL, clearance following iv administration; MRT, mean residence
time; Vss, volume of distribution at steady state.
Table 32. Pharmacokinetic Parameters of L-Iysine-d-amphetamine in Male Beagle
Dogs Following Oral Administration of L-Iys ine-d-amphetamine (1 mg/kg d-
amphetamine base).
Dose Route: Oral Dose ; 2 mg/kg (free form)

Dog ID (ng/mL) T a
• ma*
(h) AUC(O-t)
(ng"h/mL) AUC(inf)
(ng»h/mL) tin
(h) CL/F
(mL/h/kg) MRT
(h) F
(%)
1 350.2 0.5 275.3 277.1 0.24 7218 0.68 30.8
2 407.2 0.5 367,8 368.7 0.48 5424 0.74 34.3
3 227.4 0.5 310.8 312.0 0.45 6410 1.03 34.0

Mean 328.2 0.5 318.0 319.3 0.39 6351 0.81 33.0
SD 91.9 0.0 46.7 46.3 0.1 898.3 0.19 1.9
B: median (range)
Abbreviations of pharmacokinetic parameters areas follows : Cmax maximum observed plasma concentration;
Tmax, time when Cmax observed; AUC(O-t), total area under the plasma concentration versus time curve from 0 to
the last data point; AUC(O-inf), total area under the plasma concentration versus time curve; t1/2. apparent
terminal half-life; CL/F, oral clearance; MRT, mean residence time; F, bioavailability.

WO 2005/000334 PCT/US2004/017204
58
Table 33. Pharmacokinetics of L-lysine-d-amphetamine in Male Beagle Dogs
Following Intravenous Administration of L-lysine-d-amphetamine (1 mg/kg d-
amphetamine base).
Dose Route : 30-min iv Infusion Dose : 2 mg/kg of L-Iysine-d-amphetamine
(free form)

Dog ID (ng/mL) 1 nux
(h) AUC(O-t)
(ng'h/mL) AUC(inf)
(ng'h/mL) tin
(h)
I 111.2 2.0 751.9 757.6 3.35
2 116.8 0.67 668.5 673.7 3.43
3 111.4 1.0 557.8 586.1 2.65

Mean 113.2 1.00 659.4 672.5 3.14
SD 3.2 0.67-2.0 97 85.7 0.4
J: median (range)
Abbreviations of pharmacokinetic parameters are as follows : Cmax, maximum observed plasma
concentration; Tffmax, time when Cm3X observed; AUC(O-t), total area under the plasma concentration versus
time curve from 0 to the last data point; AUC(O-inf), total area under the plasma concentration versus time
curve; tI/2, apparent terminal half-life; CL/F, oral clearance; MRT, mean residence time; F, bioavailabiltty.
Table 34. Pharmacokinetics of L-lysine-d-amphetamine in Male Beagle Dogs
Following Oral Administration of L-lysine-d-amphetamine (1 mg/kg d-
amphetamine base).
Dose Route : Oral Dose : 2 mg/kg of L-lysine-d-amphetamine (free form)

Dog ID '•'max
(ng/mL) (h) AUC(0-t)
(ng'h/mL) AUC(inf)
(ng»h/mL) (h)
I 102.1 :2.0 686.34 696.89 3.93
2 127.2 2.0 937.57 946.62 3.44
3 83.7 2.0 494.61 540.38 3.06

Mean 104.3 2.0 706.2 728.0 3.48
SD 21.8 2.0 -2.0 222.1 204.9 0.4
a: median (range)
Abbreviations of pharmacokinetic parameters are as follows : Qn^, maximum observed plasma
concentration; TmM, time when Cmax observed; AUC(O-t), total area under the plasma concentration versus
time curve from 0 to the last data point; AUC(O-inf). total area under the plasma concentration versus time
curve; tI/2, apparent terminal half-life; CUV, oral clearance; MRT, mean residence time; F, bioavailability.

WO 2005/000334 PCT/US2004/017204
59
Table 35. Pharmacokinetics of d-amphetamine in Male Beagle Dogs Following
Oral Administration of L-lysine-d-amphetamine or d-amphetamine sulfate (1.8
mg/kg d-amphetamine base).

Time Mean Plasma Concentration Standard Deviation (SD) Coefficient of Variation (CV)
(hours) damphetamine L-lysined
amphetamine damphetamine L-lysine-d
amphetamine damphetamine L-lysine-d
amphetamine
0 0 0 0 0 0 0
1 431.4 223.7 140.7 95.9 32.6 42.9
2 360 291.8 87.6 93.6 24.3 32.1
4 277.7 247.5 68.1 66 24.5 26.7
6 224.1 214.7 59.3 62.1 26.5 28.9
8 175.4 150 66.7 40.1 38.0 26.7
12 81.4 47.6 58.7 19 72.1 39.9
16 33 19.6 28.1 9 85.2 45.9
24 7.2 4.5 4.5 1.7 62.5 37.8
Table 36. Pharmacokinetics of d-amphetamine in Female Beagle Dogs Following
Oral Administration of L-Iysine-d-amphetamine or d-amphetamine sulfate (1.8
mg/kg d-amphetamine base).

Time Mean Plasma Concentration Standard Deviation (SD) Coefficient of Variation (CV)
(hours) damphetamine L-lysine-d
amphetamine damphetamine L-fysine-d
amphetamine damphetamine L-lysine-d
amphetamine
0 0 0 0 0 0 0
1 217.8 308.8 141.7 40.7 65.1 13.2
2 273.5 308 113.7 29.6 41.6 9.6
4 266 260.9 132.7 37.3 49.9 14.3
6 204.7 212.1 84.5 38.7 41.3 18.2
8 160.1 164.3 72.7 43.5 45.4 26.5
12 79.4 68.7 41.3 31 52.0 45.1
16 25.5 22.3 13.4 4.7 52.5 21.1
24 5.6 5.4 4.1 1.9 73.2 35.2

WO 2005/000334 PCT/US2004/017204
60
Table 37. Pharmacokinetic Parameters of d-amphetamine in Male and Female
Beagle Dogs Following Oral Administration of L-lysine-d-amphetamine or d-
amphetamine sulfate (1.8 mg/kg d-amphetamine base).

Parameter Males Females

Compound Compound

damphetamine L-Iysine-d
amphetamine damphetamine L-lysine-d
amphetamine
AUCinf 3088.9 2382.2 2664.5 2569.9
Percent 100 77 100 96
Cmax 431.4 291.8 308.8 273.5
Percent 100 67 100 89
Tmax{hours) 1 2 1 2
Percent 100 200 100 200
Example 20. Delayed Cardiovascular Effects of L-lysine-d-amphetamine as
Compared to d-amphetamine Following Intravenous Infusion.
[216] Systolic and diastolic blood pressure (BP) are increased by d-amphetamine
even at therapeutic doses. Since L-lysine-d-amphetamine is expected to release d-
amphetamine (albeit slowly) as a result of systemic metabolism, a preliminary study
was done using equimolar doses of d-amphetamine or L-lysine-d-amphetamine to 4
dogs (2 male and 2 female). The results suggest that the amide prodrug is inactive
and that slow release of some d-amphetarmne, occurs beginning 20 minutes after the
first dose. Relative to d-amphetamine, however, the effects are less robust. For
example, the mean blood pressure is graphed in Fig. 35. Consistent with previously
published data (Kohli and Goldberg, 1982), small doses of d-amphetamine were
observed to have rapid effects on blood pressure. The lowest dose (0.202 mg/kg,
equimolar to 0.5 mg/kg of L-lysine-d-amphetamine) produced an acute doubling of
the mean BP followed by a slow recovery over 30 minutes.
[217] By contrast, L-lysine-d-amphetamine produced very little change in mean
BP until approximately 30 minutes after injection. At that time, pressure increased
by about 20-50%. Continuous release of d-amphetamine is probably responsible for
the slow and steady increase in blood pressure over the remaining course of the
experiment. Upon subsequent injections, d-amphetamine is seen to repeat its effect

