CA2802733C - Prodrugs of nh-acidic compounds: ester, carbonate, carbamate and phosphonate derivatives - Google Patents

Prodrugs of nh-acidic compounds: ester, carbonate, carbamate and phosphonate derivatives Download PDF

Info

Publication number
CA2802733C
CA2802733C CA2802733A CA2802733A CA2802733C CA 2802733 C CA2802733 C CA 2802733C CA 2802733 A CA2802733 A CA 2802733A CA 2802733 A CA2802733 A CA 2802733A CA 2802733 C CA2802733 C CA 2802733C
Authority
CA
Canada
Prior art keywords
compound
formula
optionally substituted
aryl
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CA2802733A
Other languages
French (fr)
Other versions
CA2802733A1 (en
Inventor
Laura Cook Blumberg
Julius F. Remenar
Orn Almarsson
Tarek A. Zeidan
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Alkermes Pharma Ireland Ltd
Original Assignee
Alkermes Pharma Ireland Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Alkermes Pharma Ireland Ltd filed Critical Alkermes Pharma Ireland Ltd
Publication of CA2802733A1 publication Critical patent/CA2802733A1/en
Application granted granted Critical
Publication of CA2802733C publication Critical patent/CA2802733C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/454Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. pimozide, domperidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/545Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/548Phosphates or phosphonates, e.g. bone-seeking
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/18Antipsychotics, i.e. neuroleptics; Drugs for mania or schizophrenia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D215/00Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems
    • C07D215/02Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom
    • C07D215/16Heterocyclic compounds containing quinoline or hydrogenated quinoline ring systems having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen atoms or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D215/20Oxygen atoms
    • C07D215/22Oxygen atoms attached in position 2 or 4
    • C07D215/227Oxygen atoms attached in position 2 or 4 only one oxygen atom which is attached in position 2

Abstract

The invention provides a method of sustained delivery of a lactam, imide, amide, sulfonamide, carbamate or urea containing parent drug by administering to a patient an effective amount of a prodrug compound of the invention wherein upon administration to the patient, release of the parent drug from the prodrug is sustained release. Prodrug compounds suitable for use in the methods of the invention are labile conjugates of parent drugs that are derivatized through carbonyl linked prodrug moieties. The prodrug compounds of the invention can be used to treat any condition for which the lactam, imide, amide, sulfonamide, carbamate or urea containing parent drug is useful as a treatment.

Description

PRODRUGS OF NH-ACIDIC COMPOUNDS: ESTER, CARBONATE, CARBAMATE
AND PHOSPHONATE DERIVATIVES
BACKGROUND OF THE INVENTION
(i) Field of the Invention.
The present invention relates to prodrugs of lactam, amide, imide, sulfonamide, carbamate, urea, benzamide, and acylaniline containing pharmacophores.
(ii) Background of the Invention.
Drug delivery systems are often critical for the safe and effective administration of a biologically active agent. Perhaps the importance of these systems is best realized when patient compliance and consistent dosing are taken under consideration. For instance, reducing the dosing requirement for a drug from four-times-a-day to a single dose per day would have significant value in terms of ensuring patient compliance and optimizing therapy.
Optimization of a drug's bioavailability has many potential benefits. For patient convenience and enhanced compliance it is generally recognized that less frequent dosing is desirable. By extending the period through which the drug is released, a longer duration of action per dose is expected. This will then lead to an overall improvement of dosing parameters such as taking a drug once a day where it has previously required four doses per day or dosing once a week or even less frequently when daily dosing was previously required. Many drugs are presently dosed once per day, but not all of these drugs have pharmacokinetic properties that are suitable for dosing intervals of exactly twenty-four hours.
Extending the period through which these drugs are released would also be beneficial.
One of the fundamental considerations in drug therapy involves the relationship between blood levels and therapeutic activity. For most drugs, it is of primary importance that serum levels remain between a minimally effective concentration and a potentially toxic level. In pharmacokinetic terms, the peaks and troughs of a drug's blood levels ideally fit well within the therapeutic window of serum concentrations. For certain therapeutic agents, this window is so narrow that dosage formulation becomes critical.

In an attempt to address the need for improved bioavailability, several drug release modulation technologies have been developed. For example, poorly soluble 5,5 diphenylimidazolidine-2,4-diones have been derivatized into phosphate ester prodrugs to improve solubility. (Stella et at., U.S. Patent No. 4,260,769, 1981). Enteric coatings have been used as a protector of pharmaceuticals in the stomach and microencapsulating active agents using proteinaceous microspheres, liposomes or polysaccharides have been effective in abating enzymatic degradation of the active agent. Enzyme inhibiting adjuvants have also been used to prevent enzymatic degradation.
A wide range of pharmaceutical formulations provide sustained release through microencapsulation of the active agent in amides of dicarboxylic acids, modified amino acids or thermally condensed amino acids. Slow release rendering additives can also be intermixed with a large array of active agents in tablet formulations.
While microencapsulation and enteric coating technologies impart enhanced stability and time-release properties to active agent substances these technologies suffer from several shortcomings. Incorporation of the active agent is often dependent on diffusion into the microencapsulating matrix, which may not be quantitative and may complicate dosage reproducibility. In addition, encapsulated drugs rely on diffusion out of the matrix or degradation of the matrix, or both, which is highly dependent on the chemical properties and water solubility of the active agent. Conversely, water-soluble microspheres swell by an infinite degree and, unfortunately, may release the active agent in bursts with limited active agent available for sustained release. Furthermore, in some technologies, control of the degradation process required for active agent release is unreliable. For example, because an enterically coated active agent depends on pH to release the active agent and pH and residence time varies, the release rate is difficult to control.
Several implantable drug delivery systems have utilized polypeptide attachment to drugs. Additionally, other large polymeric carriers incorporating drugs into their matrices are used as implants for the gradual release of drug. Yet another technology combines the advantages of covalent drug attachment with liposome formation where the active ingredient is attached to highly ordered lipid films.
However there is still a need for an active agent delivery system that is able to deliver certain active agents which have been heretofore not formulated or difficult to formulate in a sustained release formulation for release over a sustained period of time and which is convenient for patient dosing.
2 There is a generally recognized need for sustained delivery of drugs that reduces the daily dosing requirement and allows for controlled and sustained release of the parent drug and also avoids irregularities of release and cumbersome formulations encountered with typical dissolution controlled sustained release methods.
SUMMARY OF THE INVENTION
The present invention accomplishes this by extending the period during which a lactam, amide, imide, sulfonamide, carbamate, urea, benzamide, acylaniline, and cyclic amide containing parent drug is released and absorbed after administration to the patient and providing a longer duration of action per dose than the parent drug itself In one embodiment, the compounds suitable for use in the methods of the invention are derivatives of lactam-, amide-, imide-, sulfonamide-, carbamate-, urea-, benzamide-, acylaniline-, and cyclic amide -containing parent drugs that are substituted at the amide nitrogen or oxygen atom with prodrug moieties. Preferably, the prodrug moieties are hydrophobic and reduce the polarity and solubility of the parent drug under physiological conditions.
In one embodiment, the invention provides a prodrug compound of Formula I, II
or III:
A
\
N-Ri A\N 0 Ri X _______ ( 1X¨(/ 11 A¨S¨N/

B R1 B 0 \
Formula I Formula II Formula III
or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof;
wherein A and B together with the ¨N(C=X)- or ¨(502)N- to which they are attached forms a parent drug;
X is ¨S- or ¨0-;
R1 is selected from C(0)0R20, C(0)R20, -C(0)NR20R21, -P03MY, -P(0)2(0R20)M and -P(0)(0R20)(0R21);
wherein each R20 and R21 is independently selected from hydrogen, aliphatic, substituted aliphatic, aryl or substituted aryl;
3
4 Y and M are the same or different and each is a monovalent cation; or M and Y
together is a divalent cation.
The invention further provides a method for sustained delivery of a parent drug by the administration of a conjugate of the parent drug with a labile moiety, wherein the conjugate is represented by Formula I.
BRIEF DESCRIPTION OF THE DRAWING
The foregoing and other objects, features and advantages of the invention will be apparent from the following more particular description of preferred embodiments of the invention, as illustrated in the accompanying drawing in which like reference characters refer to the same parts throughout the different views. The drawing is not necessarily to scale, emphasis instead being placed upon illustrating the principles of the invention.
The Figure: Plasma concentration of dehydroaripiprazole after PO
administration of (10 mg/Kg) compound 323, 331 and dehydroaripiprazole to rats.
DETAILED DESCRIPTION OF THE INVENTION
In one embodiment, the invention provides a prodrug compound of Formula I, II
or III:
A
\
N-Ri A\N 0 Ri X _______ ( /X¨(/ 11 A¨S¨N/

B R1 B 0 \
Formula I Formula II Formula III
or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof;
wherein A and B together with the ¨N(C=X)- to which they are attached forms a parent drug;
X is ¨S- or ¨0-;
R1 is selected from C(0)0R20, C(0)R20, -C(0)NR20R21, -P03MY, -P(0)2(0R20)M and -P(0)(0R20)(0R21);
wherein each R20 and R21 is independently selected from hydrogen, aliphatic, substituted aliphatic, aryl or substituted aryl;

Y and M are the same or different and each is a monovalent cation; or M and Y
together is a divalent cation.
The invention further provides a method for sustained delivery of a parent drug by the administration of a conjugate of the parent drug with a labile moiety, wherein the conjugate is represented by Formula I.
In a preferred embodiment, R1 is selected from ¨C(0)0-CH2-0C(0)R21 or ¨C(0)NH-CH2-0C(0)R21 or ¨C(0)0-CH2-0C(0)N(R21)2. In a preferred embodiment, R1 is selected from ¨C(0)0-CH(CH3)-0C(0)R21 or ¨C(0)NH-CH(CH3)-0C(0)R21 or ¨C(0)0-CH(CH3)-0C(0)N(R21)2. In a preferred embodiment, R1 is selected from ¨C(0)0-CH(C6H5)-OC(0)R21 or ¨C(0)NH-CH(C6H5)-0C(0)R21 or ¨C(0)0-CH(C6H5)-0C(0)N(R21)2. In a more preferred embodiment, R21 is alkyl, substituted alkyl, alkenyl, or substituted alkenyl.
In a preferred embodiment, R1 is selected from ¨C(0)0R21 or ¨C(0)NHR21 or ¨
C(0)N(R21)2. In a more preferred embodiment, R21 is alkyl, substituted alkyl, alkenyl, or substituted alkenyl.
In one embodiment, the compounds of the invention having Formula I, II or III
are less soluble, and are preferably at least an order of magnitude less soluble, as compared to the parent drug from which they were derived. In one embodiment, the prodrugs of Formulas I
has an aqueous solubility of less than about 0.5 mg/ml, preferably less than about 0.1 mg/mL, preferably less than about 0.01 mg/mL, preferably less than about 0.001 mg/mL, preferably less than about 0.0001 mg/mL and even more preferably less than about 0.00001 mg/ml when solubility is measured in a phosphate buffer (pH 7.4) at room temperature.
In a preferred embodiment, a compound of the invention provides sustained delivery of the parent drug over hours, days, weeks or months when administered, for example, orally or parenterally, to a subject. For example, the compounds can provide sustained delivery of the parent drug for at least 8, 12, 24, 36 or 48 hours or at least 4, 7, 15, 30, 60, 75 or 90 days or longer. Without being bound by a theory, it is believed that the compounds of the invention form an insoluble depot upon parenteral administration, for example subcutaneous, intramuscular or intraperitoneal injection. In one embodiment a prodrug of the invention may further comprise a sustained release delivery system for providing additional protection of the prodrug from enzymatic or chemical degradation.
In another embodiment, the invention provides a method for sustained delivery of a parent lactam, amide, imide, sulfonamide, carbamate, urea, benzamide, or acylaniline containing drug to a subject in need thereof Each of these groups comprises an amidic N-H
group. The method comprises administering to the subject an effective amount of a prodrug
5 formed by substituting on the NH group a labile, hydrophobic prodrug moiety wherein the prodrug has reduced solubility under physiological conditions compared to the parent drug and provides for longer sustained therapeutic levels of the parent drug following administration than observed levels following administration of the parent drug. In a preferred embodiment, the amidic N-H group has a pKa of about 5 to about 22, preferably about 5 to about 21, and preferably about 5 to about 20.
In a preferred embodiment, R1 is selected from Tables 1-5.
Table 1 ) Vt 2 2 2 ? ) ) Z 2 ?

z z .
o G (7 (D¨p-1-II
0 Ca2+ e e I

(D¨P--µ111. µ112.
/ (0 0
6 0_412-OL4 /(112-<' /\ 4µtII.0 /< ZIL
\ ( ( (\ 0 vvvvIJvvv eKck0 0 (''KzkO
0 0 e)lck0 0><H

wvv vv 0>

A) 7 A)5
7 ¨
i 7 esss /0 3 /07 ( )4 / \ / \
\/¨\ i , /
\ /
/6 \

.)7y i ) ) NI)/ N).y N)-y )==y )-y )-y N N N
)) 5 )) 7 )) 13 ('s1k1)-VN
µ'13 kl)-V
))5 ))13 ))7 N N
))15 ))17 ))19
8 N)..V N
)) 21 ))23 ))9 N)cisc o o ))11 0 \
LO
/ H0-p1-1-0 /- g el-(--)-0
9 1 0¨P-1- 0¨P-1-0 ( H
7 0 ( 11 0¨P-1-'II

\

\
0----- 0----?a? JK----?a?

C /Wo \
C) C) 0----- "
rssj.
rsrsjs 0 40 Opt\ 0 0___0\

\

Ce'ss ors 0 a o<

0.pr 0,,rf orr a a orr 0,5x orr o'rs Os.r C),o'xr O'rr /LO /LO
or'r NHO NHO
VV

VV
N4 Ne N4 N4 Ne N4 Ne /LO /LO /LO

/LO ....,0 ())co0 /07 rPrr A ) 5 , A ) 3 , A) 7 rPrr ( )4 ,vvvs -[/¨\-/

N., ,L0 o o Table 2 )y\? Dy\ rµ\

yt2Z2? \ rt2222 c'22-o SO
/ )\
( \O

<' ( /1112. /\

/ ilL 0 \ (0 0 0 \ HIL

vvvvN,wv ('Kck0 0 (,1ziO

JVVVL, JVVVL, JVVVL
('/KEk0 OX

K2LC) 11 0><H

A)7 A)5 .r r, sos eNsi , 7 rssr A)3 A)7 ( )4 / \ /

SOS ov Table 3 Nil ) ) N)V N)V N )isssss5 ) /\) I

)==y )-N).../ N Ny ))13 ))5 ))13 ))7 N N N
))15 ))17 ))19 N)Y
N N
))21 ))23 ))9 o )-y ) o )11 Table 4 \
0---- 0-----\?2? JK----<\

o 0 0 0---<\
, rfss 0 0 0----?: 0 0---?0 O'rr 0'.

a0 0 0, a a0 err err orr oss ors cer cer 0.pr ,L ,L
ces ,õ0 o õ,0 o ,L0 ,L0 ,L0 /LO /LO /LO
N4 Ne N4 Ne 0 ()(7 (1o0 A)7 rPrr A)5 , o0 0 (1/11 ( \-r--A ) 3 rPrr A) 7 prrf ( ) 4 vvvs -__---o /
0 < o o rsjsress -111'1,, N, /L /LO /LO

o Lo 1 5 \o O¨P-r I 5 e 1 , Ca2+ G

e 1 , 0¨P¨r 0¨P--= 0¨n+ 0 Table 5 ..,,(1.00......-µ .,.õ(100....õ...õ.õ,(2?2, M -o 0 ooµ
oo,,,,..)2, 0 0)2L

O0µ
00µ \ 0 o\
i 4 0 A>j J 0 /t/t1) i 4 J . 0 O 0 0) O0µ
00µ \ 0 0)IZL

ill O0µ 00µ 0 2-kl) 6 i 0 j NyOO

Ny00\ 0µ

Wherein each j is independently 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, or 27.
PRODRUGS OF LACTAM, CYCLIC UREA, IMIDE, CARBAMATE CONTAINING
PHARMACOPHORES
In one embodiment, compounds of the present invention are represented by Formula IV and V as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts co-crystals and solvates thereof:
X X
) ________________ /
Ri µ'\
x1 i4 ..- X3 X ss X3 rt Formula IV Formula V
wherein _________ represents a single or double bond;
X and R1 are as defined above;
Each X1, X25 and X3 is independently selected from absent, -S-, -0-, -S(0)-, -S(0)2-, -N(Rio)-, -C(0)-, -C(0R10)(R11)-5 -[C(Rio)(Rii)k-, -C(Rio)(Rii)=C(Rio)(Rii)-;
wherein v is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10;
wherein each R10 and R11is independently absent, hydrogen, halogen, aliphatic, substituted aliphatic, aryl or substituted aryl; alternatively two R10 and R11 together with the atoms to which they are attached may form an additional optionally substituted, 3, 4, 5, 6 or 7 membered ring; and t is 0, 1, 2 or 3.
In one embodiment, compounds of the present invention are represented by Formula VI or VII as illustrated below, and the geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
F2 .F2 R1 õLI Li I
(R3)q =-.2 (R3)CI ,U2 X
Fi _______________ Ai J_ __ (R4)In Fi __ _ 1, -LN .._/)(' =
1....-........ .........,N

Formula VI Formula VII
wherein represents a single or double bond;
X5 X15 X2 and R1 are as defined above;
semicircle represents an optionally substituted cycloalkyl, cycloalkenyl, heterocyclyl or aryl containing one, two or three rings;
each F1 and F2 is independently selected from absent and R5-A-Cyl-B-D-;
wherein, A is selected from absent, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, -S-5 -0-, ¨S(0)-5 ¨S(0)2-, -S[C(R3o)(R31)L-5 ¨
S(0)[C(R30)(R3i)L-5 ¨S(0)2[C(R30)(R31)]u-, -0[C(R30)(R3i)L-, -N(R30)-5 -N(R30)[C(R31)(R32)b-, -[C(R3o)(R3i)L, -C(0)[C(R30)(R3i)b-;
wherein each u is independently 1, 2, 3, 4, 5, 6 or 7;
Cyi is absent or an optionally substituted cycloalkyl, optionally substituted cycloalkenyl, optionally substituted heterocyclyl, optionally substituted aryl or optionally substituted heteroaryl;
B is absent, or a linker;
D is selected from absent, -0-, -NR335 -C(R34)( R35)-5 ¨S-5 ¨S(0)-5 ¨S(0)2-5 -C(0)-;
Each G1 and G2 is independently selected from absent, -S-5 -0-, ¨S(0)-5 ¨S(0)2-, -SC(R40)(R41)-, ¨S(0) C(R40)(R41)-, ¨S(0)2C(R40)(R41)-5 -C(0)-5 -C(0R40)(R41)-5 -0C(R40)(R41)-5 -N(R40)-5 -C(R40)=C(R41)-, -N(R40)-C(R41)(R42)-, -[C(R40)(R41)]u-;

Each R3, R4, R5, R30, R315 R32 R335 R345 R35, R405 R415 and R42 is independently selected from absent, hydrogen, halogen, -0R10, -SRio, -NRioRii-, -C(0)R10, optionally substituted aliphatic, optionally substituted aryl or optionally substituted heterocyclyl;
Alternatively, two R3 groups together or two R4 groups together or one R3 group with one R4 group together forms an optionally substituted ring;
m and q are independently selected from 0, 1, and 2.
In a preferred embodiment, G2 is selected from ¨N- or ¨C(Rio)-=
In a preferred embodiment, the R5 moiety is an aryl or heteroaryl group selected from:
// /\ . .
R
loo ______ ) 5 ¨ _________ CI CI CI CI
Rioi \ R N __ 1 1//¨ ?
CI
CI
= ______________________________________________ = X ,N

CI Roo Rloo N ________________________________________________________________ , Rioo \ 5 Rioo4 \
\ 5 Rioo / \ 5 1\1 Rioo \ 5 ) Rioi, _____ / N
R1oK

/

R1 ____________________________________________ /01 / R101 );(-N /
R1001 \ 5 Rloo __ /
i Rio Ric'c'll __ ) /
Ric'Z/
S --i Rloo / ____ ? 1....7 N ,s _( 'Ns S
\ ______________________________ p / N=b..........
/
-100 Rioi --- S -- Rioo \ R100 // __ ) 5 N= R101 Rloo Rioi ---- N ---- Rio 1 (1) __ (1) H Rioi a/VVVlo a/VVVlo a/V1/1/1.0 N' S N' 0 NN' N.R103 _________ Rloo Rloo Rloo wherein R100 R101, and R103 are independently selected from hydrogen, halogen, optionally substituted C1-C8 alkyl, optionally substituted C2-C8 alkenyl, optionally substituted C2-C8 alkynyl, optionally substituted C3-C8 cycloalkyl, optionally substituted C1-C8 alkoxy, optionally substituted Ci-C8 alkylamino and optionally substituted C1-C8 aryl.
In a preferred embodiment, Cyi is selected from:
Rloo 1 ) Rio<\ Rioo /-1¨\
i ,-----µ
1 oe \ __________________________________________________ 'a2(N¨/ (212(\=
Rloo N R101 r1-\
¨N / \ -1\1' N1 ¨N\ 2 \ 71 i 3 r ?.2, N-......-,100 vN-... R100 ... - ¨N/ 1 rN\ ___ /0 ,a2( \_0 ....,_ NI:\)222., 1 N / \
v N . v N __v N __\
scs5 /-1-\ <??
R100 /i¨NRico N
Rioi e -Ri oi ssss\ R

ssCN)2?2, Y22z, Rioo/ N

\
R1o1 N
In a preferred embodiment, the bivalent B is a direct bond, a straight chain alkyl, C1-C10 alkenyl, C1-C10 alkynyl, C1-C10 alkoxy, alkoxyCi-Cioalkoxy, alkylamino, alkoxyCi-Cioalkylamino, Ci-Cio alkylcarbonylamino, alkylaminocarbonyl, aryloxyCi-Cioalkoxy, aryloxyCi-Cioalkylamino, aryloxyCi-Cioalkylamino carbonyl, Ci-Cio-alkylaminoalkylaminocarbonyl, Ci-Cio alkyl(N-alkyl)aminoalkyl-aminocarbonyl, alkylaminoalkylamino, alkylcarbonylaminoalkylamino, alkyl(N-alkyl)aminoalkylamino, (N-alkyl)alkylcarbonylaminoalkylamino, alkylaminoalkyl, alkylaminoalkylaminoalkyl, alkylpiperazinoalkyl, piperazinoalkyl, alkylpiperazino, alkenylaryloxyC 1 -C 1 Oalkoxy, alkenylarylaminoCl-Cioalkoxy, alkenylaryllalkylaminoCi-Cioalkoxy, alkenylaryloxyCi-Cioalkylamino, alkenylaryloxyC i-Cioalkylaminocarbonyl, piperazinoalkylaryl, heteroarylCi-Cioalkyl, heteroary1C2-Cioalkenyl, heteroary1C2-Cioalkynyl, heteroarylCi-Cioalkylamino, heteroarylCi-Cioalkoxy, heteroaryloxyCi-Cioalkyl, heteroaryloxyC2-Cioalkenyl, heteroaryloxyC2-Cioalkynyl, heteroaryloxyCi-Cioalkylamino and heteroaryloxyCl-Cioalkoxy.
In one embodiment, compounds of the present invention are represented by Formula VIII or VIIIA as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
F2 F2 Ri .02 X
D
q(R3) (R4)m q(R3) ________ k',4im lõ

Formula VIII Formula VIIIA

wherein R1, R35 R45 G15 G25 X, F25 m and q are as defined above.
In a more preferred embodiment, compounds of the present invention are represented by Formula IX or X as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

/
e......õ,...--) (R3)q)............ ___________ 0 e.......õ,..--) /Ri N
\RI (R3)q- 1 / 0 N
Formula IX Formula X
wherein R15 R3, F25 and q are as defined above.
In a preferred embodiment a compound is selected from Table IX-X. A more preferred embodiment is a compound from Table IX-X wherein R1 is selected from Tables 1-4.
Table IX-X
No Structure No Structure 411' N 0 /N
N=----Ri 0 O'R1 N
CF3 0-----\__Nr\N \--/

/ -Ri Ri 3 . 10 N N
1. .
el * N N
el N---=--( /N--k Ri 0 ) /i R 11 Ri H /

--_,rH
= N
N----"Nõ....- 0-- N 410N
CI fi NN.......,...N..õ0-- 01111 CI

\ 0 )--NH (:) ---NH
,0..._N .
N_...y R1 . NN
CI
CI .
6 . F 13 0 F
N
I N
I

/N--k ---*
Ri 0 N

/*N I.
N
= N
= N
1.1 N----=( 5 N--k Ri/ 0 F F
In a more preferred embodiment, prodrugs of domperidone are disclosed.
(Formula 4 and 11 from Table IX-X). A more preferred embodiment is a compound of Formula 4 from Table IX-X, wherein R1 is selected from Table 1. In a more preferred embodiment, a 5 compound of Formula 4 from Table IX-X, wherein R1 is selected from Tables 2-4 is disclosed.
In a more preferred embodiment, prodrugs of droperidol are disclosed. (Formula and 13, from Table IX-X). In a more preferred embodiment, a compound of Formula 6 from Table IX-X wherein R1 is selected from Table 1 is disclosed. A more preferred embodiment is a compound of Formula 6 from Table IX-X wherein R1 is selected from Tables 2-4.
In a more preferred embodiment, prodrugs of pimozide are disclosed. (Formula 7 and 14 from Table IX-X). In a more preferred embodiment, a compound of Formula 7 from Table IX-X wherein R1 is selected from Table 1 is disclosed. In a more preferred embodiment, a compound of Formula 7 from Table IX-X wherein R1 is selected from Tables 2-4 is disclosed.
In another embodiment, compounds of the present invention are represented by Formula XI or XII as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Ri q(R3) q(R3) xI

r-all1( ¨TrY(R4)m ' N ,,, 's- / \ s'= /N
G1 Ri G1 Formula XI Formula XII
wherein R1, R3, R45 X, F1, G1, G25 m and q are as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIA or XIIA as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
/ ____________ \ Rio D........zy R3)q Rio / -5/(R3) q \ ,D--...../' R5¨A¨G3 G4 __ ( ir 1 3 24 __ ( il' I
I \-li R11 (R2)P X20,., (112)P
Nv\K
(R4)11-, Rr (R4).-, 70 0 Ri Formula XIA Formula XIIA
wherein R15 R35 R45 R55 R105 R115 A, D, m, and q are as defined above;

R2 is selected from absent, hydrogen, halogen, -0R10, -SRI , -NRioRii-, optionally substituted aliphatic, optionally substituted aryl or aryl or optionally substituted heterocyclyl;
r is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11;
each G3 and G4 is independently selected from -N-, -C(Rio)-[C(Rio)(Ria-, wherein a is 0, 1 or 2;
X20 is -C(R10)- or -N-; and p is selected 0, 1, 2 or 3.
In another embodiment, compounds of the present invention are represented by Formula XIB or XIIB as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
_____________________________________________________ \ RioDy(R3) q _______________ \ RioD r y(R3) q A-N
A-N
R5 \-1-/N (1<
Rii R5 \-2 1 (r Rii (RDP
(RDP N
N (R4)m R1 (R4)m 0 Rr Formula XIB Formula XIIB
wherein R1, R2, R3, R4, R5, R10, R115 A, D, m, p and q are as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIC or XIIC as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Cl Cl NCN--(CH2*-0 N N-(CH2*-0 *
N
,0 RI
Formula XIC Formula XIIC
wherein R1, is as defined above; and w is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11.

In another embodiment, compounds of the present invention are represented by Formula XID or XIID as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
(R3)q (R3)q / __ \ 1 rD / __ \ r R5¨A¨G3 G4-B R5¨A¨G3 G4-B D

-----R

(RAI (RAI
X----1( X--( 0 o¨R1 Formula XID Formula XIID
wherein, X1, R1, R25 R35 R55 A, B, D, G35 G45 p5 ch R10 and R11 are as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIE or XIIE as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Rk ioRii Rk ( loRii R3)q R
( 3)cl Al _________________________________________________________________ 1 r D = -- 9 \

,G3--/ X__ J
X/ A (R,4). X,N/ A (R4)m-A

0 \
\ /
R2 R2 Ri Formula XIE Formula XIIE
wherein, X, R1, R25 R35 R45 A, D, G35 G45 m, q, r, R10 and R11 are as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIF or XIIF as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

R10. iRrD
C) 11 Cl R Cl Y 10. iR11 4) *
, (--N
N-R, N
/1110 , N--X'k 0 X'-i\I, /0 Formula XIF Formula XIIF
wherein, X, R1, R2, D, r, R10 and R11 are as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIG or XIIG as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
. rN
Cl 11110 , , (NN
N . Cl W
/ NI
s 'N
-N
N, 0 \
0 Ri Ri Formula XIG Formula XIIG
wherein R1, is as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIH or XIIH as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
.---R

X--<
R5-A-G3-1-j /--\G4'B ,D 0 N, N

, Ri (R) p (R2) \ P
Formula XIH Formula XIIH
wherein, X, R1, R2, R5, A, D, G3, G4 and p, are as defined above.

In another embodiment, compounds of the present invention are represented by Formula XI-I or XII-I as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Rp, 0 N¨Ri ON
/¨\ /--\
N / N . N/ N 11/
5*
Formula XI-I Formula XII-I
wherein R1, is as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIJ or XIIJ as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
/

\ (Rio R3) q Rlo ,yR3) ci ICX-......(...
R5¨A¨N R5¨A¨I¨\\-12 __ (12 X kk iir i \¨lj Rii N
(RDP i (RAI
' N N)v , (R4)m Rr (R4)m 0, Formula XIJ Formula XIIJ
wherein, X, R1, R2, R3, R4, R5, A, D, G3, G4, p, R10 and R11 are as defined above.
In another embodiment, compounds of the present invention are represented by Formula XIK or XIIK as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

1'W N , O.
I
..Ncyki^N(:) N , 141 KNoNNO-R1 Formula XIK Formula XIIK
wherein R1, is as defined above.
In a preferred embodiment a compound is selected from Table XI-XII. A more preferred embodiment is a compound from Table XI-XII, wherein R1 is selected from Table 1-4.
Table XI-XII
XI-1 Cl rN/NN70 XII- 1 Cl rNNO
Cl = NN) . Cl ,NJ
Rf N--N \

XI-2 Cl ri\y--,,Nzo XII-2 0 rN,0 ci = NN
= Cl ik N N.) Ri--N / N\ /

rN XII-3 ('N40.
N
N Cl N Cl W
/
WI / I
S'N S'N
N, ¨N
0 Ri 0\
Ri XI-4 o XII-4 )L
0 N¨R1 . .
IF\ /--\N 1, \__/ N N lik 4* . \__/

iel XII-5 =
IW N ,r) N ------I
c....,.N.,..õ----,.....----,0 "N----NA'----0 N /\/(:)/'N recy R1 1.1 XII-6 ei I

L....,- N-......------.-----0-N -^- Nr"- 0 - R1 In a more preferred embodiment, prodrugs of aripiprazole are disclosed.
(Formula 1 and 7 from Table XI-XII). In a more preferred embodiment, a compound of Formula 1 wherein R1 is selected from Table 1 is disclosed. In a more preferred embodiment, a compound of Formula 1 wherein R1 is selected from Tables 2-4 is disclosed.
In a more preferred embodiment, prodrugs of dehydroaripiprazole are disclosed.

(Formula 2 and 8 from Table XI-XII). In a more preferred embodiment, a compound of Formula 2 wherein R1 is selected from Table 1 is disclosed. In a more preferred embodiment, a compound of Formula 2 wherein R1 is selected from tables 2-4 is disclosed.
In a more preferred embodiment, prodrugs of ziprasidone are disclosed.
(Formula 3 and 9 from Table XI-XII). In a more preferred embodiment, a compound of Formula 3 wherein R1 is selected from Table 1 is disclosed. In a more preferred embodiment, a compound of Formula 3 wherein R1 is selected from Tables 2-4 is disclosed.
In a more preferred embodiment, prodrugs of bifeprunox are disclosed. (Formula and 11 from Table XI-XII). In a more preferred embodiment, a compound of Formula 4 wherein R1 is selected from Table 1 is disclosed. In a more preferred embodiment, a compound of Formula 4 wherein R1 is selected from tables 2-4 is disclosed.
Representative compounds according to the invention are those selected from the Tables A ¨F below and the geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Table A
No. Structure No. Structure \/\C) 401 47.
CI ci rNC) 40 CI 40 N j CI io N j 2.C r, N (:) 01 48. ci rNz=z 10 I

./0.,,,,N
II

3. a rN 49.
ci rNC) 110 CI 40 Nj CI 0 Nj \
N / R
,N___,N
T_-4. r N \/\C) 401 .
Ci 50 CI rNz 40 CI 40 Nj CI 0 Nj \z\
N N
0 0 0 o 5.Ci r N \/\C) 01 51. .
CI
Ci 0 N a * Nj V\
,--N
o o 6. r a N 0 lei 52. ci rNo 0 a 40 Nj ON CI 0 N j zN y N
)( ) 8 0 0 0 7. C r 53.
i CI rNC) 40 CI 40 N) CI 0 Nj I
ON ,rNN

)A 10 0 0 0 8. a rN 40/ 54. Cl rNC) 40 CI 0 N CI 0 Nj I
ON II
vN,.,N

9. r a N \/\C) 401 55.
CI r,,,c) lel CI I. N CI 0 N
(:)N N,.,N
10. r a N \/\C) 401 56.
a r,,,c) 40 Cl, N CI 0 N

(:)N vN,,,,N
11 8 II

11. 57 r 110 .
CI a rN-c) I,a is N CI 0 N

-(416 0 0 0
12. r a N \/\C) 40 58.