WO 2005/000334 PCT/US2004/017204
61
in a non-dose dependent fashion. That is, increasing dose 10-fold from the first
injection produced a rise to the same maximum pressure. This may reflect the state
of catecholamine levels in nerve terminals upon successive stimulation of d-
amphetamine bolus injections. Note that the rise in mean blood pressure seen after
successive doses of L-Iysine-d-amphetamine (Fig. 35) produces a more gradual and
less intense effect. Similar results were observed for left ventricular pressure (Fig.
36). These results further substantiate the significant decrease in d-amphetamine
bioavailability by the intravenous route when given as L-Iysine-d-amphetamine. As
a result the rapid onset of the pharmacological effect of d-amphetamine that is
sought by persons injecting the drug is eliminated.
Table 38. Effects of L-lysine-d-amphetamine on Cardiovascular Parameters in the
Anesthetized Dog - Mean Values (n=2)

TREATMENT TIME SAP %
Change DAP %
Change MAP /o
Change LVP %
Change
0.9% Saline 0 81 0 48 0 61 0 87 0
1 ml/kg 30 87 7 54 11 67 10 87 0
L-lysine-d
amphetamine 0 84 0 51 0 64 0 86 0
0.5 mg/kg 5 87 4 52 3 66 3 87 2
15 93 11 51 1 67 5 95 11
25 104 25 55 8 73 15 105 22
30 107 28 58 14 77 21 108 26
L-lysine-d
amphetamine 0 105 0 55 0 74 0 108 0
1.0 mg/kg 5 121 15 63 15 85 15 120 11
15 142 35 73 33 100 35 140 29
25 163 55 97 75 124 68 162 50
30 134 28 73 32 98 32 144 33
L-lysine-d
amphetamine 0 132 0 71 0 95 0 144 0
5.0 mg/kg 5 142 7 71 0 99 4 151 5
15 176 33 98 39 130 37 184 28
25 126 -5 69 -3 96 1 160 11
30 132 0 70 -1 99 4 163 13
SAP - systolic arterial pressure (mmHg) MAP - mean arterial pressure (mmHg)

WO 2005/000334 PCT/US2004/017204
62
DAP - diastolic arterial pressure (mmHg) LVP - left ventricular pressure (mmHg)
% Change- percent change from respective Time 0.
Table 39. Effects of d-Amphetamine on Cardiovascular Parameters in the
Anesthetized Dog - Mean values (n=2)

TREATMENT TIME SAP %
Change DAP %
Change MAP to
Change LVP %
Change
0.9% Saline 0 110 0 67 0 84 0 105 0
1 ml/kg 30 108 -2 65 -3 82 -2 101 -3
damphetamine 0 111 0 67 0 84 0 104 0
0.202 mg/kg 5 218 97 145 117 176 109 214 107
15 168 52 97 45 125 49 157 52
25 148 34 87 30 110 31 142 37
30 140 26 80 20 103 23 135 30
damphetamine 0 139 0 78 0 101 0 133 0
0.404 mg/kg 5 240 73 147 88 187 85 238 79
15 193 39 112 44 145 43 191 43
25 166 19 92 17 122 20 168 26
30 160 16 87 11 117 16 163 22
damphetamine 0 158 0 87 0 115 0 162 0
2.02 mg/kg 5 228 44 128 48 169 47 227 40
15 196 24 107 23 142 23 200 . 24
25 189 20 102 17 135 17 192 19
30 183 16 98 13 129 12 187 16
SAP - systolic arterial pressure (mmHg) MAP - mean arterial pressure (mmHg)
DAP - diastolic arterial pressure (mmHg) LVP - left ventricular pressure (mmHg)
% Change- percent change from respective Time 0.
Example 21. Pharmacodynamic (Locomotor) Response to Amphetamine vs. L-
lysine-d-amphetamine by Oral Administration
[218] Male Sprague-Dawley rats were provided water ad libitum, fasted overnight
and dosed by oral gavage with 6 mg/kg of amphetamine or L-lysine-d-amphetamine
containing the equivalent amount of d-amphetamine. Horizontal locomotor activity
(HLA) was recorded during the light cycle using photocell activity chambers (San
Diego Instruments). Total counts were recorded every 12 minutes for the duration
of the test. Rats were monitored in three separate experiments for 5, 8, and 12

WO 2005/000334 PCT/US2004/017204
63
hours, respectively. Time vs. HLA counts for d-amphetamine vs. L-lysine-d-
amphetamine is shown in Figs. 37-38. In each experiment the time until peak
activity was delayed and the pharmacodynamic effect was evident for an extended
period of time for L-lysine-d-amphetamine as compared to d-amphetamine. The
total activity counts for HLA of Lys-Amp dosed rats were increased (11-41%) over
those induced by d-amphetamine in all three experiments (Tables 40 and 41).
Table 40. Locomotor Activity of Rats Orally Administered d-amphetamine vs. L-
lysine-d-amphetamine (5 Hours)

Test Material TotaJ Activity
Counts Total Activity
Counts Above
Baseline Peak of activity
(Counts per
0.2 h) Time of Peak
(Counts per
0.2 h) Time of Last
Count Above 200
per 0.2 h
Vehicle 4689 4174 80 1.4
L-lysine-d
amphetamine 6417 5902 318 1.8 5h
damphetamine 515 0 291 0.6 2.6h
Table 41. Locomotor Activity of Rats Orally Administered Amphetamine vs. L-
lysine-d- amphetamine (12 Hours)

Test Material Total Activity
Counts Total Activity
Counts Above
Baseline Peak of activity
(Counts per
0.2 h) Time of Peak
(Counts per
0.2 h) Time of Last
Count Above 100
per 0.2 h
Vehicle 936 0 81 7.2 -
L-lysine-d
amphetamine 8423 7487 256 1.8 8.6 h
damphetamine 6622 5686 223 0.6 6.4 h
Example 22. Pharmacodynamic Response to Amphetamine vs. L-lvsine-d-
amphetamine by Intranasal Administration
[219] Male Sprague-Dawley rats were dosed by intranasal administration with 1.0
mg/kg of amphetamine or L-lysine-d-amphetamine containing the equivalent
amount of d-amphetamine. In a second set of similarly dosed animals
carboxymethyl cellulose (CMC) was added to the drug solutions at a concentration
of 62.6 mg/ml (approximately 2-fold higher than the concentration of L-lysine-d-
amphetamine and 5-fold higher than the d-amphetamine content). The CMC drug
mixtures were suspended thoroughly before each dose was delivered. Locomotor

WO 2005/000334 PCT/US2004/017204
64
activity was monitored using the procedure described in the section titled example 7.
As shown in Figs. 39-40, the activity vs. time (1 hour or 2 hours) is shown for
amphetamine/CMC vs. L-lysine-d-amphetamine and compared to that of
amphetamine vs. L-lysine-d-amphetamine CMC. As seen in Fig. 39, addition of
CMC to L-lysine-d-amphetamine decreased the activity response of IN dosed rats to
levels similar to the water/CMC control, whereas no effect was seen on
amphetamine activity by the addition of CMC. The increase in activity over
baseline of L-lysine-d-amphetamine with CMC was only 9% compared to 34% for
Lys-Amp without CMC when compared to activity observed for d-amphetamine
dosed animals (Table 42). CMC had no observable affect on d-amphetamine
activity induced by IN administration.
Table 42. Locomotor Activity of Intranasal d-amphetamine vs. L-lysine-d-
amphetamine with and without CMC