/
13. CI r,,,c) 40 59' CI rNC) lei CI is N

--.....õ, y ---41) 20 0 0 0
14. C rN
.,C) 01 60.
i CI rN IS

(:) N 0,1.r N
15. r 61.
CI CI rN

N Oy N
16. r N \/\C) 40 .
CI 62 CI rN..'C) a = N 0 I. N CI is N ,)
17. a rNC) 110 63. a (No le, CI . N.) IW 0 N CI I. N) e 2+ - I
Ca 0-P-O¨N
18. Ci rNC) 110 64. a ('NC) CI i N.) IW 0 N i N) N

I
I
19. Ci (NC) 110 65. ci ( C) Si Cl i N.) IW 0 N CI i N) IW N

I
\/
20. a rNC) 0 66. a ci /---\
Ci i N) .., --,. N
21. Ci ( NC) 0 67. a rN-..-c) CI , N) Y r N
IW / N

I
22. a r,NC) 401 68. a rNo 0 CI N) CI i N) o Ov 0 N IW / N

?LN I
H

\;
23. a r,NC) 401 69 CI r,,-.-0 0 CI 40 N) CI N
IWN .N
0 C)V

YLNI I
H
24. a rNC) 40 70. a CI I. N) CI N
O N IW / --. N

I
H
25. a rNC) 40 71.
Cl I. N) CI N
O N IW N
HN

TALC) 0 0 NI
H
26. r CI N .\/.\C) . 72. CI ro *
CI 0 N) CI N
IW H
rN N
O N )r , N\7 0 0
27. CI C) r N
, 40 73.
Cl r N .....",,,.../=,,,õ,.0 Iso CI 0 N) CI N1) si H
O N
)i. N y N

HO
28. C r,NC) 40 74.
i a r.,N\./.\(:) 0 CI is N) CI 401 N
H
N li, N
=Oy N
29. a rNC) 40 75. a r CI I. N) CI I. N
Oy N Cy N
30. r 76 N \/\C) 401 .
CI CI rI\JC) 0 CI 40 Nj CI

O N N N
II
31.r N \/\C) IS 77.
CI CI rN 0 CI 0 N CI is N j
32. a rN 0 78. CI rN01 CI 0 N CI 0 N j N H2N rN
33. a rN,C) CI
0 79.
r-N
Cl 0 N1.) CI *
O H
N N
e_N Nj , 0 0
34. CI rN 0 80. CI
a 40 N j a 0 Nj 0 o-N120 NN
35. C r \/\C) 0 81. r CI 40 N) CI 0 N
NH

e_N )(N
36. C r \/\C) 0 82.
I N CI rN 0 CI is N j CI 40 0 N j 1.rN1H N

N
HO ---- ,---
37.
c, (,NC) IS 83. c, rN.-c) ) 0 N c N
HO--\___ ,---- H

\--\
OH
38.CI r,NC) 0 84. CI rNC) 6 CI las N CI 0 N) ( N1 $--N H2NYN '
39. CI r,NC) 0 85. CI rNC) 0 Cl 0 N,) CI i N) , N IW H2N j-NrN
H

CN
40. CI r,NC) 40 86. Cl rNC) a CI las N) CI r N) IW i 7 N rY
41. c, (NC)40 87. 01 CI las N) / NNO N
42. c, rNC) 40 88. 01 CI I. N) WI N

L
43. c, (NC) 40/ 89. 01 CI I. N) WI

Nc) =

0 c(
44. ci rNC) lei 90. a ci lei N1) N
OyN
45. ci rN(:) 0 91. a ci 0 N1) 0 Cl N
=C)1.rN
No 0 I.

(")
46. ci C)S
CI is N1) C)i-N

Table B
No. Structure No. Structure 100.143.
a r N .\/.\C) 0 ci rNic) 0 CI 0 N CI N) 0.1_,N

101.144.
a r N 0 CI rNi-c) 0 Cl 0 N ,) CI r N
1 1W Oy-N
I

102.145.
a r N \/\.C) 0 CI NC) 40 CI 0 Nk) CIr N) 1 IW C)r-N
I

103.146.
a r N \/\C) . ci rNic) 0 CI 40 N CI N) I

104. r \/\C) . 147. CI N 40 CI
N
CI 01 Nk) CI 40 N
1 .
/0õ.__,N I
I I

105. rNõ.......,.....,........õ,.0 0 a 148. ci rN
CI -c) 0 is N CI 0 N
\ I
1 N 7 ,N
.R,,- N
0 o 106.
Cl r N 0 149. lei CI is N CI . N \7\
I

N )( ),) 10 0 0 0 107. Ci r N 0 150. a rN 0 CI 01 N CI 0 Nj 7\ 1 108. a r N 0 a N 151. CI rN-c) 0 C) N CI 0 N

7:N y N

109. CI r CI N 152. CI r N
0 CI s N

-r= i 8 II

110. r 151 Cl r N (:) lei CI
CI 01 N ,) CI s N
C) N 1 I
N N I
-r= i 13 II

111. r N 0 0 CI
CI N 154.
is CI 0 N

I
C) N 1 N N
-r= i 13 II

112. Ci rNC) 0 a N
155. ci (õc) 0 01 ,) I CI si N) 0N 1 srNN
II

113. Ci rNC) 0 156. CI rNo 0 CI 0 N CI 0 N) ===õ%,÷ y ----a)22 0 0 0 114. Ci a N
157. ci (,,--0 0 0 ,.) 0 CI 0 N) /
q N 1 0 N
'y II I

115. Cl rNC) 0 CI N
158. ci r,N\/\C) i&
is ) CI . N) -..,.., II I

116. Ci rNC) 0 159. CIri\l CI N
'0 0 CI 0 N si L) 0 N (:) 1N

, 2 117. arNC) 0 160. ci r,,,,NC) a 40 0 N CI 0 N,) 1 n N I

, 3 118. Ci rNC) 0 161.
CI ri\l'0 10 CI 0 N CI flo 1\1) k 1 Oy: C)1.r I

119. Ci ( NC) 0 162. ci rNC) 40 CI 0 N CI 0 N) e o I

Ca2+ 0-P-0¨N
Oy: II

120. r CI N.\/.\v 0 163. CI rNIC) 0 CI 0 N1) CI
I IW N I
O N ,,y , 7 1 , , , 121. r N.\./.\.0 0 164. CI r NC) 0 CI
CI 0 N CI N) I IW N I

r I -H

122. CI r N .\./\. 0 0 165. a Cl r---\
CI 0 N . N N----\,--,,r *
\____/

o 0N
---...
--, N
V
yLN 0 V
o o H

123. CI ( N.\./.\v 0 166. CI r NC) 0 CI 0 N CI is N1) I N I
o (:)v N /

)LN 0 H I

\/
124. Cl r N.\/.\v 0 167. CI r NC) 0 CI 0 N CIis N1) N I N I
0--,õ---H

125. a r N 40 168. CI rN .
CI 0 N CIi N

O N /

?; 0 I

126. a r N 40 169. ci rN .
CI
Cl 0 N N r IW N
1 , N 1 O /

HOv \/\
127. Ci rN 0 170.
ci rN 0 CI 40 N CI is N

v N HN N
oill NH 0 -L0 0 0 128. a r N lei 171. I. CI 01 N CI io N

I
Ov N
N

129. a r N 0 172. ci r, N
v./\.20 0/
CI is N CI 1 ,N H
.'N I
O N
\ / 9 IIN

130. Ci r N 0 173. ci rNv./\.20 0/

H
N ir-N I
O N

131. r 174.
CIci CI is N CI 0 N

)' Y

, 132. CI1N-- 0 175.
a (N--040 CI 0 N1) CI 0 N>
, O 1d 0 I \--N N7N
I I

133. CI r,NC) 0 176. CI rNC) 0 CI * N) CI i N) O I IW
I
12\--N H2N(N

134. ,OS 177. rN(:) 0 CI CI
CI is N) CI 0 N) O I
Th,õ-N I
e_N H2N

135. r N .\./.\(:) 0 178.
CI
a Ci 0 N) CI 0 NJ
H
O I

HO -- ,--- N 0 N N
)r NH
136. (,NC) 0 179.
CI
CI
Ci 0 N) Cl 0 Nj .
I

N N
,--N
HO-N____ * 0 0 \-\
OH
Cl 137. r.,,N,..-......_,,,,0 0 180. CI
('NC) 0 CI 0 N, )\ N I
) CI s N) I
(-N
/N) 0 0 0 138. r CI N .\./.\(:) 0 181. CI rNC) 0 CI 0 N.) CI * N) 0\ I
) N HOH.r N I

139. r NCI
0 401 182.
ci r,,--c) 0 I I
,W N

140. r N CI
.\/.\(:) 401 183.
ci r N 0 c, 0 N CI io N

\/\) / NNC)ON1 H2N y N

141.. N
N CI
r N 1:) 184 401 r (:) 0 CI
CI 0 N CI N j H2N )-LN N I

NhCO 0 142. CI r N .\/.\C) 40 185.
ci r N a CI N CI N) I
H IW 2N j=L N mr0 N H2N
)-LN N

Table C
No. Structure No. Structure 200.
CI N r 0 I. 222.
CI
CI 40 N a io N1) NI
N

y0 c)( 201. a r N
0 223. a ( N \./\ 101 CI 40 N a io N) N
N

/y )h.( 202. a r N 0 224. a CI 40 N a 0 N
N
N

\/y 203. Ci rNC) . 225. a rNo s CI 0 N) CI i N
IW N
N
/

\/y0 204. Ci rNC) 0 226. a rNc) s CI 0 N) CI i N
IW N
N
H,3h(C) 205. Ci rNC) 0 227. a CI 0 N) CI i N
IW N
N
o 206. Ci rNC) 0 228. a rNc) s Cl 0 N) CI i N
IW N
N

207. Ci rNC) 0 229. a rNc) s CI 0 N) CI i N
IW N
N

208. a(N- 0 230. a (N-- 0 CI 0 N) CI r N
IW N
N
H-Th'r o o 209.231.
a r NC) 40 a rNc) s c, 40, N) CI 0 N1) N
N

210. Ci r NC) 110 232. a rNc) s a 40 N) ON CI * N) N

_--N.( 211.233.
c r NC) 0 a rNo s CI 0 N) CI 0 N1) N
N
\ 0 0 N-i 212.234.
a r NC) 40 a r-N,--0 c, 40, N) CI 0 N1) NI
/
N

N-o 213.235.
a r NC) 40 a (-N,--0 c, 40, N) CI 0 N) N
----Y
N

214.236.
a r NC) 01 a rNc) I.
c, lei N) Cl * N) N
N

215.a r NC) 401 237. CI rNc) I.
c, 40, N) CI = N) IW N
N

216. a r N C) 401 238. CI rNc) I.
c, I. N.) CI = N) IW N
N

217.a rNi\./\.0 is 239. CI rNc) I.
c, 40, N) Cl = N) IW N
N
0 ENJIC) 218. a r NC) 401 240. CI rNo s CI
c, lei N) isi NJ) N
N
Ay0 219. a r NC) 401 241. CI rNc) I.
CI is N.) CI O N) N
N
= H 0 Oy0 N

220.a r NC) 401 242. CI (N-- ) 0 c, is N.) CI 0 N) N
. Oy N N
47 221.243.
CI ,N\/\C) 0 c, C, 0 N 0 1\ CI r N
N r1 \ \
48 Table D
No. Structure No. Structure 300. CI
322. r,NC) 0 CI
CI is Nk) CI 0 N1.) 301. 323.
N
r,NC) 5 CI
CI N CI
40 CI s k) /y )().( 302. 324.
N1) r,NC) 0 Cl CI N CI

\/y0 303. 325.
N
r,NC) 0 CI
CI N CI
0 CI 0 k) . -..

304. CI
326.
r N r,NC) r&
CI
CI 0 N CI 0 N1.) 1 H,301( 305. r N 327.
CI a CI is N CI 0 N1) 0 N I N I
0 ('-rz?1=(
49 306. Ci rNC) 40 328. a rNC) 0 CI is N) CI 0 N) N I

307. Ci (NC) lei 329. a rNC) 0 CI 0 N) CI 0 N.) N I

y0 308. Ci rNC) 40 330. a rNC) 0 CI is N) CI 0 N) I
N

o 309.CI r,NC) 40 331. CI
CI 0 N.) a 0 N.) I
N

¨\ I' /¨

,,,Thro O-P-0 g 310. Ci rNC) 40 332. 5 CI I. N) Cl s N) 0\\N I N I

---a)1 _--N1 311.CI r,N.\./.\. IS 333. a rNC) 0 CI 0 N) CI s Nk) I N I
N
\ 0 0 N---i 312. Ci rNC) 0 334. a 0 CI 0 N) CI 0 N) I
N

c/ 0 0 ( 0 313. a rNC) 5335. a( CI 0 N) CI 0 N
I N I
N
---\ 0 314. a rNC) 40 336.
a r N *
CI 0 N) CI s N
I N I
N

315. a rNC) 40 337.
a r N 0 Cl 0 N) CI 0 N
I
N

316. a rNC) 110 338.
a r N 0 CI 0 N.) Cl 0 NJ) I N I
N
\IL_ hl 0 317. CI rN.-(:) 0339.
a ( N O
CI is N) CI s N
I N I
N

318.
CI rNC) 40 340. a r N *
CI 0 N) CI s N
I N I
N

319. r \/\C) 40 341. a rNC) *
CI N
CI 0 N CI r N

.1H 0 Oy0 NI

320. CI r,N(N- 40 342. a rNC) *
CI 01 N) CI r Nk) I\1 1 IW
* N I
NC) Oy0 321. r \/\C) 40 343. a rNC) *
CI N
CI 0 N CI r N

0y0 \\

322. /\/(:) 0 344. a rNo *
(---N
a Nj CI I
N---) NI

. 0 Table E
No. Structure No. Structure 400. 0 0 415. 0 0).LNA 0AA
oiy __la N
Nr-\N . 5 /---\
N N .
41 = \___/
401. 0 0 416. 0 0 0NA ).L
Oiy 4I se ()ANA
1.1/---\N 41 . 9 \__/ N N
41 = \___/
402.
v 0 417. 0 0 41 le OA
N\
Ot.,y 1\11-\1\1 = \ /13 /--\

403.
v 0 418. 0 0 C1N---k 0A N).)?
0/_\ = la 1\1/-\N se 1 5 N/ --\N 4. ( 6 40 II \__/
404.
v 0 419. 0 0 ,___\ õ 9 N N IP Nr-\N ii ( 8 41 .
405.
v 0 420. 0 0 IDNNI----kNH = = 0AN
/---\
N N 111 /--\ __Al 0 1 =
\/ N N
406.
v 0 421. 0 0 0N---1(N,\ . . 0AN
NN4. __ /
0 NN . )..1 2 41 II \____/

407. o o 422.
o ,\ _ )L )L
N--I) 9 o N
/--\/-\
N N 11 N\ __ t =
408. = 1 0 423. o _ 41 0 N-j\
/ __ \ CAN
NnN
\ __ / =
\___/ = =
409. 0 0 424.
o 0 4* * oAN)\---"( N N
r\N = \ __ / lit \___/ . =
410. o 425.
o ON--1-iii () N
Nr-\-IN .
= 11 . .
i \
411. 0 426. o )(o N-%......õ) /---\ *
N N /----\
N\ iN *
= =
/ ( . =
\--412. 0 427. 0 0 0)LN
ONAii) /-\
/----\ . _ N N -0 N\ iN .
\_._i 41 =
= = ----_ s /

413. 0 428.
o 0 ON 1( k_, N
rTh N

Ni-N . N \ iN 1, Q
41 II = 11 /
I
414. 0 429.
o 0)N 0 (:) /---\
N N
IL/ N N
41 . 414 II
I
I
Table F
No. Structure No. Structure 500.
, 519.
S IW , N N 0"*.N N0 N N
501.
, 520.
i 0, NI N
O N IWW N ONNO
N N
502. . 521.
S
, õ.
, 1 (:)NiN0 1.1 N N ON NO

503. . 522.
jo O N
I
ONNC) W NI ONNO
N
----_7--- --0 5 0 504. . 523.
ill N
N

ONNO

505.
524.
Oj N o .õ. =-=* õ,,I
.,.-..c., N ONNO
506.
525.
ill ONN

ONNO
507.
o 526.
OaN 0 / 1 N ONNO
508.
527.
Oi .,-..... ).--õ ,,=,k, N ONNO

509.
rel 528.
w o N O j NNO IW N
..,..-.* õ,,I
,..=,c., ON NO
510.
rel 529.
o tw N O j NNO IW N
..,..-.* õ,,I
,..=,c., ON NO
511.
S
530.

IW N

I
C) 0 N
512.
r&IO 1 531.
ill O
IW
I
,..... NNO
N ,-..,. õ.....k, IW N ONNO NI v,Lo .d A
513.
o 1 532.
Ojo N ONNO IW N C) NNO
Lo N 0 Fr A
514.
lel 533.

ONNO IW
I
ON NO
N
N oLo N 0 A

515.
r&I. 534.