Drug n Total Activity Counts
(1h) Total Activity Counts
Above Baseline Percent (/amphetamine
Amphetamine 3 858 686 100
damphetamine CMC 3 829 657 100
L-lysinc-d-
amphetamine 4 408 237 35
L-lysine-d-
amphetamine CMC 4 232 60 9
Water 1 172 0 0
Water CMC 1 172 0 0
Example 23. Pharmacodvnamic Response to Amphetamine vs. L-lvsine-d-
amphetamine by Intravenous (TV) Administration
[220] Male Sprague-Dawley rats were dosed by intravenous administration with
1.0 mg/kg of d-amphetamine or L-Iysine-d-amphetamine containing the equivalent
amount of amphetamine. The activity vs. time (3 hours) is shown for d-
amphetamine vs. L-lysine-d-amphetamine (Fig. 41). The activity induced by L-
lysine-d-amphetamine was substantially decreased and time to peak activity was
delayed. The activity expressed as total activity counts over a three hour period of
time is shown in Fig. 41. The increase in activity over baseline of L-lysine-d-

WO 2005/000334 PCT/US2004/017204
65
amphetamine was 34% for L-Iysine-d-amphetamine when compared to activity
observed for d-amphetamine dosed animals (Table 43).
Table 43. Total activity counts after d-amphetamine vs. L-lysine-d-amphetamine
Following Intravenous (IV) Administration,

Drug n Total Activity Counts
3h Above Baseline Percent damphetamine
damphetamine 3 1659 1355 100
L-lysine-d-
amphetamine 4 767 463 34
Water 1 304 0 0
Example 24: Decrease in toxicitv of orally administered L-lysine-d-amphetamine
[221] Three male and three female Sprague Dawley rats per group were given a
single oral administration of L-lysine-d-amphetamine at 0.1, 1.0, 10, 60, 100 or 1000
mg/kg (Table 44). Each animal was observed for signs of toxicity and death on
Days 1-7 (with Day 1 being the day of the dose) and one rat/sex/group was
necropsied upon death (scheduled or unscheduled).
Table 44. Dosing Chart Oral Administration of L-lysine-d-amphetamine Toxicity
Testing.

Groups No. of Animals Test Article Dosages (mg/kg) Concentrations
(mg/mL)
M F
1 3 3 L-lysine-J-amphetamine 0.1 0.01
2 3 3 L-Iysine-d-amphetamine 1.0 0.1
3 3 3 L-Iysine-d-amphetamine 10 1.0
4 3 3 L-lysine-d-amphetamine 60 6.0
5 3 3 L-tysine-d-amphetamine 100 10
6 3 3 L-lysine-d-amphetamine 1000 100
[222] Key observations of this study include:
• All animals in Groups 1-3 showed no observable signs throughout the
conduct of the study.

WO 2005/000334 PCT/US2004/017204
66
• AH animals in Groups 4-6 exhibited increased motor activity within
two hours post-dose and which lasted into Day 2.
• One female rat dosed at 1000 mg/kg was found dead on Day 2.
Necropsy revealed chromodacryorrhea, chromorhinorrhea, distended
stomach (gas), enlarged adrenal glands, and edematous and distended
intestines.
• A total of 4 rats had skin lesions of varying degrees of severity on
Day 3.
• One male rat dosed at 1000 mg/kg was euthanatized on Day 3 due to
open skin lesions on the ventral neck.
• All remaining animals appeared normal from Day 4 through Day 7.
[223] Animals were observed for signs of toxicity at 1, 2 and 4 h post-dose, and
once daily for 7 days after dosing and cage-side observations were recorded.
Animals found dead, or sacrificed moribund were necropsied and discarded, A total
of one animal/sex/group was necropsied upon scheduled or unscheduled death.
[224] Cage-side observations and gross necropsy findings are summarized in Table
5. The data are not sufficient to establish a lethal dose, however, the study indicates
that the lethal oral dose of L-lysine-d-amphetamine is above 1000 mg/kg, because
only one death occurred out of a group of six animals. Although a second animal in
this dose group was euthanatized on Day 3, it was done for humane reasons and it
was felt that this animal would have fully recovered. Observations suggested drug-
induced stress in Groups 4-6 that is characteristic of amphetamine toxicity (NTP,
1990; NIOSH REGISTRY NUMBER: SI1750000; Goodman et. al. 1985). All
animals showed no abnormal signs on Days 4-7 suggesting full recovery at each
treatment level.
[225] The lack of data to support an established lethal dose is believed to be due to
a putative protective effect of conjugating amphetamine with lysine. Intact L-lysine-
d-amphetamine has been shown to be inactive, but becomes active upon metabolism
into the unconjugated form (d-amphetamine). Thus, at high doses, saturation of

WO 2005/000334 PCT/US2004/017204
67
metabolism of L-lysine-d-amphetamine into the unconjugated form may explain the
lack of observed toxicity, which was expected at doses greater than 100 mg/kg,
which is consistent with d-amphetamine sulfate (NTP, 1990). The formation rate of
d-amphetamine and the extent of the formation of amphetamine may both attribute
to the reduced toxicity. Alternatively, oral absorption of L-lysine-d-amphetamine
may also be saturated at such high concentrations, which may suggest low toxicity
due to limited bioavailability of L-lysine-d-amphetamine.
Example 25: In Vitro Assessment of L-lvsine-d-amphetamine Pharmacodvnamic
Activity.
[226] It was anticipated that the acylation of amphetamine, as in the amino acid
conjugates discussed here, would significantly reduce the stimulant activity of the
parent drug. For example, Marvola (1976) showed that N-acetylation of
amphetamine completely abolished the locomotor activity increasing effects in mice.
To confirm that the conjugate was not directly acting as a stimulant, we tested
(Novascreen, Hanover, MD) the specific binding of Lys-Amp (10"9 to 10'5 M) to
human recombinant dopamine and norepinephrine transport binding sites using
standard radioligand binding assays. The results (see Table 45) indicate that the
Lys-Amp did not bind to these sites. It seems unlikely that the conjugate retains
stimulant activity in light of these results. (Marvola, M. (1976). "Effect of acetylated
derivatives of some sympathomimetic amines on the acute toxicity, locomotor
activity and barbiturate anesthesia time in mice." Acta Pharmacol Toxicol (Copenh)
38(5): 474-89).
Table 45. Results From Radioligand Binding Experiments with L-lvsine-d-
amphetamine

Assay Radioligand Reference
Compound Ki (M) for
Ref. Cpd. Activity*
NE
Transporter [3H]-Nisoxetine Desipramine 4.1xl(Ty No
DA
Transporter [3HJ-WIN35428 GBR-12909 7.7 x 10 -y No
*No activity is defined as producing between -20% and 20%- inhibition of radioligand binding
(Novascreen).

WO 2005/000334 PCT/US2004/017204
68
Example 26: In Vitro Assessment "Kitchen Tests" to Release Amphetamine.
[227] It was anticipated that attempts would be made by illicit chemists to treat the
compound with various easily accessible physical and chemical methods by which
to release free amphetamine from the conjugate. An abuse-resistant preparation
would have the additional feature of not releasing d-amphetamine when exposed to
water, acid (vinegar), base (baking powder and baking soda), and heat. In several
tests with L-lysine-d-amphetamine and GGG-Amp, no amphetamine was detected
after the following treatments:

Vinegar Tap Water Baking Powder Baking Soda
L-Iysine-d-
amphetamine 0% 0% 0% 0%
Gly3-Amp 0% 0% 0% 0%
Samples were heated to boiling for 20-60 minutes in each test.
Example 27. Bioavailability of Various Amino Acid-Amphetamine Compounds
Administered by Oral, Intranasal. and Intravenous Routes.
[228] Oral Administration. Male Sprague-Dawley rats were provided water ad
libitum, fasted overnight, and dosed by oral gavage with amphetamine or amino
acid-amphetamine conjugates containing the equivalent amount of amphetamine.
[229] Intranasal Administration. Male Sprague-Dawley rats were dosed by
intranasal administration with 1.8 mg/kg of amphetamine or lysine-amphetamine
containing the equivalent amount of amphetamine.
[230] The relative in vivo performance of various amino acid-amphetamine
compounds is shown in Figs. 42-50 and summarized in Table 46. Intranasal
bioavailability of amphetamine from Ser-Amp was decreased to some degree
relative to free amphetamine. However, this compound was not bioequivalent with
amphetamine by the oral route of administration. Phenylalanine was bioequivalent
with amphetamine by the oral route of administration, however, little or no decrease
in bioavailability by parenteral routes of administration was observed. Gly3-Amp
had nearly equal bioavailability (90%) by the oral route accompanied by a decrease