Ili o w .Ni N0 ...
IW N N ONNO
N ic)Lo N
c jix0--516.
j 535. "
NI

ONNO IW
I
ONNO
N
N
- /
/
517.
r&IO , 536.

i IW N
ON N0 N eN N

-,.
/
\ / V
-._ 518.
f&IO 537.
O NiN 0 II.
IW
W
I
N N ONNO
N N

V
I
L.

\:\
In another embodiment, the invention relates to a compound of Formula LI and LII:

F F
. 41 li\T _____________________________ 0 N/ 0 N _____________________________________________ / ________________ N / , d iR1 0 > ____ N ________________________ ) __ N \R1 0 ) N ) N
N \ \ N \ \
Formula LI Formula LII
In another aspect of the invention, compounds of Formula LI and LII are selected from Table G:
Table G
No. Structure No. Structure 700. F 741F
. 4ilt ii-0 / 0 \\
O>- d ):)/
01 NI)-/ ) N -N
) 0 \ N \ \ 0 \
0 \
701. F 742. F
. 40 NI/ -) 0 )_N
\\
/-N
0NN-N/\ )_N\ )i 0 > N/ \ / N\ 0.--1---<
N1- \

702. F 743. F
. .
NO NO
)_N
\\
\

\ 2-N\ //\ 101 N
0 N \ 0 703. F 744. F
. .
NO
0 ) )_N
\\
\ - N
1\1 NI/
1\1/ \ / \ )/ /--) 10 -N1µ )- N \ y(:)..1.<
N \

704. F 745. ' . .

)_N 0 N /
O>¨ NI/ / / \
0 Nx ) N \
),..,'n'''f..41';
\ / \ N \ 0 705. F 746. F
.e \ 0 O>-/N )_ i N
N
\ (48 lo N\ /
\ 1 N\ ) N\ I/
0 N 0 \
706. F 747. F
4Ik =
/
NI/ ¨ 0 N ¨C) \\
)_N
0 I \I¨ NI/ )¨ 40 N\ N/ ) N
\
N \ N\ (410 Ni \ \ , "
o 0 ) 707. F 748. F
= . /
d 0 N ¨C) \\

N1\i _/ ) /-N I. N \ N/ ) N \
N
\ (412 Ni¨ \ \ , "
N \ 0 708. F 749. F
=.
I" 0 N( ) 0 \\
)O N-1\i / )_ N
\ )/ (413 40 N\ N/ \_N
N \ Ni- \ / N\ 1 N
0 0 ))) 5 709. F 750. F
411.4i NI/, 0 d ) 0 0 I\IN/
N\ )i (414 0 N \ N/ \
/
Ni \ o )) 8 N \

710. F 751. F
411.( 0 fa NI' (:) 1\1/-0 Ni\i/ )-N) N / ) N )_N
N"
\ >/416 10 ) N
1\ \ \
N \ 0 o )))13 711. F 752. F
.*
N\')01 \ I/ - 0 0 Ni\i/ )_ \ j<
N
\ )i ( / 418 0 N \ N" \
N \ N \ i \,_N i "
o 0 y)13 712. F 753. F
.40 NI/ 0 d ) 0 \\
0 N-ii )_ 40 N\ \
N \ N\ )/ (420 Ni- N/ \ / \ 1 N
0 0 ))8 713. F 754. F
411'411 N/-0 N/ ) 0 0 N-ii )_ )-\ /----N\ )i (4'22 10 N
N N" ) N -1::( N \ N \ \

714. F 755. F
11-0 N(¨) 0 \\
)=N
\_ N /N
0 N >-N/ 10 NN/ ) N\ >--0/--) N/ - \
i\l/ \ /-\ 0)/ (0 715. F 756. o . /¨\ (------( N ('0) 5 N---=----._( \\ N--- 0 /-N
N F =
NI/ 0( NO
*N-----(1 o 716. F 757.
(------f (,)9 = ,_, NN--,......õ
N po N---_ 0 \\
\ i¨N
0N/ N)¨N/
N
) 2 N--f NO
\ /¨ \ 0i 41It 717. F 58. F
4ilt 4Ik , N 0 N /
\ /¨N

N \ \ 0 \
i\i/ \ /¨ NI\ c:?/' ' 3 o 718. F 759.
(--- (-)15 e N.' /-\ N------ _--("---õ,A

\ /¨N
0N)¨N/
N)/' N----µ
F .
NOi\l/ \ /¨ \ 0 ) 41li 719. F 760. F
1111, 411.
N /- N/-= NN/
i_N H2 \_ /¨N
oi \
N/ \ /¨N\ 1:?/' ' 5 720. F 61. F
4Ik =
i\l/ (:) \\ 0 N-i\i/ \-N>-N
\ /¨N
0 N)_N, N' \ /-N\ ) ) N \ / \
' 7 --,.
\ .......
/
/

721. F 762. F
. lik il\ o o N \ 0 \
N /¨N N/ )¨N HN \ /
./( )_[\?¨:

N \ NF

¨ /
722. F 763. F
.
N/ (:) o N/ (:) \\
\ ¨N /¨N
I. Nµ /) NI/ HN
¨ \ i\ 0 I \1_ N NF
0 N \----\o \/
723. F 764. F
.
=
Ni o o N/ 0 \\
\
.\
0 N, N" HN
//¨ \ i¨N\ / 40 I\1¨N/ \_Nrki N NF
0 \ / \
N

/
____ \ /
_____ 724. F 765. F
.=
N/ o o N/\ 'O
\\
¨N
0 N / ¨I\ j )_N)-N , 0 N, / / HN
N1\ )-N\ /
NF
N N \ \

/
/
\ _ 725. F ________________________________ 766. F
4IkS
N/-0 1\1/-0 _ \_ /-N 0 I\J/
40 N)-N/
N/ ) \ __ /-1\1 \ / C. N \

0) r \_ 726. F 767. F
4Ik .

\\
0 N, / /-N 0 I \I- NI/ )-N/-N
NH
/
/1-N )-N\ )/ ________________ /OH
N \ N \ \ (:

727. F 768. F
= 441k d (:) N/-0 ____________________ A A
/¨N __________________________ I NI\ n./
\ 0 NI, /
N//¨IN\ _________ )¨N\ ___ NH ___ = /2¨N, )¨N\ o 0 N ` 0 728. F ____________________________________________________________________ 769. F
. =
1\1/-0 NO
\\
/-N \_ /-I. 1\1)_N/
N -0/ __ ( 4 0 N)_N/
N N\O
N/ \ __ )- \ 0 Ni \ / \
(N) 729. F 770. F
. 4Ik NO NO)-N ____________ /- N / \-0 N)-N/ )-NV % 4 0 N¨ N\ / N\ 0 NI/ \ __ \

730. F 771. F
. eqiik d 0 i\l'-0 \\
\ /-N 0 I \I_ NI/
0 N)-N/
N\\ 0 N, \ /- \ . N \ /- \
0 . NH2 731. F 772. F
=.
N''O N/, 0 \ N / )_ N NH2 0 i\i-N/ )-N)- k 0 N N N )i N \ \ N \ \ 0 732. F 773. F
= 40 i\l/-0 N/ 0 \\
\ /¨N -N /
0 N) I"
Ni \ /-N\ --- N \
0 \10 \ /
, 733. F 774. F
e 41k i\l/-0 kl/-0 \\
\_ /-N
0 N)_N/ 40 N-i\i \-N
/ >-Nii NH2 N/ \ /-N\ ----<) N \

OH

734. F 775. F
=4k N/-0 N" )=O
- N )-1\1___0 \\
=0 14N)-N/\ )-11:0N)/, c 0 N) d \
Ni \ / \

735. F 776. F
= 411It I" 0 d -(:) )¨N N\ N/ \

0 N )¨N NrEi---OF 0 N /¨ \ / \
\ \

736. F 777. F
.*
N\' \_ ¨N r---/ N
0 N)¨N/
N\ _N

m OF 01 -1\i/ µ ) \ r r¨NH2 NI, \ / '\---1 N \

H o 737. F 778. F
4lik 41k Nii-0 Nii-0 \\ 0 0 N¨Ni/ \_Ni¨N /¨N
\ /--\ I. N\ I"
N
r \ ) \ )/ \ /NH
N \ /¨ \ cr N\__7-0 \
738. F 779. F
* =
N/\ 0 1\1/-0 )-N N
Nr?-d\ )-NN/ )-1\1/ 0 1\1)-N/
N
)- \ (:3 \
\
is HN

739. F 780. F
= 4Ik N/-\0 N/-\0 \ \
\ s 0 N)-N/ /-N\ -0 IN/ \ /-N\ // \ J I-N\ ) )/' -N\ NH/
0 0 0 19'9 740. F 781. F
4ikt 4Ik d 0d 0 / \ \\
,-N
= N)-N, 0 ( s N / \
N
I\1/ \ =)¨ \ \ J ' 0 0 1-N\ /-N\ 0)/
Compounds of Formula IX, X, XI, XII and in particular compounds of Tables A-E
are useful for the treatment of neurological and psychiatric disorders including schizophrenia, mania, anxiety and bipolar disease. These compounds provide sustained release of parent pharmacophores by cleavage of the labile moiety, R1.
In another embodiment, compounds of the present invention are represented by Formula XIII or XIV as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Ri 1 0 0----Ri N?R
loo Rioo R103¨ R103¨

..... R101 .... R101 R102 ilK100 f(102---- )(100 Formula XIII Formula XIV
wherein Rloo, R1015 R1025 and R103 are independently selected from absent, hydrogen, halogen, -0R10, -SRio, -NRioRii-, optionally substituted aliphatic, optionally substituted aryl or aryl or optionally substituted heterocyclyl;
alternatively, two R100, and R101 together form an optionally substituted ring; and X100 is ¨CH- or ¨N-.
A preferred embodiment is a compound selected from Table XIII-XIV. A more preferred embodiment is a compound from Table XIII-XIV wherein R1 is selected from tables 1-4.

Table XIII-XIV
R\1 o 11 Ri 1 \

Br ---N
I.
Br 'N
N
\ / N
\ /

R\1 o 12 Ri \

NI <
Nzz-.-__ <

Cl 11 ----N OH
Cl li -----N OH
*
*
R\1 0 13 Ri 3 \

I
02, NT I.
---N
* Cl * Cl R\1 o 14 Ri \
. NI 0 Ni Cl N1 NI

N
() Cl -----40 Cl 0 Cl Ri 15 0----Ri \ 0 0 N NI 0 I (0_\
el<-`
Cl NH 0 Cl NH 0 R1 16 Ri 6 \ 0 \
-----__ N
OH
OH
Cl NH
Cl e N
I

NH

R\1 o 17 R1 7 \
. i N N
Cl N
Cl I. N

40 Cl 0 . Cl Ri 18 R1 8 \ 0 \
. NI 0 _N<

02iNxT ---N
Cl *
R\1 o 19 0----Ri NI
Cl Cl li. 40, 0 1 I 0 NI Ri . N
el Br N_....
Br N.....

PRODRUGS OF ACYLANILINES
In another embodiment, compounds of the present invention are represented by Formula XV or XVI as illustrated below, or its geometric isomers, enantiomers, 5 diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
R50 Ri R50 I
R51 01 N... R55 R51 NR55 Formula XV Formula XVI
wherein R1 is as defined above;
each R505 R515 R525 R535 R54 and R55 is independently selected from hydrogen, halogen, -0R10, 10 -SRI , -NRioRi1-, optionally substituted aliphatic, optionally substituted aryl or aryl or optionally substituted heterocyclyl;
alternatively, two or more R505 R515 R525 R535 R54 and R55 together form an optionally substituted ring.
A preferred embodiment is a compound selected from Table XV-XVI. A more preferred embodiment is a compound from Table XV-XVI wherein R1 is selected from Tables 1-4.

Table XV-XVI
1 o 40 0 OH OH
H

N Nip I

r 41 0 r Ri, N

H
N N

)N N
I

0 101 1 0 0 I.
0 0 Ce N N
I

RI 0 44 o N N\
NH NH

le 0 . () OH

0 OH 45 .,..Ri 0 OH

I
Ri 7 Ri 46 N
I
N

0 Ri 8 -,...,Ø 47 0 Ri1 1 I
N\s/
0 oCo Ri 9 R, 48 I I
* N I
* I \ \ \
0 0, 0Ri 49 R1 ) /

0 = NH /AL F
W
F N F 0 . )=N
H /..\ F
W F
Cl F
N I Cl Nr() NH
/
/NH

RI
*N OH * N
/ N \ / N OH
0 ---- 0 10 *
1211 =
. F F

N, 1 N

Ri I

I.
0 101 (i) F3C 1:1X)1 F30% j<v le 01 IW 0 IW NC Ri F
NC F

15 Ri 54 I
01 N.
1\1(:, Br Ri Br I

0, OH
0 Ri 17 R1 ,56 I
0 N_I\T NN
0 40 () Ri 18 ri'l . 0 NRI 1 el 57 . 0 N 10 I\T

--N '---N
N, )-_/----\N 1-__/----\
N )---_/- \__/N---\
IN)--Nr ---/ --\--OH 0 N,TX >--NH `-OH
S
10 111 -S 0, CI CI RI

N N
Ri 0 * NI

Ri I

0 ) 0, Ri 22 Ri 61 I
F3 C 0 N. F 3 C 0 02N 02N Ri NI\I R1 . N
N
I
1.1 () C10 N C) Cl 0 l\c 0 C) H I H
01 N ,N
II
0 9 * N
* 0 OH Ri I

Ri 1 Ck F3._,0 F3C

(31 Ri I
0 1\1/\/0 N
0 oCo Ri 27 Ri 66 0¨R1 I
0 NNJ N¨K /-0 ) = N
28 0 RI Cl 067 0 Cl 0 H H
N I.
HO N)Y 01 ycH HO OH
0 0 C) 0 CN CN

I

0 lio I oCo I
Ri 0)f . N 0 N,......õ,.Ø 0 N 0 1\ir 0, I
S 0 I S Ri I
01 N 0 1\T/
0 () Ri 32 Cl 71 Cl Cl R1 . Cl 0 1\1 el 0 OH 0, OH
02N 02N Ri 33 Ri 72 /\o I
0 N 0 1\T/
0 () Ri I
Ri 0 N,N 0 N,N1 0 0, Ri Ii 74 0 1/

NO () Ri H

H
OH

36 Ri 75 I
0 N.,,,/,====-....õ0 0 NNO
0 Ri ao N.,......õ.....^.....v^,) OH o * NI\T OH 0 0 NjO 10 RI
*

Ri I
0 N,.........õ======...,õ,,N.,,õ 0 N.c.,õ:õ.========--..õ,,,,...N.,,, 0 oCo Ri N I\T el HO HO
N 10 0 N 0 () RI

CI
CI
Thiazolidinones In another embodiment, compounds of the present invention are represented by Formula XVII, XVIII or XIX as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

Ri FiN... Fi Fi S---___< s=-........<

/
0 0 Ri Formula XVII Formula XVIII Formula XIX
wherein F1 and R1 are as defined above.

A preferred embodiment is a compound of Formula XX, XXI or XXII as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Cy2, Cy2 O'R1 S
Ri Formula XX Formula XXI
Cy SyN
Formula XXII
wherein R1 is as defined above;
Cy2 is an optionally substituted heterocyclic ring; and X5 is selected from absent, ¨S-, -0-, -S(0)-, -S(0)2-, ¨N(Rio)-, -C(ORio)(Rii)-, -[C(R10)(R11)]v-,-0[C(R1o)(R11)]v-,-0[C(R1o)(R11)]v0-,-S[C(R1o)(R1i)L0-, -NR12[C(R10)(R1i)L70-, -NR12[C(R10)(R1i)k S-, -S[C(Rio)(RiAv-, -C(0)[C(R10)(R1Av-, and -C(Rio)(Rii)=C(Rio)(Rii)-; wherein v is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10.
A preferred embodiment is a compound of Formula XXIV as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

S
Ri Formula XXIV.

In a more preferred embodiment of Formula XXIV, R1 is selected from Tables 1-4.
A preferred embodiment is a compound of Formula XXV as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

µ

N ______________ Sr ____________ \

Formula XXV
In a more preferred embodiment of Formula XXV, R1 is selected from Tables 1-4.
A preferred embodiment is a compound of Formula XXVI as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

N _______________ )_ S,N
\
N/
Formula XXVI.
In a more preferred embodiment of Formula XXVI, R1 is selected from Tables 1-4.
A preferred embodiment is a compound of Formula XXVII as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

/ ____________________ 0 41 / ___________ SrN.,...

Formula XXVII.
In a more preferred embodiment of Formula XXVII, R1 is selected from Tables 1-4.
A preferred embodiment is a compound of Formula XXVIII as illustrated below, or its 5 geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
Ri N µ
) / ____________________ 0 it / ______________________________________ \
Sr Formula )(XVIII.
In a more preferred embodiment of Formula )(XVIII, R1 is selected from tables 1-4.
A preferred embodiment is a compound of Formula XXIX as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
0 .0 /(/_)/ s,N
0, Ri Formula XXIX.
In a more preferred embodiment of Formula XXIX, R1 is selected from tables 1-4.