WO 2005/000334 PCT/US2004/017204
69
in Cmax {14%). Additionally, Gly3-Amp showed a decrease in bioavailabiiity
relative to amphetamine by intranasal and intravenous routes.
Table 46. Percent Bioavailabiiity of Amino Acid Amphetamine Compounds
Administered by Oral, Intranasal or Intravenous Routes

Drug Oral Inlranasal Intravenous

Percent AUC Percent Omax Percent AUC Percent Cmax Percent AUC Percent Omax
Amphetamine 100 100 100 100 100 100
E-Amp 73 95 NA NA NA NA
EE-Amp 26 74 NA NA NA NA
L-Amp 65 81 NA NA NA NA
S-Amp 79/55 62/75 76 65 NA NA
GG-Amp 79 38 88 85 NA NA
GGG-Amp 111/68 74/73 32 38 45 46
F-Amp 95 91 97 95 87 89
EEF-Amp 42 73 39 29 NA NA
FF-Amp 27 64 NA NA NA NA
Gu!onate-Amp 1 1 0.4 0.5 3 5
K-Amp 98 55 0.5 0.5 3 3
KG-Amp 69 71 13 12 NA NA
cfcK-Amp 16 7 2 2 NA NA
LE-Amp 40 28 6 6 NA NA
H-Amp 16 21 22 42 NA NA
Example 28. Decreased Oral Cmax of d-Amphetamine Conjugates.
[231] Male Sprague-Dawley rats were provided water ad libitum, fasted overnight
and dosed by oral gavage with amphetamine conjugate or d-amphetamine sulfate.
All doses contained equivalent amounts of d-amphetamine base. Plasma d-
amphetamine concentrations were measured by ELISA (Amphetamine Ultra,
109319, Neogen, Corporation, Lexington, KY). The assay is specific for d-
amphetamine with only minimal reactivity (0.6%) of the major d-amphetamine
metabolite (para-hydroxy-d-amphetamine) occurring. Plasma d-amphetamine and L-
lysine-d-amphetamine concentrations were measured by LC/MS/MS where
indicated in examples.

WO 2005/000334 PCT/US2004/017204
70
Example 29. Decreased Intranasal Bioavailability (AUC and Cmax) of d-
Amphetamine Conjugates.
[232] Male Sprague-Dawley rats were provided water ad libitum and doses were
administered by placing 0.02 ml of water containing amphetamine conjugate or d-
amphetamine sulfate into the nasal flares. All doses contained equivalent amounts of
d-amphetamine base. Plasma d-amphetamine concentrations were measured by
ELISA (Amphetamine Ultra, 109319, Neogen, Corporation, Lexington, KY). The
assay is specific for d-amphetamine with only minimal reactivity (0.6%) of the
major d-amphetamine metabolite (para-hydroxy-d-amphetamine) occurring. Plasma
d-amphetamine and L-lysine-d-amphetamine concentrations were measured by
LC/MS/MS where indicated in examples.
Example 30. Decreased Intravenous Bioavailabilitv (AUC and Cmax) of d-
Amphetamine Conjugates.
[233] Male Sprague-Dawley rats were provided water ad libitum and doses were
administered by intravenous tail vein injection of 0.1 ml of water containing
amphetamine conjugate or d-amphetamine sulfate. All doses contained equivalent
amounts of d-amphetamine base. Plasma d-amphetamine concentrations were
measured by ELISA (Amphetamine Ultra, 109319, Neogen, Corporation, Lexington,
KY). The assay is specific for d-amphetamine with only minimal reactivity (0.6%)
of the major d-amphetamine metabolite (para-hydroxy-J-amphetamine) occurring.
Plasma d-amphetamine and L-lysine-d-amphetamine concentrations were measured
by LC/MS/MS where indicated in examples.
Example 31. Attachment of Amphetamine to Variety of Chemical Moieties
[234] The above examples demonstrate the use of an amphetamine conjugated to a
chemical moiety, such as an amino acid, which is useful in reducing the potential for
overdose while maintaining its therapeutic value. The effectiveness of binding
amphetamine to a chemical moiety was demonstrated through the attachment of
amphetamine to Iysine (K), however, the above examples are meant to be illustrative
only. The attachment of amphetamine to any variety of chemical moieties (i.e.
peptides, glycopeptides, carbohydrates, nucleosides, or vitamins)as described below
through similar procedures using the following exemplary starting materials.

WO 2005/000334 PCT/US2004/017204
71
Amphetamine Synthetic Examples
Synthesis of Gly2-Amp
Gly2-Amp was synthesized by a similar method except the amino acid
starting material was Boc-Gly-Gly-OSu.
Synthesis of Glu2-Phe-Amp
GIu2-Phe-Amp was synthesized by a similar method except the amino acid
starting material was Boc-Glu(OtBu)-Glu(OtBu)-OSu and the starting drug
conjugate was Phe-Amp (see Phe-Amp synthesis).
Synthesis of His-Amp
His-Amp was synthesized by a similar method except the amino acid starting
material was Boc-His(Trt)-OSu.
Synthesis of Lys-Gly-Amp
Lys-Gly-Amp was synthesized by a similar method except the amino acid
starting material was Boc-Lys(Boc)-OSu and the starting drug conjugate was
Gly-Amp (see Gly-Amp synthesis).
Synthesis of Lys-Glu-Amp
Lys-Glu-Amp was synthesized by a similar method except the amino acid
starting material was Boc-Lys(Boc)-OSu and the starting drug conjugate was
Glu-Amp.
Synthesis of Glu-Amp
Glu-Amp was synthesized by a similar method except the amino acid starting
material was Boc-Glu(OtBu)-OSu.
Synthesis of (d)-Lys-(l)-Lys-Amp
(d)-Lys-(l)-Lys-Amp was synthesized by a similar method except the amino
acid starting material was Boc-(d)-Lys(Boc)~(l)-Lys(Boc)-OSu.
Synthesis of Gulonic acid-Amp
Gul-Amp was synthesized by a similar method except the carbohydrate
starting material was gulonic acid-OSu.
Example 32. Lack of detection of L-lysine-d-amphetamine in Brain Tissue
Following Oral Administration.
[235] Male Sprague-Dawley rats were provided water ad libitum, fasted overnight
and dosed by oral gavage with L-lysine-d-amphetamine or d-amphetamine sulfate.
All doses contained equivalent amounts of d-amphetamine base. As shown in Figs.
51A-B, similar levels of d-amphetamine were detected in serum as well as in brain
tissue following administration of d-amphetamine sulfate or L-Iysine-d-
amphetamine. The conjugate L-lysine-d-amphetamine, however, was present in

WO 2005/000334 PCT/US2004/017204
72
appreciable amounts in serum but was not detected in brain tissue indicating that the
conjugate does not cross the blood brain barrier to access the central nervous system
site of action.
Example 33. Clinical Pharmacokinetic Evaluation and Oral Bioavaiiability of L-
lysine-d-amphetamine Compared to Amphetamine Extended Release Products
Adderall XR® and Dexadrine Spansule® Used in the Treatment of ADHD
Table 47. Treatment Groups and Dosage for Clinical Pharmacokinetic Evaluation of
L-lysine-d-amphetamine Compared to Adderall XR® or Dexadrine Spansule®
Drug Treatment
Group Number of
Subjects Dose Dose
(mg) Dose (amphetamine
base)
L-lysine-
d-amphetamine A 10 1 x 25 mg
capsule 25 7.37
L-lysine-
d-amphetamine B 10 3 x 25 mg
capsules 15 22.1
Dexadrine
Spansule C 10 3 x 10 mg
capsules 30 22.1
Adderall XRW D 10 1 x 30 mg
capsules plus
1 x5 mg
capsule 35 21.9
[236] A clinical evaluation of the pharmacokinetics and oral bioavaiiability of L-
lysine-d-amphetamine in humans was conducted. L-lysine-d-amphetamine was
orally administered at doses approximating the lower (25 mg) and higher (75 mg)
end of the therapeutic range based on d-amphetamine base content of the doses.
Additionally, the higher dose was compared to doses of Adderall XR® (Shire) or
Dexadrine Spansule® (GlaxoSmithKline) containing equivalent amphetamine base
to that of the higher L-lysine-d-amphetamine dose. Treatment groups and doses are
summarized in Table 47. All levels below limit quantifiable (blq treated as zero for purposes of pharmacokinetic analysis.
[237] The concentrations of d-amphetamine and L-lysine-d-amphetamine intact
conjugate following administration of L-lysine-d-amphetamine at the low and high
dose for each individual subject as well as pharmacokinetic parameters are presented
in Tables 48-51. The concentrations of d-amphetamine following administration of

WO 2005/000334 PCT/US2004/017204
73
Adderall XR® or Dexadrine Spansule® for each individual subject as well as
pharmacokinetic parameters are presented in Tables 52 and 53, respectively.
Concentration-time curves showing L-Iysine-d-amphetamine intact conjugate and d-
amphetamine (ng/mL, Figures 52A and 53A and uM, Figures 52B and 53B) are
presented in Figures 52 and 53. Extended release of d-amphetamine from L-lysine-
d-amphetamine was observed for both doses and pharmacokinetic parameters (Cmax
and AUC) were proportional to dose when the lower and higher dose results were
compared (Table 43, 50 and 54; Figures 52 and 53). Significant levels of d-
amphetamine were not observed until one-hour post administration. Only small
amounts (1.6 and 2.0 percent of total drug absorption, respectively for 25 and 75 mg
doses; AUCinf - molar basis) of L-lysine-d-amphetamine intact conjugate were
detected with levels peaking at about one hour Table 49 and 51). The small amount
of intact conjugate absorbed was rapidly and completely eliminated with no
detectable concentrations present by five hours even at the highest dose.
[238] In a cross-over design (identical subjects received Adderall XR® doses
following a 7-day washout period), the higher L-lysine-d-arnphetamine dose was
compared to art equivalent dose of Adderall XR®. Adderall XR® is a once-daily
extended release treatment for ADHD that contains a mixture of d-amphetamine and
l-amphetamine salts (equal amounts of d-amphetamine sulfate, d-/l-amphetamine
sulfate, d-amphetamine saccharate, and d-/l-amphetamine aspartate). An equivalent
dose of extended release Dexadrine Spansule® (contains extended release
formulation of d-amphetamine sulfate) was also included in the study. As observed
in pharmacokinetic studies in rats, oral administration of L-lysine-d-amphetamine
resulted in d-amphetamine concentration-time curves similar to those of Adderall
XR® and Dexadrine Spansule® (Figures 54 and 55). The bioavailability (AUCinf) of
d-amphetamine following administration of L-lysine-d-amphetamine was
approximately equivalent to both extended release amphetamine products (Table
54). Over the course of twelve hours, typically the time needed for effective once-
daily treatment of ADHD, the bioavailability for L-lysine-d-amphetamine was
approximately equivalent to that of Adderall XR® (d-amphetamine plus l-
amphetamine levels) and over twenty percent higher than that of Dexadrine

WO 2005/000334 PCT/US2004/017204
74
Spansule®. Based on the results of this clinical study, L-lysine-d-amphetamine
would be an effective once-daily treatment for ADHD. Moreover, L-lysine-d-
amphetamine afforded similar pharmacokinetics in humans and animal models,
namely, delayed release of d-amphetamine resulting in extended release kinetics.
Based on these observations L-lysine-d-amphetamine should also have abuse-
resistant properties in humans.

Table 48. Individual Subject d-amphetamine Concentrations and Pharmacokinetic Parameters Following Oral Administration of a 25 mg Dose
of L-lysine-d-amphetamine to Humans.

Time
Hours Subject
102 Subject
103 Subject
105 Subject
107 Subject
110 Subject
112 Subject
113 Subject
116 Subject
117 Subject
120 Mean SD CV%
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0.5 0 0 0.625 0 0 0 0 0.78 0.769 0 0.2 0.4 162.1
1 4.29 2.95 8.67 3.36 8.33 1.1 10 10.5 14 3.15 6.6 4.2 63.6
1.5 10 12.7 16 13.8 21.4 3.94 24.7 19.5 24 15.1 16.1 6.5 40.3
2 16.3 18.4 17 21 25.9 9.29 30.9 23.6 30 21.7 21.4 6.6 30.8
3 16.5 19.6 16.7 26.1 27 17.7 30.2 23.5 27.6 28.9 23.4 5.3 22.7
4 23.9 18.8 14.1 24.5 30.1 17.9 33.2 21.2 24.7 25.3 23.4 5.7 24.3
5 21.2 18.9 14.6 21.6 22.6 17.2 27 20 20.2 24.2 20.8 3.5 16.9
6 21.8 18 12.5 21.6 23.7 15.7 25.8 18.2 20.3 20.5 19.8 3.9 19.6
7 18.9 15.8 12.1 17.8 20.6 14.5 26.6 21 18.3 21.8 18.7 4.1 21.9
8 19.3 16.6 10.4 17.9 20 14.2 25.7 13.6 18.8 20.1 17.7 4.2 24.1
10 18.8 13.6 9.8 15.3 19.3 13.7 22.4 15.1 15.3 15.9 15.9 3.5 22.1
12 15.8 12.6 6.92 11.5 15.8 11.2 17.9 12 13.7 15.2 13.3 3.1 23.6
16 13.4 10.5 6.56 9.53 14.3 10.7 12.5 10.3 10 13 11.1 2.3 20.5
24 8.03 5.81 2.65 4.9 5.8 5.9 6.57 6.13 4.52 5.45 5.6 1.4 25.1
48 1.57 1.36 0 1.26 0.795 1.44 1.24 1.23 0.864 0.586 1.0 0.5 46.1
72 0 0 0 0 0 0 0 0 0 0 0 0 0
Parameter Subject
102 Subject
103 Subject
105 Subject
107 Subject
110 Subject
112 Subject
113 Subject
116 Subject
117 Subject
120 Mean SD CV%
AUC0.,2h
(ng.h/mL) 204.0 177.4 140.4 204.9 242.7 152.4 284.6 199.2 225,5 223.3 205.4 42.5 20.7
AUC!ast (ng.h/mL 463.3 375.1 201.4 378.5 462.7 350.7 515.2 397.9 395.7 426.1 396.7 84.8 21.4
AUQnf (ng.h/mL) 486.7 397.1 233.5 398.8 472 374 532.5 416.4 407 432.2 415.0 80.1 19.3
Cmax (ng/mL) 23.9 19.6 17 26.1 30.1 17.9 33.2 23.6 30 28.9 25.0 5.6 22.3
Tmax (hours) 4 3 2 3 4 4 4 2 2 3 3.1 0.876 28.2
T1/2 (hours) 10.32 11.18 8.36 11.18 8.16 11.227 ; 9.68 10.43 9.06 7.22 9.68 1.43 14.7