A preferred embodiment is a compound of Formula XXX as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

0 *

Formula XXX.
In a more preferred embodiment of Formula XXX, R1 is selected from Table 1.
A preferred embodiment is a compound of Formula XXXI as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
O'R1 \N
HO ''0 Formula XXXI.
In a more preferred embodiment of Formula XXXI, R1 is selected from Table 1.
A preferred embodiment is a compound of Formula XXXII as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

0 *

Formula XXXII.

In a more preferred embodiment of Formula XXXII, R1 is selected from Table 1.
In a preferred embodiment a compound of Formula XX-XXII is selected from table XX-XXII below, wherein R1 is as described above. A more preferred embodiment is a compound of table XX-XXII wherein R1 is selected from Tables 1-4.
Table XX-XXII

o . io 41 ib )-1\r yNj RI
< )¨Nr-j Q \
S
IT

S
\

\ rN S N..., Y ' Ri---Q \
"N N
iT S N
), 0 0, 7 o 8 o¨Ri o *
HO . HO 40 ¨RI o * \
o s/N
,=0 s,N
\\

F

0 #0 = 0 S-.....fN 00 S =
HO C)RI 0 '0 F OM \N F
0 I 10 S,C
0 0 S-----µ

b 13 = 14 0 yo s--<
0 S¨_fN
\
N
¨RIo o 411, \ , /
ill, 0 1\1\
o Ri \
Ek Nyci 10 N
0 -N¨R 1 S.--µ.
o 0 its N\

o \

S----N
10 \N

0--Ri \\O

,I\T 0 c) 101 R1 0 I. ON

S
21 o o 0 ol N N
/ S
Ri Thiazolidinedione prodrugs of Formula XVII to XXXII are useful for the treatment of type 2 diabetes mellitus. Herein provided is a method of treating type 2 diabetes mellitus by the administration of a prodrug of Formula XVII to XXXII, in particular a compound of table XX-XXII above wherein the prodrug provides sustained release of the parent drug. The parent drug results from the cleavage of the labile R1 moiety.
In some embodiments, a compound of Formula XXVII is selected from table H:
Table H
No. Structure No. Structure 1000. 0 NO* 1021. o 2% s___( __\-----\

0 / \%

O (ij CI\
1001. 0 NO* 1022. o s____ ___\-----\
0 s-4 1002. 0 1023. 0 2\TO *_cf----\ 1\10 1003. io 0 1024. 0 ..õ....N,..,õ,.....õ,-..,.õ,-0 s__<_\\)-----\____ 1004. o 1025. 0 1 S¨Al t N.,...(0--., W

1005. o 1026. 0 = * s_____ I 1\10 )9<

1006. o 1027. 0 I\TO */N(1) 40 s__<
1=

WJ

1007. 0 1028. 2 IV..(1) * /0 /I\ /C1 I = N

)9.

1008. 0 0 1029. 2 2V(i) *
/W
I = N
I

1009. 0 1030. 2 = I /1\10 I

1010. 0 0 1031. 2 IV..(1) * /
I = N

N.,õ.70 )9<

1011. 0 0 1032. 2 2V(i) *
I = N

2<

1012. 0 1033. o 0 s_.4 /1\I%/ \ /0 I N __ /
I
N.,.../) 1013. ,0 1034. 0 IVO
I * S-1KNICiii3 NO

1014. 0 0 1035. 0 (NC, *
= N¨ I
I

0 N :
1015. ,0 1036. 0 NO
I * S¨ --1'(N__ 2\10 401 s_.4 I

I

1016. o 1037. o 2\T O * S- <

N
0 < --) \--N\
1017. 2 1038.

N(i) r . s___<
, 1 / \
N
0 ___________________________________ ( \ \
/
1018. o 1039.

1\i'D 0 s_4 NO 0 S--ic N
o ----, \ /
1019. o 1040.

1\1() , 0 N-....õ.

---- I ----/
\
1020. o 1041.
p __ , N __ 7 BARBITURATES
In another embodiment, compounds of the present invention are represented by Formula XXXIII-XXXVII as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

Rum Ri Ri NX
Xf Xf Xi ,,,,,...,,,.............2 _______,,,...,,,.........õ.õ....X2 ,=====,,,,,,,,..õ...õ,,X2 R Rum Rum Xi Xi Xi Formula XXXIII Formula XXXIV Formula XXXV
Rum Ran 1 Xf N X
Xi- - IZi 1 T I Nx2 ,=====,,,.õ.õ.õ,x2 A10, x10 Formula XXXVI Formula )(XXVII
wherein, X, X1, X25 R1005 R1015 and R1 are as defined above;
Xio is -S or -O.
In a preferred embodiment a compound from Table XXXIII-XXXVII is provided. A
more preferred embodiment is a compound of table XXXIII-XXXVII wherein R1 is selected from tables 1-4.
TABLE XXXIII-XXXIV
1 R1 <o 9 0 HN
0 NH 0 < N-Ri \N /0 10 \ <
o N-R1 \
( 3 11 \ 00 N <
N <
0 N-Ri 0 N-Ri ( = 0 ------ -, 4 / N <0 12 R1 0 \
HN <
0 N-Ri 0 NH
( ( Ri 13 HN-\
\
N-\ 0 N-R1 4110 0 =0 6 o 14 RI\ S
N
Ri =-=..1 <

1 >
<

\

7 /s 15 /S
HN RN
0 N-Ri 0 N-Ri < ( \
8 R1 S- M 16 R1 /s \N\ \N < 0 NH

< ( Pyridone pyrimidone and pyrimidione prodruo In another embodiment, compounds of the present invention are represented by Formula )(XXVIII or XXXIX as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:
yRi X

Ri N
N I

(R (R3)q 3)q II
I
K ,X12 A ,X12 (R4)M X11 (R4)1111 X11 Formula )(XXVIII Formula XXXIX
wherein X, R1, R3, R4, m and q are as defined above;
X11 is ¨N- or X12 is ¨C(0)-, ¨C(S)-, -C(R10)(R11)- or ¨C(Rio)(0R11)-; and X13 is ¨0, -5, -N(R10)(R11), -0R10.
A preferred embodiment is a compound selected from table XXXVIII-XXXIX. A
more preferred embodiment is a compound from table )(XXVIII- XXXIX wherein R1 is selected from Tables 1-4.

Table XXXVIII
1 o 9 o H2N R1 NC Iti V N

N
N
2 o 10 o 1Zi F Iti N N

N/o HO

111 i H
OH
3 o 11 o F3c N

NO

NO
HO HO

H'------1 H.------- 0--___......., 1_ /
H H H
OH OH
4 o 12 o N
N
c) N ,OH
N

OH

'Iv-4N R1 0 111t H10 OH
HO /

I

Ri N N N
NO

H
HO O

F
OH
OH
7 0 15 o N

H
H

NH
N
I
NO
F/N/o HO
OH
PRODRUGS OF BENZAMIDE PHARMACOPHORES
In another embodiment, compounds of the present invention are represented by Formula XL or XLI as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

R51 0 N......R55 R51 ",,N55 I
Ri Formula XL Formula XLI
wherein R1, R50, R515 R525 R535 R54 and R55 are as defined above.
Table XL-XLI
1 0 r 22 oRi (--, //1\I 0 N
N I N
\ R:C)N N// * 0 NC.:\1) N 0 \
H
H
I N
I
2 00 0 r- 23 IZi KS %,// r---1 0 irciN r 3 0 I
3 1 24 .

\

I Ri I
4 0 25 IZi 0 r 0 H

R1 , cF3 0 CF3 F ) 0 CF3 r 3k_f_., HOO 26 R 1,, HO, -- 0 N

HI\ic-___-N,c! FIN_c-NL:

N /
N N
N

0 1TL.Y 0 1.1-r() -õ
N r__--.:--: I....c._ N_.--zr-N____c_ N
---"N N
---N

7 HOO 28 Ri HOO

0 N(3 0 1\1 HN
I

HN
H H H H
...,,.>NNNH2 N

0 \
* N N
I
Ri OH
r 0 FIN
HN *

r 0 H H H H

R1 ' 0 Cl 0 NI
Cl 0 \N

..õ......Nõs .
H H
e II _____________________________ s 40 () II

= = 31 R1.õ

S 1\T /s 0 \el H2N/ 0 i\
-j- H2N
Ri Cl Cl H OH 0 32 .
N
OH R1Nõ

0 OH r(1 NH
R H
N

OH

oR 1 Cl * NI\II Cl 0 \1\11\11 I

I

13 Cl 34 Cl NI

OH 0 el I

1\1 Cl / Cl Ri 0 _s X >

15 r 36 R,.._ r . 0 ,-----..,,,,,N..........,...-N * 1\1N
I

* *
* RI 1 0 OH Ri 0 OH
17 \0 0 ) 38 R1 ) * i)I I\ICN) Br I Br I

) 39 R1...,..

0 V) ) H2N 1(2: NCN) H21\1- 01 NC5 I
Ri 0 1.0 Ri..., 1.1.

I
Ri HO 0 20 o 41 o 1111' R,, N * N
I
01 Ri * OH el * OH
Cl Cl --R

/N N
/ N N
zN * zN
N N

PRODRUGS OF IMIDE PHARMACOPHORES
In another embodiment, compounds of the present invention are represented by Formula XLII, XLIII or XLIV as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

X X X
R100µ Rioo\ Rioo \) ;µ N
Rim 1! Rim _IT! Ri0 M
X-,.....,.... Js.1;:.. D ...,..,Lx 1 X 1:...:. ,...".

Formula XLII Formula XLIII Formula XLIV
wherein R1 R100, Run, X, X1 and X2 are as defined above; alternatively R100 and Run together with the atoms to which they are attached form an optionally substituted 3, 4, 5, 6, or 7 membered ring.
A preferred embodiment is a compound selected from table XLII-XLIV. A more preferred embodiment is a compound from table XLII- XLIV wherein R1 is selected from Tables 1-4.
Table XLII-XLIV
XLII- XLIII- XLIV-N-z_1\ 1 0 1 0 Ri I. N¨ )-01 0 N
0 1(1) N -N
0 0 Ri NH2 0 0 \

2 0 2 Ri 2 0 N N N

o'it1 1111 1110. 1110 3 0 3 R1 \o 3 0 i\i'R1 /N
/L N
I R

4 0 4 R., 4 o ' -0 I\TR1 N
N I
R
o 1 * N
* N

o/R1 5 0 N' N

. N 1 01 I. Ri Od( \O
0"--( >.,...( 0 0----Ri 0 0 0----Ri Ole N-R1 ille \N Ole /N

NH2 NH2 Ri 9 09 0] 9 ----N-NN---(N-N 1 ri r) r ' yN-F i yN
n,--,,2,,v , 0, 02N--'0 0 02N7---0 0 10 R,.._ 10 0 ' 0 N N
N
I
0 el ---/
II Nj c 1111 NJ
c 110 N
c 0 11 11 o r) N-N7''''r 1---) N_N7-3---0-Ri 1--) N¨Nr'r r, /,T,----0 n x[V----0 )=-----N
V---%0 -2, , ,,-, 21 N )-1\1 02N )----N

0 \
RI
12 0 12 Rp., 12 o 1\i'R1 i-l-N
N 0.y.-..õN 0 õ...-",,,.....2\Tõ,.....õk ,.Ri NNLC) HNJ
HI\T
HN.,,,....õ, 0 13 0 13 Ri--,o 13 0 1\i'R1 V.N
NNC,1 oNNIC, N7,...,,,,,NN./.......,1 orRI
HI\T HN.,............--14 R, 14 R, 14 R1 N 0 \ 0 \
N 0 b i N
----*
..----/
-----/ * *
101 \ 1 . \ N =N N
N N \ A___ \ \ 0 /
\Ok__ N
/ /
\
N N
\ \

0 N¨ )-0/R1 0 N 0 01 N _I\ 0 / N\ / N
0 0 R1 0 0 0 0\
RI

16 p 16 0 16 . N-RI . HN---(N ----( /
* 0 = 0 = - - - R 1 . 0 In another embodiment, compounds of the present invention having the Formula VI or VII is selected from Table VI-VII.
Table VI-VII

101 %o o\r o o 2 Ri 27 Ri i& N 0 N 0 OH OH
?0 ?0 3 Ri 28 Ri i& N 0 N 0 OH
/
OH H
?0 ?0 4 Ri 29 Ri 4=, I. N 0 4, 0 I\1 0 ,N ,N
11\ )/ \/\(:) //
N¨N 11 N%. \/\(:) ¨N
5 Ri 30 Ri P=, P, ,NO
,N, N¨N NI\ /2 N¨N

6 Ri 31 N N 0, 1 40 Ri is CI

\/----- 32 0 \/----NH NH
O.
. r I I

8 Ri 33 0 N
\ o R1 o .. o 1. fk o Q

1/4. o o ----i o ----µ o o 9 OH Ri 34 OH

40 N 0 I. I\1 O.
/
H H
N N
HO HO
Ri 35 s N\ ,R1 .N

N
N
11 ( 36 ( /
0 N---.7 0 N--/
/----/ /----.../
N N
, H
/\ H
/ \

H F

\
N N

Ri N

4110, / o,R1 11 N
Ri 14 / \ 39 / \
NC NC

/

15 / \ 40 / \
NC NC

/

...," õ,...Ri N
N S S

= 0\ = 0\

/ / ilZi Ri 0 i\i) 71 H
/ __ N 1\1No S N )HO I\I 0, 1 I
N/\N R1 0 \

I& 0) 0 R1 FIN, -Tv \o---- 1 ,1,1 Cl N N" 0 . CF3 \
Ri lel CF3 F
19 Ri 44 N 1\1 d 0 Ri Cl CF3 H3c V

I
/1\INF12 Cl"' N0 N () I I

Cl H
21 NO2 46 0µ
H
Cl 0 N(i) / \
0 = rs 1 \ /N \N
Cl N 0 F II

i\T I. Ri Cl =
1\1 F 0 IT
N
401 \ . oz N a H

N
_\
41'N_) ( /N
/

o'Th\T\

24 N _\ 49 e N 1 RHI) I
i ) ( , 25 o RI ( 50 0 Z
NNH--N N

0 I. CFI

Pei CFI
F

N
\ 1-1N----N
0jr0 N CFI

. R1 0 RI

F

........),........0O2H

I
N
--- N
0 N \ o N

....õ.. 0 0 Ri 0. cF3 C

PRODRUGS OF SULFONAMIDE PHARMACOPHORES
In another embodiment, compounds of the present invention are represented by Formula III as illustrated below, or its geometric isomers, enantiomers, diastereomers, racemates, pharmaceutically acceptable salts and solvates thereof:

A¨S¨N
11 \

Formula III
A preferred embodiment is a compound selected from Table III. A more preferred embodiment is a compound from Table III wherein R1 is selected from Tables 1-4.

Table III

HN
N----0=S=0 H2 NH
S

S
/ i / 0 Rr..--NH
Cl HN
\
RI
2 0)N 38 Cl i >
ol . 70 NH
/
) 7 N HN¨Ri Ri¨NH
N
0 .

C) 401 N
1\111 0 / Co 0 0 i RI

RK

4Ci SCF3 OH RI N
0, .,---N'....,, 0 I
, I NII
1\T

RI

a Oq 1\1N 0 C) NH H

o A

1\114 /
RI

(i)y 0, R1.., ....... s 0-.-- 11 N=N . NH
N /

HO
,-, v NH2 0 7 oCo 43 oCo II NH2 0= 11 NH2 / /
R1¨N R1¨N
)---0 N 0 _ ¨0 c) 0-=1 11 NH2 o=s la N=N = NH2 R1¨N R1-NH
)--)---0 _ )-----N \

0 0%/I
( S
H /
N 10 1 \ ) c / e I R1¨NH

/
Ri ci H

0----) = co . /NH
R1¨NH 0 II 0 I

ID\ Ri o oA0 1 N
0X 0 0=S=0 01\TRI

0=S=0 N

1\lti Rr NrN
%
if 7 -----NH
12O 0 H 48 Cl 0-A = OH
/ ClCI lio N
Ri-NH CI
RN /CI
o/I\TH
) \ l NO
0 Cl0 12iNH
13HN¨R1 49 \
c. z 0 Ri 0 40 N, _ CON¨NH
\ 1 = CI 0 00N 0 \

N\

H //
N-.,,,_ //,_ vSHy Cl 10 Ns / S
Ri 1 -1 0 NH
N¨N C) 1 N

R
H 51 Cl 01 .NIl / C
S
OH c H

HN\
N/ \
RI N 1\T
N

16 Cl Ns 52 a. N

, 0%
(if \
, O
,\ o HN-R1 HN¨R1 17 53 o o e 0.,. OH
NH
1001 /0 r R
/S.,,,. ,...,........-....N.. NH

N-54Cl ce \ 100 NW
cil\IFI
NH .FIN¨R1 HO = 0 s") 0-\
NH
/

V

N/ /
FIN¨Ij 4.0 \ / NH2 N / II
Rio 40 fat 0 "0 HN\

V/
S¨NH2 S¨NH2 = 0 II

=

RI
Ri 0 CI

21 o o V/
S¨NH2 s-NH2 HN¨SI I Cl I I
HN¨S 4.

Ri 0 1 0 Cl Cl Cl Vii\T

NH

HN lj 11 Cl Cl 23 H 59 Cl Cl 40 N 0 0 1\11-CC) 0=S . NH
0 \ 0 ....õ---,S¨NH
FIN¨R1 R1 10 \\

24 0 59 Cl 0 HN 41Ik N c,NH
HO

% \

FIN¨R1 ¨ .

C' 3\1/1\ F
HN¨S . 0 / lj -------N
HN
µ
, Ri\s, s S
F

0\

26 o 61 o 0 ip SV
N NH
....,,) S70 RI
/
NH
/
HN

H
Cl 10 N 101 F3C N
0 1\1 NH 0 \ 0-4-s0 N
e \ FIN-R1 Cl 0 1=1 F3 C 0 N.

NH NH
ys e \ liN) O \

29 H FA 64 Cl 0 H
Cl___ N/ O I\IN *
ys 0H e \
Oys HN-R1 0 e \

HNJI . / ? 1 / H
Ri 0 0, 0 N
= e \
FIN-RI 0 C j N
/

31 0 o\ 66 0 Cl /N R1() N
H
o 0 NH
s \
Ri d Cl 0 N 2\N
H H
1\e Q R1\ Ni . Q R1\ Ni /\N N
N---- N----H H

33 0 0 68 ,(6 N

_NN R V 00 / J
\ H
N\
1:) P HN
Q R1\ .,,/ . 0 N
il R V 41, R1\ Cl 1\ s Cl s N N N
1\1N / I\IN-0 RN .
Cl \ = N
N\ N

QRIN )1 .
N

....õ¨, Ri....,....
....õ.---............
N N N
\ H

o=s=o 1:; ?
RiN q N /
HI "

o H2N
Chlorothiazide and hydrochlorothiazide compounds of Formula III and in particular Table III are useful for the treatment of hypertension, congestive heart failure, osteoporosis, symptomatic edema peripheral edema, kidney stones, diabetes, nephrogenic diabetes insipidus, hypercalcaemia, Dent's disease and Meniere's disease. Compounds of Formula III
and Table III provide sustained release of parent drugs by cleavage of the labile R1 moiety.
Compounds of Formula III, for example 111-63 to 111-71 are useful as prodrugs for the treatment of diabetes.
Scheme 1:
A A
\ \ 0 Ri N¨R1N I I /
A ¨ S ¨ N
X ______ ( X ______________________ I I \
i 0 B

Formula I Formula ll Formula III
\ / /

\
NH I I
A¨ S ¨NH
X _______________________ ( I I \

B
Formula XLV Formula XLVI

The invention further relates the sustained delivery of a compound of Formula XLV
or XLVI by the administration of a compound of Formula I-III as shown in Scheme I. Upon administration of a compound of Formula I-III, the labile R1 moiety may be cleaved off enzymatically, chemically or through first phase metabolism giving a compound of Formula XLV or XLVI. Without being bound to any theory, it is postulated that for some of the compounds of Formula I-III, the release of a compound of Formula XLV or XLVI
upon cleavage of the R1 moiety results in a therapeutically active agent. For example such active ingredient can be aripiprazole, ziprasidone or bifeprunox. In one embodiment, the sustained release comprises a therapeutically effective amount of a compound of Formula XLV or XLVI in the blood stream of the patient for a period of at least about 8, preferably at least about 12, more preferably at least about 24 and even more preferably at least about 36 hours after administration of a compound of Formula I-III. In one embodiment, the compound of Formula XLV or XLVI is present in the blood stream of the patient for a period selected from: at least 48 hours, at least 4 days, at least one week, and at least one month. In one embodiment, a compound of Formula I-III is administered by injection.
Compounds of Formula IX, X, XI, XII, XIII, XIV, XXXIII, XXXIV, XXXV, XXXVI, and )(XXVII are useful for the treatment of neurological and psychological disorders. Neurological and psychiatric disorders include, but are not limited to, disorders such as cerebral deficit subsequent to cardiac bypass surgery and grafting, stroke, cerebral ischemia, spinal cord trauma, head trauma, perinatal hypoxia, cardiac arrest, hypoglycemic neuronal damage, dementia (including AIDS-induced dementia), Alzheimer's disease, Huntington's Chorea, amyotrophic lateral sclerosis, ocular damage, retinopathy, cognitive disorders, idiopathic and drug-induced Parkinson's disease, muscular spasms and disorders associated with muscular spasticity including tremors, epilepsy, convulsions, cerebral deficits secondary to prolonged status epilepticus, migraine (including migraine headache), urinary incontinence, substance tolerance, substance withdrawal (including, substances such as opiates, nicotine, tobacco products, alcohol, benzodiazepines, cocaine, sedatives, hypnotics, etc.), psychosis, schizophrenia, anxiety (including generalized anxiety disorder, panic disorder, social phobia, obsessive compulsive disorder, and post-traumatic stress disorder (PTSD)), mood disorders (including depression, mania, bipolar disorders), circadian rhythm disorders (including jet lag and shift work), trigeminal neuralgia, hearing loss, tinnitus, macular degeneration of the eye, emesis, brain edema, pain (including acute and chronic pain states, severe pain, intractable pain, neuropathic pain, inflammatory pain, and post-traumatic pain), tardive dyskinesia, sleep disorders (including narcolepsy), attention deficit/hyperactivity disorder, eating disorders and conduct disorder.
Definitions Listed below are definitions of various terms used to describe this invention.
These definitions apply to the terms as they are used throughout this specification and claims, unless otherwise limited in specific instances, either individually or as part of a larger group.
The term "aliphatic group" or "aliphatic" refers to a non-aromatic moiety that may be saturated (e.g. single bond) or contain one or more units of unsaturation, e.g., double and/or triple bonds. An aliphatic group may be straight chained, branched or cyclic, contain carbon, hydrogen or, optionally, one or more heteroatoms and may be substituted or unsubstituted. In addition to aliphatic hydrocarbon groups, aliphatic groups include, for example, polyalkoxyalkyls, such as polyalkylene glycols, polyamines, and polyimines, for example.
Such aliphatic groups may be further substituted. It is understood that aliphatic groups may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, and substituted or unsubstituted cycloalkyl groups as described herein.
The term "acyl" refers to a carbonyl substituted with hydrogen, alkyl, partially saturated or fully saturated cycloalkyl, partially saturated or fully saturated heterocycle, aryl, or heteroaryl. For example, acyl includes groups such as (C1-C6) alkanoyl (e.g., formyl, acetyl, propionyl, butyryl, valeryl, caproyl, t-butylacetyl, etc.), (C3-C6)cycloalkylcarbonyl (e.g., cyclopropylcarbonyl, cyclobutylcarbonyl, cyclopentylcarbonyl, cyclohexylcarbonyl, etc.), heterocyclic carbonyl (e.g., pyrrolidinylcarbonyl, pyrrolid-2-one-5-carbonyl, piperidinylcarbonyl, piperazinylcarbonyl, tetrahydrofuranylcarbonyl, etc.), aroyl (e.g., benzoyl) and heteroaroyl (e.g., thiopheny1-2-carbonyl, thiopheny1-3-carbonyl, furany1-2-carbonyl, furany1-3-carbonyl, 1H-pyrroy1-2-carbonyl, 1H-pyrroy1-3-carbonyl, benzo[b]thiopheny1-2-carbonyl, etc.). In addition, the alkyl, cycloalkyl, heterocycle, aryl and heteroaryl portion of the acyl group may be any one of the groups described in the respective definitions. When indicated as being "optionally substituted", the acyl group may be unsubstituted or optionally substituted with one or more substituents (typically, one to three substituents) independently selected from the group of substituents listed below in the definition for "substituted" or the alkyl, cycloalkyl, heterocycle, aryl and heteroaryl portion of the acyl group may be substituted as described above in the preferred and more preferred list of substituents, respectively.

The term "alkyl" is intended to include both branched and straight chain, substituted or unsubstituted saturated aliphatic hydrocarbon radicals/groups having the specified number of carbons. Preferred alkyl groups comprise about 1 to about 24 carbon atoms ("C1-C24") preferably about 7 to about 24 carbon atoms ("C7-C24"), preferably about 8 to about 24 carbon atoms ("C8-C24"), preferably about 9 to about 24 carbon atoms ("C9-C24"). Other preferred alkyl groups comprise at about 1 to about 8 carbon atoms ("C1-C8") such as about 1 to about 6 carbon atoms ("C1-C6"), or such as about 1 to about 3 carbon atoms ("C1-C3").
Examples of C1-C6 alkyl radicals include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, n-pentyl, neopentyl and n-hexyl radicals.
The term "alkenyl" refers to linear or branched radicals having at least one carbon-carbon double bond. Such radicals preferably contain from about two to about twenty-four carbon atoms ("C2-C24") preferably about 7 to about 24 carbon atoms ("C7-C24"), preferably about 8 to about 24 carbon atoms ("C8-C24"), and preferably about 9 to about 24 carbon atoms ("C9-C24"). Other preferred alkenyl radicals are "lower alkenyl"
radicals having two to about ten carbon atoms ("C2-C10") such as ethenyl, allyl, propenyl, butenyl and 4-methylbutenyl. Preferred lower alkenyl radicals include 2 to about 6 carbon atoms ("C2-C6").
The terms "alkenyl", and "lower alkenyl", embrace radicals having "cis" and "trans"
orientations, or alternatively, "E" and "Z" orientations.
The term "alkynyl" refers to linear or branched radicals having at least one carbon-carbon triple bond. Such radicals preferably contain from about two to about twenty-four carbon atoms ("C2-C24") preferably about 7 to about 24 carbon atoms ("C7-C24"), preferably about 8 to about 24 carbon atoms ("C8-C24"), and preferably about 9 to about 24 carbon atoms ("C9-C24"). Other preferred alkynyl radicals are "lower alkynyl"
radicals having two to about ten carbon atoms such as propargyl, 1-propynyl, 2-propynyl, 1-butyne, 2-butynyl and 1-pentynyl. Preferred lower alkynyl radicals include 2 to about 6 carbon atoms ("C2-C6").
The term "cycloalkyl" refers to saturated carbocyclic radicals having three to about twelve carbon atoms ("C3-C12"). The term "cycloalkyl" embraces saturated carbocyclic radicals having three to about twelve carbon atoms. Examples of such radicals include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
The term "cycloalkenyl" refers to partially unsaturated carbocyclic radicals having three to twelve carbon atoms. Cycloalkenyl radicals that are partially unsaturated carbocyclic radicals that contain two double bonds (that may or may not be conjugated) can be called "cycloalkyldienyl". More preferred cycloalkenyl radicals are "lower cycloalkenyl" radicals having four to about eight carbon atoms. Examples of such radicals include cyclobutenyl, cyclopentenyl and cyclohexenyl.
The term "alkylene," as used herein, refers to a divalent group derived from a straight chain or branched saturated hydrocarbon chain having the specified number of carbons atoms. Examples of alkylene groups include, but are not limited to, ethylene, propylene, butylene, 3-methyl-pentylene, and 5-ethyl-hexylene.
The term "alkenylene," as used herein, denotes a divalent group derived from a straight chain or branched hydrocarbon moiety containing the specified number of carbon atoms having at least one carbon-carbon double bond. Alkenylene groups include, but are not limited to, for example, ethenylene, 2-propenylene, 2-butenylene, 1-methy1-2-buten- 1-ylene, and the like.
The term "alkynylene," as used herein, denotes a divalent group derived from a straight chain or branched hydrocarbon moiety containing the specified number of carbon atoms having at least one carbon-carbon triple bond. Representative alkynylene groups include, but are not limited to, for example, propynylene, 1-butynylene, 2-methy1-3-hexynylene, and the like.
The term "alkoxy" refers to linear or branched oxy-containing radicals each having alkyl portions of one to about twenty-four carbon atoms or, preferably, one to about twelve carbon atoms. More preferred alkoxy radicals are "lower alkoxy" radicals having one to about ten carbon atoms and more preferably having one to about eight carbon atoms.
Examples of such radicals include methoxy, ethoxy, propoxy, butoxy and tert-butoxy.
The term "alkoxyalkyl" refers to alkyl radicals having one or more alkoxy radicals attached to the alkyl radical, that is, to form monoalkoxyalkyl and dialkoxyalkyl radicals.
The term "aryl", alone or in combination, means a carbocyclic aromatic system containing one, two or three rings wherein such rings may be attached together in a pendent manner or may be fused. The term "aryl" embraces aromatic radicals such as phenyl, naphthyl, tetrahydronaphthyl, indane and biphenyl.
The terms "heterocyclyl", "heterocycle" "heterocyclic" or "heterocyclo" refer to saturated, partially unsaturated and unsaturated heteroatom-containing ring-shaped radicals, which can also be called "heterocyclyl", "heterocycloalkenyl" and "heteroaryl"
correspondingly, where the heteroatoms may be selected from nitrogen, sulfur and oxygen.
Examples of saturated heterocyclyl radicals include saturated 3 to 6-membered heteromonocyclic group containing 1 to 4 nitrogen atoms (e.g. pyrrolidinyl, imidazolidinyl, piperidino, piperazinyl, etc.); saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms (e.g. morpholinyl, etc.);
saturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms (e.g., thiazolidinyl, etc.). Examples of partially unsaturated heterocyclyl radicals include dihydrothiophene, dihydropyran, dihydrofuran and dihydrothiazole. Heterocyclyl radicals may include a pentavalent nitrogen, such as in tetrazolium and pyridinium radicals. The term "heterocycle" also embraces radicals where heterocyclyl radicals are fused with aryl or cycloalkyl radicals. Examples of such fused bicyclic radicals include benzofuran, benzothiophene, and the like.
The term "heteroaryl" refers to unsaturated aromatic heterocyclyl radicals.
Examples of heteroaryl radicals include unsaturated 3 to 6 membered heteromonocyclic group containing 1 to 4 nitrogen atoms, for example, pyrrolyl, pyrrolinyl, imidazolyl, pyrazolyl, pyridyl, pyrimidyl, pyrazinyl, pyridazinyl, triazolyl (e.g., 4H-1,2,4-triazolyl, 1H-1,2,3-triazolyl, 2H-1,2,3-triazolyl, etc.) tetrazolyl (e.g. 1H-tetrazolyl, 2H-tetrazolyl, etc.), etc.;
unsaturated condensed heterocyclyl group containing 1 to 5 nitrogen atoms, for example, indolyl, isoindolyl, indolizinyl, benzimidazolyl, quinolyl, isoquinolyl, indazolyl, benzotriazolyl, tetrazolopyridazinyl (e.g., tetrazolo[1,5-b]pyridazinyl, etc.), etc.; unsaturated 3 to 6-membered heteromonocyclic group containing an oxygen atom, for example, pyranyl, furyl, etc.; unsaturated 3 to 6-membered heteromonocyclic group containing a sulfur atom, for example, thienyl, etc.; unsaturated 3- to 6-membered heteromonocyclic group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms, for example, oxazolyl, isoxazolyl, oxadiazolyl (e.g., 1,2,4-oxadiazolyl, 1,3,4-oxadiazolyl, 1,2,5-oxadiazolyl, etc.) etc.;
unsaturated condensed heterocyclyl group containing 1 to 2 oxygen atoms and 1 to 3 nitrogen atoms (e.g.
benzoxazolyl, benzoxadiazolyl, etc.); unsaturated 3 to 6-membered heteromonocyclic group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms, for example, thiazolyl, thiadiazolyl (e.g., 1,2,4- thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, etc.) etc.; unsaturated condensed heterocyclyl group containing 1 to 2 sulfur atoms and 1 to 3 nitrogen atoms (e.g., benzothiazolyl, benzothiadiazolyl, etc.) and the like.
The term "heterocycloalkyl" refers to heterocyclo-substituted alkyl radicals.
More preferred heterocycloalkyl radicals are "lower heterocycloalkyl" radicals having one to six carbon atoms in the heterocyclo radical.
The term "alkylthio" refers to radicals containing a linear or branched alkyl radical, of one to about ten carbon atoms attached to a divalent sulfur atom. Preferred alkylthio radicals have alkyl radicals of one to about twenty-four carbon atoms or, preferably, one to about twelve carbon atoms. More preferred alkylthio radicals have alkyl radicals which are "lower alkylthio" radicals having one to about ten carbon atoms. Most preferred are alkylthio radicals having lower alkyl radicals of one to about eight carbon atoms.
Examples of such lower alkylthio radicals include methylthio, ethylthio, propylthio, butylthio and hexylthio.
The terms "aralkyl" or "arylalkyl" refer to aryl-substituted alkyl radicals such as benzyl, diphenylmethyl, triphenylmethyl, phenylethyl, and diphenylethyl.
The term "aryloxy" refers to aryl radicals attached through an oxygen atom to other radicals.
The terms "aralkoxy" or "arylalkoxy" refer to aralkyl radicals attached through an oxygen atom to other radicals.
The term "aminoalkyl" refers to alkyl radicals substituted with amino radicals.
Preferred aminoalkyl radicals have alkyl radicals having about one to about twenty-four carbon atoms or, preferably, one to about twelve carbon atoms. More preferred aminoalkyl radicals are "lower aminoalkyl" that have alkyl radicals having one to about ten carbon atoms. Most preferred are aminoalkyl radicals having lower alkyl radicals having one to eight carbon atoms. Examples of such radicals include aminomethyl, aminoethyl, and the like.
The term "alkylamino" denotes amino groups which are substituted with one or two alkyl radicals. Preferred alkylamino radicals have alkyl radicals having about one to about twenty carbon atoms or, preferably, one to about twelve carbon atoms. More preferred alkylamino radicals are "lower alkylamino" that have alkyl radicals having one to about ten carbon atoms. Most preferred are alkylamino radicals having lower alkyl radicals having one to about eight carbon atoms. Suitable lower alkylamino may be monosubstituted N-alkylamino or disubstituted N,N-alkylamino, such as N-methylamino, N-ethylamino, N,N-dimethylamino, N,N-diethylamino or the like.
The term "substituted" refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: halo, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, thiol, alkylthio, arylthio, alkylthioalkyl, arylthioalkyl, alkylsulfonyl, alkylsulfonylalkyl, arylsulfonylalkyl, alkoxy, aryloxy, aralkoxy, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkyl, amino, trifluoromethyl, cyano, nitro, alkylamino, arylamino, alkylaminoalkyl, arylaminoalkyl, aminoalkylamino, hydroxy, alkoxyalkyl, carboxyalkyl, alkoxycarbonylalkyl, aminocarbonylalkyl, acyl, aralkoxycarbonyl, carboxylic acid, sulfonic acid, sulfonyl, phosphonic acid, aryl, heteroaryl, heterocyclic, and aliphatic. It is understood that the sub stituent may be further substituted.