WO 2005/000334 PCT/US2004/017204

Table 49. Individual Subject L-lysine-d-amphetamine Intact Conjugate Concentrations and Pharmacokinetic Parameters Following Oral
Administration of a 25 mg Dose of L-lysine-d-amphetamine to Humans.
Time
Hours Subject
102 Subject
103 Subject
105 Subject
107 Subject
110 Subject
112 Subject
113 Subject
116 Subject
117 Subject
120 Mean SD CV%
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0.5 4.1 5.5 10.0 0.0 3.6 0.0 9.2 9.6 8.9 0.0 5.1 4.2 82.0
1 9.2 11.2 15.2 12.5 9.1 2.7 20.1 10.5 10.8 10.9 11.2 4.5 39.7
1.5 4.0 4.4 6.1 7.5 3.6 6.2 6.6 2.8 4.2 8.4 5.4 . 1.8 34.1
2 2.1 1.4 2.5 2.9 1.9 4.0 2.3 0 1.7 3.1 2.2 1.1 48.8
3 0 0 0 0 0 0 0. 0 0 0 0 0 0
4 0 0 0 0 0 0 0 0 0 0 0 0 0
5 0 0 0 0 0 0 0 0 0 0 0 0 0
6 0 0 0 0 0 0 0 0 0 0 0 0 0
7 0 0 0 0 0 0 0 0 0 0 0 0 0
8 0 0 0 0 0 0 0 0 0 0 0 0 0
10 0 0 0 0 0 0 0 0 0 0 0 0 0
12 0 0 0 0 0 0 0 0 0 0 0 0 0
16 0 0 0 0 0 0 0 0 0 0 0 0 0
24 0 0 0 0 0 0 0 0 0 0 0 0 0
48 0 0 0 0 0 0 0 0 0 0 0 0 0
72 0 0 0 0 0 0 0 0 0 0 0 0 0
Parameter Subject
102 Subject
103 Subject
105 Subject
107 Subject
110 Subject
112 Subject
113 Subject
116 Subject
117 Subject
120 Mean SD cv%
AUCiast
(ng.h/mL) 9.18 10.95 16.31 10.68 8.583 5.439 18.51 10.77 12.35 10.41 11.32 3.74 33.1
AUQnf
(ng.h/mL) 10.62 11.64 17.66 12.65 9.759 - 19.56 - 13.3 12.83 13.50 3.40 25.2
Cmax (ng/mL) 9.18 11.2 15.2 12.5 9.05 6.18 20.1 10.5 10.8 10.9 11.56 3.80 32.9
Tmax (hours) 1 1 1 1 1 1.5 1 1 1 1 1.05 0.16 15.1
T|/2 (hours) 0.47 0.34 0.38 0.47 0.44 - 0.32 - 0.38 0.55 0.419 0.077 18.5

WO 2005/000334 PCT/US2004/017204

Table 50. Individual Subject d-amphetamine Concentrations and Pharmacokinetic Parameters Following Oral Administration of a 75 mg Dose
of L-lysine-d-amphetamine to Humans.
Time
Hours Subject
101 Subject
104 Subject
106 Subject
108 Subject
109 Subject
111 Subject
114 Subject
115 Subject
118 Subject
119 Mean SD CV%
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0.5 0 0.748 0.506 0 0 0.779 0.525 0 3 1.85 0.7 1.0 132.2
1 11.9 14.4 12.6 7.26 5.9 10.3 7.2 23.1 23 27.9 14.4 7.7 53.6
1.5 40.3 34.6 30.4 22.8 19.3 38.4 19 52.8 51.5 55.8 36.5 13.8 37.8
2 84.6 48.9 68.2 34.8 32.7 57.2 33.1 91.3 61.7 70.4 58.3 21.0 36.0
3 72.9 64.3 55.7 60.3 62.3 61.1 44.8 95.8 62.1 83.6 66.3 14.5 21.9
4 84.6 65.3 58.8 51.1 77.9 63.3 47.6 89.2 54.2 86 67.8 15.5 22.8
5 65 .55.6 60.2 74 83.9 59.1 56.9 77.7 54.9 82.8 67.0 11.5 17.2
6 71 53.5 49.4 51.5 78.3 50.8 55.1 68.8 52.9 64 59.5 10.2 17.1
7 53.8 55.7 52.9 69.5 73.1 52.9 55.9 71.2 45.1 74.6 60.5 10.5 17.4
8 63.7 40.3 47.3 45.7 72.2 46.5 54.2 61.1 44.3 66.2 54.2 10.9 20.2
10 43.7 41.7 37 58.4 67 44.3 48.4 68 34.1 55.9 49.9 11.9 24.0
12 46.4 26.1 36.7 37.4 49.9 32.4 37.1 54.1 34.5 45.1 40.0 8.6 21.6
16 35.4 22.2 25.7 48 44.9 24.3 28.9 44.7 31.7 34.5 34.0 9.2 27.1
24 16.4 11.4 14.9 13.2 18.4 16.8 20.5 21.7 15.7 18.1 16.7 3.1 18.8
48 2.74 2.14 4.17 2.73 3.75 4.81 2.81 4.26 3.36 3.4 0.9 25.9
72 0 0 0 1.07 0.661 0.687 1.49 0 0 0.553 0.4 0.5 120.2
Parameter Subject
101 Subject
104 Subject
106 Subject
108 Subject
109 Subject
111 Subject
114 Subject
115 Subject
118 Subject
119 Mean SD CV%
AUC0-i2h
(ng.h/mL) 666.2 525.9 531.6 570.3 704.8 545.6 513.7 790.9 523.4 742.8 611.5 104.5 17.1
AUClast
(ng.h/mL) 1266 918.7 1031 1257 1442 1123 1223 1549 1143 1417 1237.0 194.0 15.7
AUCinf
(ng.h/mL) 1301 948.3 1072 1278 1451 1133 1251 1582 1154 1425 1259.5 191.3 15.2
Cma, (ng/mL) 84.6 65.3 68.2 74 83.9 63.3 56.9 95.8 62.1 86 74.0 12.9 17.4
Tmax (hours) 4 4 2 5 5 4 5 3 3 4 3.9 1.0 25.5
Tt/2 (hours) 8.78 9.59 10.02 13.26 9.24 10.41 12.8 8.05 10.92 9.47 10.3 1.7 16.3

WO 2005/000334 PCT/US2004/017204

Table 51. Individual Subject L-lysine-d-amphetamine Intact Conjugate Concentrations and Pharmacokinetic Parameters Following Oral
Administration of a 75 mg Dose of L-Iysine-d-amphetamine to Humans.
Time
Hours Subject
101 Subject
104 Subject
106 Subject
108 Subject
109 Subject
111 Subject
114 Subject
115 Subject
118 Subject
119 Mean SD cv%
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0.5 10.4 22.6 6.92 10.3 0 9.21 7.88 14.5 87.8 35.5 20.5 25.6 124.7
1 48 40.5 29 41.5 21.2 30.8 23.4 127 88.9 80.1 53.0 34.6 65.2
1.5 28.4 15.7 16.1 20.3 26.5 19 12.7 38.7 28.6 38 24.4 9.2 37.5
2 8.87 5.53 4.91 9 18.1 5.62 6.29 12.1 9.75 11.3 9.1 4.0 44.0
3 2.15 1.29 1.76 1.82 10.6 0 2.31 2.57 1.73 1.73 2.6 2.9 111.6
4 0 0 1.09 0 4.65 0 1.53 1.01 0 0 0.8 1.5 176.9
5 0 0 0 0 0 0 0 0 0 0 0 0 0
6 0 0 0 0 0 0 0 0 0 0 0 0 0
7 0 0 0 0 0 0 0 0 0 0 0 0 0
8 0 0 0 0 0 0 0 0 0 0 0 0 0
10 0 0 0 0 0 0 0 0 0 0 0 0 0
12 0 0 0 0 0 0 0 0 0 0 0 0 0
16 0 0 0 0 0 0 0 0 0 0 0 0 0
24 0 0 0 0 0 0 0 0 0 0 0 0 0
48 0 0 0 0 0 0 0 0 0 0 0 0 0
72 0 0 0 0 0 0 0 0 0 0 0 0 0
Parameter Subject
101 Subject
104 Subject
106 Subject
108 Subject
109 Subject
111 Subject
114 Subject
115 Subject
118 Subject
119 Mean SD cv%
AUClast
(ng.h/mL) 51.2 44.2 32.0 43.7 50.4 30.9 29.8 102.1 110.8 86.1 58.1 30.2 52.0
AUCinf
(ng.h/mL) 52.5 45.0 33.0 44.9 52.3 34.2 31.4 102.9 111.7 87.0 59.5 29.9 50.2
Cmax (ng/mL) 48.0 40.5 29.0 41.5 26.5 30.8 23.4 127.0 88.9 80.1 53.6 34.1 63.6
TmaK (hours) 1 1 1 1 1.5 1 1 1 1 1 1.05 0.16 15.1
T1/2 (hours) 0.43 0.4 0.61 0.43 1.02 0.41 0.75 0.56 0.38 0.35 0.534 0.211 39.6

WO 2005/000334 PCT/US2004/017204

Table 52. Individual Subject d-amphetamine Concentrations and Pharmacokinetic Parameters Following Oral Administration of a 35 mg Dose
of Adderall XR® (equivalent to 75 mg dose of L-Iysine-d-amphetamine based on amphetamine base content) to Humans.

Time
Hours Subject
101 Subject
104 Subject
106 Subject
108 Subject
109 Subject
111 Subject
114 Subject
115 Subject
118 Subject
119 Mean SD cv%
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0.5 7.9 2.3 2.8 0.6 2.2 5.7 0 16 2.3 5.3 4.5 4.7 104.3
1 37.6 28.9 23.3 13.7 29.8 38.2 17.9 46.2 28.8 48.8 31.3 11.5 36.6
1.5 49.9 42.3 31.1 23,7 39.1 34.4 30.8 65.4 34.1 53 40.4 12.5 31.0
2 65.9 45.8 29.2 37.4 46.2 65.4 40 64.4 37 67.8 49.9 14.6 29.2
3 95.3 51.7 36.7 23.6 64.7 62.9 44.7 56.5 31.1 64.8 53.2 20.7 38.9
4 83.7 73.3 56.7 40 67 76.6 56.3 53.1 33.5 73.3 61.4 16.3 26.6
5 77.4 75.2 71.6 62.1 75.9 76.4 51.5 61.4 56.8 82.4 69.1 10.3 14.9
6 71.5 72.1 64 59.8 66.9 63.5 56.8 59.8 58.7 85.7 65.9 8.7 13.2
7 72.3 63.6 71 57.9 70.6 69.7 51.9 48.1 53.7 79.7 63.9 10.5 16.4
8 60.4 57.1 53.8 53 72 66.9 56.2 56.4 51.7 66.7 59.4 6.9 11.6
10 50.4 45.5 53 50.7 67.6 57.4 49.1 66.6 48 71.3 56.0 9.3 16.6
12 42.5 41.3 45.4 32.9 53.1 46 37.3 74.7 42.2 60.2 47.6 12.2 25.7
16 31.1 29.6 35.7 39 45.2 33.9 34.3 64.9 29 40.5 38.3 10.6 27.7
24 14.9 15.1 22.1 19.5 21.7 21.2 20.7 35.7 17.9 20.5 20.9 5.8 27.7
48 2.5 4.2 3.8 5.9 5.4 3.8 7.3 5.1 3.9 3 4.5 1.4 32.1
72 0 0.3 1 1 0.3 1.1 2.7 0.3 0 0 0.7 0.8 124.7
Parameter Subject
101 Subject
104 Subject
106 Subject
108 Subject
109 Subject
111 Subject
114 Subject
115 Subject
118 Subject
119 Mean SD CV%
AUC0-i2h
(ng.h/mL) 731.2 625.0 582.6 504.3 711.6 698.5 535.4 683.5 509.8 793.2 637.5 101.1 15.9
AUClast
(ng.h/mL) 1270 1230 1343 1269 1568 1436 1354 1920 1101 1520 1401.1 229.0 16.3
AUCinf
(ng.h/mL) 1301 1234 1358 1286 1571 1454 1418 1923 1164 1557 1426.6 218.9 15.3
Cmax (ng/mL) 95.3 75.2 71.5 62 75.9 76.5 56.8 74.7 58.8 85.8 73.3 11.9 16.3
Tmax (hours) 3 5 5 5 5 4 6 12 6 6 5.70 2.41 42.2
T1/2 (hours) 8.65 9.01 10.57 11.58 8.37 10.78 16.4 7.25 11.05 8.54 10.22 2.59 25.3

WO 2005/000334 PCT/US2004/017204

Table 53. Individual Subject d-amphetamine Concentrations and Pharmacokinetic Parameters Following Oral Administration of a 30 mg Dose
of Dexadrine Spansule® (equivalent to 75 mg dose of L-lysine-d-amphetamine based on amphetamine base content) to Humans.
Time
Hours Subject
102 Subject
103 Subject
105 Subject
107 Subject
110 Subject
112 Subject
113 Subject
116 Subject
117 Subject
120 Mean SD CV%
0 0 0 0 0 0 0 0 0 0 0 0 0 0
0.5 1.2 2.68 1.37 1.4 1.16 2.36 6.75 2.63 4.95 3.43 2.8 1.8 65.5
1 14.8 26.5 16.7 21.4 25.2 12.7 33.1 22.3 26 21.5 22.0 6.1 27.8
1.5 24.2 36.9 23.2 28.5 37.2 21.3 42.4 29.2 33.7 39.2 31.6 7.3 23.2
2 28.6 43.4 27.3 34.6 38.5 27.6 46.2 31.3 38.5 42 35.8 6.9 19.4
3 27.4 37.3 30.6 40.1 41.7 30.9 52 36.5 42.9 60.1 40.0 10.0 25.2
4 27.1 44.1 33.5 48.7 45.2 34.7 49.1 40.7 42.4 53.2 41.9 8.1 19.2
5 35.1 53 40.2 43.4 46.5 42.4 . 58,1 47 52.1 68.7 48.7 9.7 20.0
6 33.8 58.5 40.2 46.5 43.5 37.5 56.2 40 51 63 47.0 9.8 20.8
7 37.2 50.7 31.2 41.4 44.9 42 57.8 43.6 51.6 65.7 46.6 10.1 21.7
8 35.9 54.3 34.9 45 45 36 58.7 41.8 53.9 59.2 46.5 9.5 20.4
10 33.1 49.1 34.3 35.5 45 37 51.4 38.9 46.3 60.1 43.1 8.8 20.4
12 34 51 28.6 34.1 40.8 32.6 51.6 37.7 38.1 50.9 39.9 8.4 21.1
16 30.2 40.8 25.2 28 33 25.8 41 26.8 29.6 44.9 32.5 7.1 22.0
24 20.5 27.8 18.2 19.5 17.1 17.8 22.5 19.1 15.5 27.3 20.5 4.2 20.3
48 3.83 6.89 3.7 5.11 2.56 4.31 6.51 4.43 2.77 5.47 4.6 1.4 31.8
72 0.715 1.63 1 1.7 0 0.622 1.29 1.22 0 1.31 0.9 0.6 64.0
Parameter Subject
102 Subject
103 Subject
105 Subject
107 Subject
110 Subject
112 Subject
113 Subject
116 Subject
117 Subject
120 Mean SD CV%
AUCo.12h
(ng.h/mL) 356.2 539.8 366.4 444.3 480.8 387.0 591.4 436.5 512.8 634.2 474.9 94.7 19.9
AUClas[
(ng.h/mL) 1033 1517 966 1135 1065 1003 1473 1100 1048 1589 1193 236 19.8
AUCinf
(ng.h/mL) 1043 1544 983.5 1168 1097 1013 1495 1121 1085 1610 1216 238 19.5
Cmax (ng/mL) 37.2 58.5 40.2 48.7 46.5 42.4 58.7 47 53.9 68.7 50.18 9.74 19.4
Tmax (hours) 7 6 5 4 5 5 8 5 8 5 5.80 1.40 24.1
T1/2 (hours) 9.92 11.74 12.07 13.8 8.7 10.76 11.47 12.23 9.36 10.92 11.10 1.50 13.6

WO 2005/000334 PCT/US2004/017204

Table 54. Pharmacokinetic Parameters of Amphetamine Following Oral Administration of L-Iysine-d-amphetamine, Adderall XR® or Dexadrine
Spansule®.