For simplicity, chemical moieties that are defined and referred to throughout can be univalent chemical moieties (e.g., alkyl, aryl, etc.) or multivalent moieties under the appropriate structural circumstances clear to those skilled in the art. For example, an "alkyl"
moiety can be referred to a monovalent radical (e.g. CH3-CH2-), or in other instances, a bivalent linking moiety can be "alkyl," in which case those skilled in the art will understand the alkyl to be a divalent radical (e.g., -CH2-CH2-), which is equivalent to the term "alkylene." Similarly, in circumstances in which divalent moieties are required and are stated as being "alkoxy", "alkylamino", "aryloxy", "alkylthio", "aryl", "heteroaryl", "heterocyclic", "alkyl" "alkenyl", "alkynyl", "aliphatic", or "cycloalkyl", those skilled in the art will understand that the terms alkoxy", "alkylamino", "aryloxy", "alkylthio", "aryl", "heteroaryl", "heterocyclic", "alkyl", "alkenyl", "alkynyl", "aliphatic", or "cycloalkyl"
refer to the corresponding divalent moiety.
The terms "halogen" or "halo" as used herein, refers to an atom selected from fluorine, chlorine, bromine and iodine.
The terms "compound" "drug", and "prodrug" as used herein all include pharmaceutically acceptable salts, co-crystals, solvates, hydrates, polymorphs, enantiomers, diastereoisomers, racemates and the like of the compounds, drugs and prodrugs having the formulas as set forth herein.
Substituents indicated as attached through variable points of attachments can be attached to any available position on the ring structure.
As used herein, the term "effective amount of the subject compounds," with respect to the subject method of treatment, refers to an amount of the subject compound which, when delivered as part of desired dose regimen, brings about management of the disease or disorder to clinically acceptable standards.
"Treatment" or "treating" refers to an approach for obtaining beneficial or desired clinical results in a patient. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following:
alleviation of symptoms, diminishment of extent of a disease, stabilization (i.e., not worsening) of a state of disease, preventing spread (i.e., metastasis) of disease, preventing occurrence or recurrence of disease, delay or slowing of disease progression, amelioration of the disease state, and remission (whether partial or total).
The term "labile" as used herein refers to the capacity of the prodrug of the invention to undergo enzymatic and/or chemical cleavage in vivo thereby forming the parent drug. As used herein the term "prodrug" means a compounds as disclosed herein which is a labile derivative compound of a heteroaromatic NH-containing parent drug which when administered to a patient in vivo becomes cleaved by chemical and/or enzymatic hydrolysis thereby forming the parent drug such that a sufficient amount of the compound intended to be delivered to the patient is available for its intended therapeutic use in a sustained release manner.
Pharmaceutical Compositions The pharmaceutical compositions of the present invention comprise a therapeutically effective amount of a compound of the present invention formulated together with one or more pharmaceutically acceptable carriers or excipients.
As used herein, the term "pharmaceutically acceptable carrier or excipient"
means a non-toxic, inert solid, semi-solid, gel or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. Some examples of materials which can serve as pharmaceutically acceptable carriers are sugars such as lactose, glucose and sucrose;
cyclodextrins such as alpha- (a), beta- (f3) and gamma- (y) cyclodextrins;
starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt;
gelatin; talc;
excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols such as propylene glycol;
esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline;
Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
The pharmaceutical compositions of this invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. In a preferred embodiment, administration is parenteral administration by injection.
The pharmaceutical compositions of this invention may contain any conventional non-toxic pharmaceutically-acceptable carriers, adjuvants or vehicles. In some cases, the pH
of the formulation may be adjusted with pharmaceutically acceptable acids, bases or buffers to enhance the stability of the formulated compound or its delivery form. The term parenteral as used herein includes subcutaneous, intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions, may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable suspension or emulsion, such as INTRALIPID , LIPOSYN OR OMEGAVEN , or solution, in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. NTRALIPID is an intravenous fat emulsion containing 10-30%
soybean oil, 1-10% egg yolk phospholipids, 1-10% glycerin and water. LIPOSYN is also an intravenous fat emulsion containing 2-15% safflower oil, 2-15% soybean oil, 0.5-5% egg phosphatides 1-10% glycerin and water. OMEGAVEN is an emulsion for infusion containing about 5-25% fish oil, 0.5-10% egg phosphatides, 1-10% glycerin and water.
Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, USP and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.

Additional sustained release in accordance with the invention may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form.
Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissues.
In one preferred embodiment, the formulation provides a sustained release delivery system that is capable of minimizing the exposure of the prodrug to water.
This can be accomplished by formulating the prodrug with a sustained release delivery system that is a polymeric matrix capable of minimizing the diffusion of water into the matrix.
Suitable polymers comprising the matrix include polylactide (PLA) polymers and the lactide/glycolide (PLGA) co-polymers.
Alternatively, the sustained release delivery system may comprise poly-anionic molecules or resins that are suitable for injection or oral delivery. Suitable polyanionic molecules include cyclodextrins and polysulfonates formulated to form a poorly soluble mass that minimizes exposure of the prodrug to water and from which the prodrug slowly leaves.
Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or: a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
Examples of embedding compositions that can be used include polymeric substances and waxes.
Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, eye ointments, powders and solutions are also contemplated as being within the scope of this invention.
The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof Powders and sprays can contain, in addition to the compounds of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons.
Transdermal patches have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.

For pulmonary delivery, a therapeutic composition of the invention is formulated and administered to the patient in solid or liquid particulate form by direct administration e.g., inhalation into the respiratory system. Solid or liquid particulate forms of the active compound prepared for practicing the present invention include particles of respirable size:
that is, particles of a size sufficiently small to pass through the mouth and larynx upon inhalation and into the bronchi and alveoli of the lungs. Delivery of aerosolized therapeutics, particularly aerosolized antibiotics, is known in the art (see, for example U.S. Patent No.
5,767,068 to VanDevanter etal., U.S. Patent No. 5,508,269 to Smith et al., and WO
98/43650 by Montgomery).
By a "therapeutically effective amount" of a prodrug compound of the invention is meant an amount of the compound which confers a therapeutic effect on the treated subject, at a reasonable benefit/risk ratio applicable to any medical treatment. The therapeutic effect may be objective (i.e., measurable by some test or marker) or subjective (i.e., subject gives an indication of or feels an effect).
In accordance with the invention, the therapeutically effective amount of a prodrug of the invention is typically based on the target therapeutic amount of the parent drug.
Information regarding dosing and frequency of dosing is readily available for many parent drugs from which the prodrugs of the invention are derived and the target therapeutic amount can be calculated for each prodrug of the invention. In accordance with the invention, the same dose of a prodrug of the invention provides a longer duration of therapeutic effect as compared to the parent drug. Thus if a single dose of the parent drug provides 12 hours of therapeutic effectiveness, a prodrug of that same parent drug in accordance with the invention that provides therapeutic effectiveness for greater than 12 hours will be considered to achieve a "sustained release".
The precise dose of a prodrug of the invention depends upon several factors including the nature and dose of the parent drug and the chemical characteristics of the prodrug moiety linked to the parent drug. Ultimately, the effective dose and dose frequency of a prodrug of the invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level and dose frequency for any particular patient will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or contemporaneously with the specific compound employed; and like factors well known in the medical arts.
EXAMPLES
Various changes and modifications to the disclosed embodiments will be apparent to those skilled in the art and such changes and modifications including, without limitation, those relating to the chemical structures, substituents, derivatives, formulations and/or methods of the invention may be made.
General methodology for the preparation of lactam compounds can be found in the following publications: U.S.
Patent No. 7,160,888; U.S. Patent No. 5,462,934; U.S. Patent No. 4,914,094;
U.S. Patent No.
4,234,584; U.S. Patent No. 4,514,401; U.S. Patent No. 5,462,934; U.S. Patent No. 4,468,402;
WO 2006/090273 A2; WO 2008/150848 Al; WO 2006/112464 Al; WO 2008/132600 Al.
Hexyl 744-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydrociuinoline-1(2H)-carboxylate (Example 1, Compound 59) CI rN
CI ai HN Cly0 CI rsNC) CI
igr To a solution of diisopropylamine (1.11 mL, 7.87 mmol) in 2-methyltetrahydrofuran (37 mL) at -5 C was added n-BuLi (3.0 mL, 2.5 M in hexanes, 7.49 mmol) slowly. After 20 minutes, the reaction was cooled to -78 C and Aripiprazole (1.68 g, 3.74 mmol) was added. After a further 10 minutes, hexylchloroformate (1.53 mL, 9.37 mmol) was added. The reaction was held at -78 C for 2 hours before allowing to warm to room temperature overnight. The reaction was quenched with saturated aqueous ammonium chloride solution (30 mL) and extracted with ethyl acetate (2 x 30 mL). The organics were combined, washed with saturated aqueous sodium hydrogen carbonate solution (30 mL), brine (30 mL), dried over MgSO4 and concentrated. The crude product was purified by column chromatography on silica eluting with 0 to 3% tetrahydrofuran in ethyl acetate. The product was triturated in heptane to remove aliphatic impurities and then filtered and dried to afford Compound-59 (0.487 g) as a colorless solid.
1H-NMR (300 MHz, CDC13) 6 7.14 (2H, m), 7.07 (1H, d), 6.95 (1H, m), 6.62 (1H, dd), 6.54 (1H, d), 4.38 (2H, t), 3.95 (2H, t), 3.06 (4H, m), 2.89 (2H, t), 2.66 (6H, m), 2.47 (2H, t), 1.90 ¨ 1.65 (6H, m), 1.49 ¨ 1.30 (6H, m), 0.88 (3H, t). [M+H] ' = 576.2.
Isopropyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-l-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (Example 2, Compound 75) To a solution of diisopropylamine (1.11 mL, 7.87 mmol) in 2-methyltetrahydrofuran (37 mL) at -5 C was added n-BuLi (3.0 mL, 2.5 M in hexanes, 7.49 mmol) slowly. After 20 minutes, the reaction was cooled to -78 C and Aripiprazole (1.68 g, 3.74 mmol) was added. After a further 10 minutes, isopropylchloroformate (9.37 mL, 1.0 mol in toluene, 9.37 mmol) was added. The reaction was held at -78 C for 2 hours before allowing to warm to room temperature overnight. The reaction was quenched with saturated aqueous ammonium chloride solution (30 mL) and extracted with ethyl acetate (2 x 30 mL). The organics were combined, washed with saturated aqueous sodium hydrogen carbonate solution (30 mL), brine (30 mL), dried over MgSO4 and concentrated. The crude product was purified by column chromatography on silica eluting with 1: 1 ethyl acetate to dichloromethane to 2%
methanol in 1: 1 ethyl acetate to dichloromethane to give the product. The product was recrystallized from isopropanol to remove aliphatic impurities. The product was not sufficiently pure so was purified by column chromatography eluting with 0 to 10%
tetrahydrofuran in ethyl acetate. The product was triturated in heptane and filtered to afford Compound-75 (0.593 g) as a colorless solid.
1H-NMR (300 MHz, CDC13) 6 7.16 (2H, m), 7.05 (1H, d), 6.95 (1H, m), 6.60 (1H, dd), 6.52 (1H, d), 5.22 (1H, quintet), 3.95 (2H, t), 3.07 (4H, m), 2.89 (2H, t), 2.66 (6H, m), 2.47 (2H, t), 1.81 (2H, m), 1.74 (2H, m), 1.42 (3H, s), 1.40 (3H, s). [M+H] ' = 534.2.

7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-N-ethyl-3,4-dihydro-2-oxoquinoline-1(2H)-carboxamide (Example 3, Compound 72) To a solution of aripiprazole (1.5 g, 3.35 mmol) in dichloromethane (33 mL) was added triethylamine (0.56 mL, 4.01 mmol) and ethyl isocyanate (0.53 mL, 6.69 mmol).
The reaction was stirred at room temperature for 5 days. The reaction was quenched with water and extracted with dichloromethane (3 x 20 mL). The organics were combined, washed with saturated aqueous sodium hydrogen carbonate (20 mL) and brine (20 mL), dried over MgSO4 and concentrated. The product was purified by column chromatography on silica eluting with 0 ¨ 3% methanol in 1: 1 ethyl acetate to dichloromethane. The product was triturated in heptane to remove aliphatic impurities and then filtered and dried. The material was dissolved in ethyl acetate and washed with saturated aqueous sodium hydrogen carbonate (6 x 15 mL) to afford Compound-72 (0.482 g) as a pink solid.
1H-NMR (300 MHz, CDC13) 6 8.44 (1H, t), 7.14 (2H, m), 7.05 (1H, d), 6.94 (2H, m), 6.65 (1H, dd), 3.96 (2H, t), 3.43 (2H, quintet), 3.07 (4H, m), 2.79 (2H, m), 2.67 (6H, m), 2.47 (2H, t), 1.80 (2H, m), 1.69 (2H, m), 1.26 (3H, t). [M+H] = 519.2.
N-benzy1-7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxamide (Example 4, Compound 79) To a solution of aripiprazole (1.5 g, 3.35 mmol) in dichloromethane (33 mL) was added triethylamine (0.56 mL, 4.01 mmol) and benzyl isocyanate (0.82 mL, 6.69 mmol).
The reaction was stirred at room temperature for 48 hours. The reaction was quenched with water and extracted with dichloromethane (3 x 20 mL). The organics were combined, washed with brine (20 mL), dried over MgSO4 and concentrated. The residue was purified by column chromatography on silica eluting with 0 ¨ 10% tetrahydrofuran in ethyl acetate. The product was triturated in heptane to remove aliphatic impurities and then filtered and dried to afford Compound-79 (0.575 g) as a colorless solid.
1H-NMR (300 MHz, CDC13) 6 8.90 (1H, t), 7.44 ¨7.28 (5H, m), 7.15 (2H, m), 7.05 (1H, d), 6.97 (2H, m), 6.66 (1H, dd), 4.60 (2H, d), 3.96 (2H, t), 3.07 (4H, m), 2.81 (2H, m), 2.67 (6H, m), 2.48 (2H, t), 1.81 (2H, m), 1.69 (2H, m). [M+H] ' = 581.2.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-3,4-dihydroquinolin-2-y1 dibenzylcarbamate (Example 5, Compound 242) A mixture of aripiprazole (1.5 g, 3.3 mmol), dibenzylcarbamoyl chloride (1.74 g, 6.7 mmol) silver carbonate (3.75 g, 13.4 mmol) and 2-methyltetrahydrofuran (30 mL) was heated at reflux for 4 days. The reaction mixture was cooled, diluted with ethyl acetate and water, and then filtered through celite*. The organic phase was separated, dried over MgSO4 and evaporated. The residue obtained was further purified on silica eluting with ethyl acetate/tetrahydrofuran to give after evaporation of the major product containing fractions, Compound-80 (1.02 g) was a yellow oil.
1H-NMR (300 MHz, CDC13) 6 7.44-7.21 (m, 8H), 7.20-7.10 (in, 4H), 7.04 (d, 1H), 7.00-6.93 (m, 1H), 6.57 (dd, 1H), 6.40 (d, 1H), 4.80 (d, 1H), 4.54 (d, 1H), 4.35 (d, 1H), 4.26 (d, 1H), 3.90-3.81 (m, 1H), 3.74-3.67 (m, 1H), 3.15-3.03 (m, 4H), 2.92-2.80 (m, 2H), 2.75-2.60 (m, 6H), 2.65 (t, 2H), 2.84-2.63 (in, 4H). [M+11]1 = 671.3.
7-(4-(4-(2,3-dichlorophenyl)piperazin- -yl)butoxy)-3,4-dihydroquin olin-2-y1 diethylcarbamate (Example 6, Compound 234) Compound-51 was synthesized in a similar manner to Compound-80, (0.80 g) as a yellow oil.
1H-NMR (300 MHz, CDC13) 6 7.17-7.12 (2H, m), 7.07 (d, 1H), 7.99-7.92 (m, 1H), 6.57 (dd, 1H), 6.39 (dd, 1H), 4.00-3.85 (m, 2H), 3.76-3.62 (m, 2H), 3.54-3.40 (m, 2H), 3.25-3.15 (m, 2H), 3.12-3.01 (in, 4H), 2.92 (t, 2H), 2.76-2.56 (m, 6H), 2.47 (t, 2H), 1.84-1.60 (m, 4H), 1.31 (t, 3H), 1.10 (t, 3H). [M+H] = 547.2.
Methyl 7-(4-(4-(2,3-dichlorophenybpiperazin-l-yl)butoxy)-2-oxo-3A-dihydrouuinoline-1(2H)-carboxylate (Example 7, Compound 74) To a solution of diisopropylamine (0.99 mL, 7.02 mmol) in 2-methyltetrahydrfuran (33 mL) at -5 C was added n-BuLi (2.67 mL, 2.5 M in hexanes, 6.69 mmol) slowly. After minutes, the reaction was cooled to -78 C and aripiprazole (1.50 g, 3.34 mmol) was added.
After a further 10 minutes, methylchloroformate (0.65 mL, 8.36 mmol) was added. The reaction was held at -78 C for 2 hours before allowing to warm to room temperature. After 2 hours the reaction was quenched with saturated aqueous ammonium chloride solution (30 mL) and extracted with ethyl acetate (2 x 30 mL). The organics were combined, washed with saturated aqueous sodium hydrogen carbonate solution (30 mL), brine (30 mL), dried over MgSO4 and concentrated. The crude product was purified by column chromatography on silica eluting with 1: 1 ethyl acetate to dichloromethane to 10% methanol in 1: 1 ethyl acetate to dichloromethane to give the product. The product was triturated in heptane to remove aliphatic impurities and then filtered to afford Compound-74 (0.824 g) as a colorless solid.
Trademark*