Parameter Drug

L-lysine-
d-
amphetamine
25 mg Percent1 L-lysine-
d-
amphetamine
75 mg Percent Adderall
XR® Percent Dexadrine
Spansule® Percent
AUC0-i2h
(ng.h/mL) 205.4 33.6 611.5 100 637.5 104 474.9 78
AUClasl (ng.hymL) 396.7 31.5 1237 100 1401.1 113 1193 96
AUCinf (ng.h/mL) 415.0 32.9 1260 100 1427 113 1216 97
Cmax (ng/mL) 25.0 33.8 74 100 73.3 99 50.2 68
Tmax (hours) 3.1 79.5 3.9 100 5.7 146 5.8 149
T1/2 (hours) 9.68 94 10.3 100 10.22 99 11.1 108
Percent relative to L-lysine-d-amphetamine 75 mg dose

WO 2005/000334 PCT/US2004/017204

WO 2005/000334 PCT/US2004/017204
82
[239] It will be understood that the specific embodiments of the invention shown
and described herein are exemplary only. Numerous variations, changes,
substitutions and equivalents will occur to those skilled in the art without departing
from the spirit and scope of the invention. In particular, the terms used in this
application should be read broadly in light of similar terms used in the related
applications. Accordingly, it is intended that all subject matter described herein and
shown in the accompanying drawings be regarded as illustrative only and not in a
limiting sense and that the scope of the invention be solely determined by the
appended claims.
82

CLAIMS
1. A compound comprising amphetamine covalently attached to a chemical moiety, selected
from L-lysine-d- amphetamine, a salt thereof and an ester thereof.
2. The compound according to claim 1, which is L-lysine-d-amphetamine.
3. The compound according to claim 1, which is a salt or an ester of L-lysme-d-amphetamine.
4. The compound according to claim 3, which is L-lysine-d-amphetamine mesylate.
5. The compound according to claim 3, which is L-lysine-d-amphetamine hydrochloride.
6. The pharmaceutical composition in a form suitable for oral administration, comprising L-
lysine-d-amphetamine or a salt or ester thereof and a pharmaceutically acceptable additive

7. The pharmaceutical composition according to claim 6, consisting essentially of L-lysine-d-
amphetamine or a salt or ester thereof and a pharmaceutically acceptable additive.
8. The pharmaceutical composition according to claim 6 or 7, wherein said L-lysine-d-
amphetamine salt is L-lysine-d-amphetamine mesylate.
9. The pharmaceutical composition according to claim 6 or 7, wherein said L-lysine-d-
amphetamine salt is L-lysine-d-amphetamine hydrochloride.

10. The pharmaceutical composition according to any one of claims 6 to 9, wherein said form
suitable for oral administration is selected from the group consisting of a tablet, a capsule, a
caplet, an oral solution, a powder, a granule, a sachet, a syrup, an emulsion and a suspension.
11. The pharmaceutical composition according to any one of claims 6 to 10, wherein the
pharmaceutically acceptable additive comprises at least one additive selected from the group
consisting of diluents, binders and adhesives, lubricants, plasticizers, disintegrants, colorants,
bulking substances, flavorings, sweeteners, buffers, adsorbents and combinations thereof.
12. The use of L-lysine-d-amphetamine or of an ester or a salt thereof for preparing a
medicament for the treatment of ADHD.
83

13. The use according to claim 12, characterized in that the salt of L-lysine-d-amphetamine is
selected from L-lysine-d-amphetamine mesylate and L-lysine-d-amphetamine hydrochloride.
14. The pharmaceutical composition of claim 6, wherein said L-lysine-d-amphetamine or salt
thereof is in an amount sufficient to provide a pharmacologically effective amount to treat a
patient in need of amphetamine.
15. The pharmaceutical composition of claim 14, wherein said L-lysine-d-amphetamine or salt
thereof is in an amount sufficient to provide a therapeutically bioequivalent AUC when
compared to amphetamine alone, but in an amount insufficient to provide a Cmax which results in
euphoria.
94


The invention describes compounds compositions and methods or using the some comprising a chemical moiety
covalently attached to amphetamine. These compounds and compositions are useful for reducing or preventing abuse and overdose
of amphetamine. These compounds and compositions find particular use in providing an abuse-resistant alternative treatment for
certain disorders, such as attention deficit hyperactivity disorder (ADHD), ADD, narcolepsy, and obesity. Oral bioavailability of
amphetamine is maintained at therapeutically useful doses. At higher doses bioavailability is substantially reduced, thereby providing
a method of reducing oral abuse liability. Further, compounds and compositions of the invention decrease the bioavailability of
amphetamine by parenteral routes, such as intravenous or intranasal administration, further limiting their abuse liability.

Documents:

02683-kolnp-2005-abstract.pdf

02683-kolnp-2005-claims.pdf

02683-kolnp-2005-description complete.pdf

02683-kolnp-2005-drawings.pdf

02683-kolnp-2005-form 1.pdf

02683-kolnp-2005-form 13.pdf

02683-kolnp-2005-form 3.pdf

02683-kolnp-2005-form 5.pdf

02683-kolnp-2005-international publication.pdf

02683-kolnp-2005-priority document.pdf

2683-kolnp-2005-assignment.pdf

2683-KOLNP-2005-CORRESPONDENCE 1.1.pdf

2683-KOLNP-2005-CORRESPONDENCE.pdf

2683-kolnp-2005-correspondence1.1.pdf

2683-kolnp-2005-examination report.pdf

2683-kolnp-2005-form 13.pdf

2683-kolnp-2005-form 18.pdf

2683-kolnp-2005-form 3.pdf

2683-kolnp-2005-form 5.pdf

2683-kolnp-2005-gpa.pdf

2683-kolnp-2005-granted-abstract.pdf

2683-kolnp-2005-granted-claims.pdf

2683-kolnp-2005-granted-description (complete).pdf

2683-kolnp-2005-granted-drawings.pdf

2683-kolnp-2005-granted-form 1.pdf

2683-kolnp-2005-granted-specification.pdf

2683-KOLNP-2005-OTHERS 1.1.pdf

2683-kolnp-2005-reply to examination report.pdf

2683-KOLNP-2005.pdf


Patent Number 250464
Indian Patent Application Number 2683/KOLNP/2005
PG Journal Number 02/2012
Publication Date 13-Jan-2012
Grant Date 04-Jan-2012
Date of Filing 23-Dec-2005
Name of Patentee NEW RIVER PHARMACEUTICALS INC
Applicant Address THE GOVERNOR TYLER, 1881 GROVE AVENUE, RADFORD, VA
Inventors:
# Inventor's Name Inventor's Address
1 KRISHNAN SUMA 1210 DRAPER ROAD, BLACKSBURG, VA 24060
2 OBERLENDER ROB 914 ALLENDALE COURT, BLACKSBURG, VA 24060
3 LAUDERBACK CHRISTOPHER 465 BRUSH MOUNTAIN ROAD, BLACKSBURG, VA 24060
4 MICKLE TRAVIS 13700 COOPER CROFT RUN, NW, APT. B, BLACKSBURG, VA 24060
5 BISHOP BARNEY 5003 FLEMING DRIVE, ANNADALE, VA 22003
6 MONCRIEF JAMES SCOTT 615 CHARLES STREET, CHRISTIANSBURG, VA 24073
PCT International Classification Number A61K 38/00
PCT International Application Number PCT/US2004/017204
PCT International Filing date 2004-06-01
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 60/473,929 2003-05-29 U.S.A.
2 60/567,801 2004-05-05 U.S.A.