1H-NMR (300 MHz, CDC13) 6 7.15 (2H, m), 7.06 (1H, d), 6.95 (1H, m), 6.63 (1H, dd), 6.56 (1H, d), 3.99 (3H, s), 3.95 (2H, t), 3.07 (4H, m), 2.89 (2H, t), 2.67 (6H, m), 2.48 (2H, t), 1.81 (2H, m), 1.72 (2H, m). [M+H] ' = 506.1.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-1-isobutyry1-3,4-dihydroq uinolin-2(1H)-one (Example 8, Compound-16) To a solution of diisopropylamine (1.3 mL, 8.9 mmol) in tetrahydrofuran (25mL) at 78 C was added n-BuLi (2.2M in hexanes, 4.1 mL, 8.9 mmol). The reaction mixture was warmed to 0 C and after 10min was re-cooled to 78 C and aripiprazole (2.0 g, 4.5 mmol) added. The reaction mixture was stirred for 30min and then isobutyryl chloride (0.7 mL, 6.7 mmol) added. After 2 h the reaction mixture was warmed to room temperature and stirred for 1 h. A
second reaction was carried out under exactly the same conditions and the two reactions were combined. This mixture was washed with water, dried over MgSO4, filtered and evaporated.
The residue was dissolved in diethyl ether and 4M HC1 in diethyl ether added to form a precipitate. The mother liquor was decanted and the residual gum was washed with diethyl ether. The gum was then partitioned between ethyl acetate and saturated sodium bicarbonate, and the organic layer separated. After drying over MgSO4 and evaporation, the residue was further purified on silica eluting with 1:1:0.1 dichloromethane/ethyl acetate/methanol to give Compound-16 (1.3 g) as a yellow solid.
1H-NMR (300 MHz, CDC13) 6 7.16-7.13 (m, 2H), 7.08 (d, 1H), 6.98-6.92 (m, 1H), 6.65-6.59 (m, 2H), 3.93 (t, 2H), 3.48 (dt, 1H), 3.12-3.01 (m, 4H), 2.86 (dd, 1H), 2.72-2.59 (m, 6H), 2.47 (t, 2H), 1.84-1.64 (m, 4H), 1.25 (d, 6H). [M+H] ' = 518.2.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-3,4-dihydroquinolin-2-ylpivalate (Example 9, Compound-216) To a stirred solution of aripiprazole (0.1g, 0.223 mmol) in pyridine (1mL) at 0 C was added pivaloyl chloride (0.055 mL, 0.446 mmol). After stirring at 0 C for 5 minutes the reaction was allowed to warm to room temperature. After a further 5 minutes the temperature was increased to 50 C for approximately 19 hours. The reaction was allowed to cool to room temperature. The reaction was repeated in a similar manner using of aripiprazole (1.75g, 3.90 mmol). The two reaction mixtures were combined and quenched with approximately methanol (5mL). The majority of the pyridine was removed in vacuo and the residue partitioned between dichloromethane (30mL) and saturated NaHCO3 solution (30mL). The aqueous phase was extracted with dichloromethane (2x30mL) and the combined organic extracts washed with brine (20mL) and dried over MgSO4. After filtration, the volatiles were removed (toluene and methanol/dichloromethane azeotrope) and the residue purified by silica chromatography eluting first with dichloromethane followed by ethyl acetate/dichloromethane/methanol (1:1:0.04) to give the title compound (1.19g, 54%).
1H NMR (CDC13, 300MHz) 6 7.13-7.17 (m, 2H), 7.09 (d, 1H), 6.99-6.92 (m, 1H), 6.57 (dd, 1H), 6.24 (d, 1H), 3.90 (t, 2H), 3.2-3.0 (m, 4H), 2.94-2.83 (m, 2H), 2.78-2.4 (m, 8H), 1.85-1.45 (m, 4H), 1.3 (s, 9H); m/z (M11) 532.26.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-1-hexanoy1-3,4-dihydroq uinolin-2(1H)-one (Example 10, Compound-4) To a stirred solution of diisopropylamine (1.26 mL, 8.92 mmol) in 2-methyltetrahydrofuran (40 mL) at 7 C was added 1.47 M butyl lithium in hexanes (6.07 mL, 8.92 mmol) dropwise keeping the temperature between 0 C and 5 C. After stirring at 7 C for 20 minutes the reaction was cooled to 78 C. A suspension of aripiprazole (2 g, 4.46 mmol) in methyltetrahydrofuran (40 mL) was added to the lithium diisopropylamide (LDA) solution keeping the temperature below -65 C. After 10 minutes hexanoic anhydride (2.58 mL, 11.15 mmol) was added dropwise and the reaction stirred at 78 C under argon gas.
After 2.5 hours the reaction was allowed to warm to room temperature (removal of bath). After a further 40 minutes the reaction was quenched with saturated aqueous NH4C1 (50 mL) and diluted with ethyl acetate (100 mL). The aqueous layer was extracted with ethyl acetate (2 x 50 mL) and the combined organics washed with water (50 mL), saturated aqueous NaHCO3 (3 x
50 mL), brine (50 mL) and dried over MgSO4. After filtration, the volatiles were removed. The crude mixture was purified by silica chromatography eluting with 1% methanol (1:1 ethyl acetate /
dichloromethane). This material was further purified by partitioning between ethyl acetate (50 mL) and sat. aqueous NaHCO3 (50 mL). The organic layer was washed with sat. aqueous NaHCO3 (2 x 50 mL), brine (50 mL) and dried over MgSO4. After filtration, the volatiles were removed to give Compound-4, 0.95 g.
1H-NMR (300MHz, CDC13) 6 7.18-7.12 (2H, m), 7.12-7.05 (1H, m), 6.99-6.92 (1H, m), 6.81 (1H, d), 6.66 (1H, dd), 3.95 (2H, t), 3.13-3.01 (4H, bs), 2.97 (2H, t), 2.87-2.78 (2H, m), 2.74-2.57 (6H, m), 2.48 (2H, t), 1.87-1.64 (6H, m), 1.43-1.29 (4H, m), 0.90 (3H, m). m/z [M+H]
546.1.

7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-1-dodecanoy1-3,4-dihydroquinolin-2(1H)-one (Example 11, Compound-7) Compound-7 was synthesized in a similar manner to Compound-4 in Example 10.
1H-NMR (300MHz, CDC13) 6 7.18-7.12 (2H, m), 7.12-7.05 (1H, m), 6.99-6.92 (1H, m), 6.81 (1H, d), 6.65 (1H, dd), 3.95 (2H, t), 3.17-3.01 (4H, bs), 2.97 (2H, t), 2.88-2.78 (2H, m), 2.75-2.56 (6H, m), 2.49 (2H, bt), 1.87-1.56 (6H, m), 1.45-1.17 (16H, m), 0.87 (3H, t) m/z [M+H] 629.9.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)quinolin-2-y1 hexyl carbonate (Example 12, Compound-328) To a mixture of dehydro-aripiprazole (1.5 g, 3.4 mmol), potassium tert-butoxide (0.75 g, 6.7 mmol) and 2-methyltetrahydrofuran (30 mL) at 0 C was added hexyl chloroformate (1.32 mL, 8.1 mmol). The reaction mixture was stirred for 2h, allowed to self warm to room temperature and stirred for a further 4h. The reaction mixture was then diluted with water and extracted with ethyl acetate. The organic phase was dried over MgSO4, filtered and evaporated. The residue was further purified on silica eluting with ethyl acetate/tetrahydrofuran and the major product containing fractions evaporated to give a yellow solid. This was triturated in heptane (30mL) for 2.5h, filtered and dried to give Compound-328 (1.55g) as a white solid.
1H-NMR (300 MHz, CDC13) 6 8.15 (d, 1H), 7.71 (d, 1H), 7.32 (d, 1H), 7.21-7.12 (m, 3H), 7.09 (d, 1H), 6.97-6.91 (m, 1H), 4.29 (t, 1H), 4.13 (t, 1H), 3.12-3.01 (m, 4H), 2.73-2.55 (m, 4H), 2.50 (t, 2H), 1.95-1.85 (m, 2H), 1.82-1.70 (m, 4H), 1.50-1.28 (m, 6H), 0.94-0.86 (m, 3H). [M+H] = 574.2.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)quinolin-2-y1 isopropyl carbonate (Example 13, Compound-323) Compound-323 was synthesized in a similar manner as Compound-328 in Example 12.
1H-NMR (300 MHz, CDC13) 6 8.14 (d, 1H), 7.70 (d, 1H), 7.32 (d, 1H), 7.20-7.12 (m, 3H), 7.07 (d, 1H), 6.97-6.91 (m, 1H), 5.16-4.98 (m, 1H), 4.13 (t, 2H), 3.11-3.02 (m, 4H), 2.72-2.59 (m, 4H), 2.50 (t, 2H), 1.94-1.84 (m, 2H), 1.80-1.68 (m, 2H), 1.40 (d, 6H). [M+H]' =
532.1.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)quinolin-2-y1 diethylcarbamate (Example 14, Compound-334) A mixture of dehydro-aripiprazole (1.50 g, 3.36 mmol), triethylamine (1.03 mL, 7.39 mmol), diethyl carbamoyl chloride (1.02 mL) were combined in tetrahydrofuran (30 mL).
This was then heated to 100 C for 6 hours by microwave. The reaction was quenched with water (50 mL) and extracted with dichloromethane (2 x 100 mL). The combined organics were dried (MgSO4) and concentrated. The crude product was purified by column chromatrography on silica eluting with ethyl acetate to 20% tetrahydrofuraniethyl acetate to give the product. The product was then triturated with heptane to remove aliphatic impurities and then dried to give Compound-334 (1.54 g) as a light brown oil.
1H-NMR (300 MHz, CDC13) 6 8.10 (1H, d), 7.69 (1H, d), 7.31 (1H, d), 7.16-7.07 (4H, m), 6.99-6.92 (1H, m), 4.12 (2H, t), 3.54-3.39 (4H, 2 x q), 3.12-2.96 (4H, br s), 2.78-2.54 (4H, br s), 2.50 (2H, t), 1.97-1.62 (4H, m), 1.32-1.16 (6H, 2 x t). [M+H] ' = 545.2.
N,N-diethy1-7-(4-(4-(2,3-dichlorophenybpiperazin- 1-yl)butoxy)-2-oxoquinoline-1(2H)-carboxamide (Example 15, Compound-150) Compound-334 was synthesized as described above. Compound-334 (2.17 g, 3.99 mmol) was dissolved in pyridine (10 mL) and heated in the microwave to 175 C for 5 hours. The reaction was diluted with ethyl acetate (10 mL) and concentrated, co-evaporating with toluene (3 x 5 mL). The product was purified by column chromatography on silica eluting with 10 to 30% tetrahydrofuran in ethyl acetate to provide Compound-150 (0.81 g) as a yellow oil.
1H-NMR (300 MHz, CDC13) 6 7.64 (1H, d), 7.45 (1H, d), 7.16 (2H, m), 6.96 (1H, m), 6.89 (1H, dd), 6.54 (1H, d), 6.47 (1H, d), 4.04 (2H, m), 3.78 (1H, m), 3.57 (1H, m), 3.28-3.00 (6H, m), 2.66 (4H, m), 2.49 (2H, m), 1.85 (2H, m), 1.72 (2H, m), 1.39 (3H, t), 1.08 (3H, t).
[M+H] ' = 545.2.
7-(4-(4-(2,3-dichlorophenybpiperazin-l-yl)butoxy)quinolin-2-y1 dibenzylcarbamate (Example 16, Compound-342) To a solution of dibenzylcarbamoyl chloride (1.7 g, 6.7 mmol) in pyridine (15 mL) was added dehydro-aripiprazole (1.5 g, 3.4 mmol) and the reaction mixture heated at reflux for 4h. The reaction mixture was concentrated and the residue co-evaporated with toluene (x3).
The residue was dissolved in ethyl acetate (ethyl acetate), washed with water and dried over MgSO4. After evaporation the residue was further purified on silica eluting with ethyl acetate and after drying gave Compound-342 (0.71g) as a white solid.

1H-NMR (300 MHz, CDC13) 6 8.13 (d, 1H), 7.70 (d, 1H), 7.42-7.29 (m, 11H), 7.20-7.08 (m, 4H), 7.00-6.92 (m, 1H), 4.64 (s, 2H), 4.56 (s, 2H), 4.14 (t, 2H), 3.15-3.02 (m, 4H), 2.74-2.60 (m, 4H), 2.58-2.48 (m, 2H), 1.98-1.88 (m, 2H), 1.83-1.70 (m, 2H). [M+H] ' =
669.1.
N,N-dibenzy1-7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-2-oxoquinoline-1(2H)-carboxamide (Example 17, Compound-179) A solution of Compound-342 (0.94 g, 1.4 mmol) in pyridine (10 mL) was heated in a microwave at 175 C for 10h . The reaction mixture was evaporated and then co-evaporated with toluene. The residue was further puriifed on silica eluting with ethyl acetate to give after evaporation of the product containing fractions, Compound-179 (0.36 g) as a yellow oil.
1H-NMR (300 MHz, CDC13) 6 7.65 (1H, d), 7.51 (2H, d), 7.39 (5H, m), 7.23 (2H, m), 7.16 (4H, m), 6.96 (1H, m), 6.77 (1H, dd), 6.50 (1H, d), 6.39 (1H, s), 4.89 (1H, d), 4.64 (1H, d), 4.27 (2H, d), 3.85 (1H, m), 3.65 (1H, m), 3.10 (4H, m), 2.68 (4H, m), 2.52 (2H, m), 1.79 (2H, m), 1.72 (2H, m). [M+H] ' = 669.1.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)quinolin-2-y1 diethyl phosphate (Example 18, Compound-331) To a solution of dibenzyl phosphate (2.48 g, 8.91 mmol) in dichloromethane (25 ml) was added N,N-dimethyl formamide (1 drop) followed by oxalyl chloride (0.75 mL, 8.91 mmol).
After 2 hours the reaction mixture was concentrated in vacuo. The residue was dissolved in 2-methyltetrahydrfuran (5 mL) and added to a suspension of dehydro-aripiprazole (1.66 g, 3.71 mmol) and potassium t-butoxide (0.92 g, 8.17 mmol) in 2-methyltetrahydrfuran (35 mL) at 0 C under argon gas, then allowed to gradually warm to room temperature. After stirring overnight the reaction was quenched with water (25 mL) and 28% aq NH3 (15 mL) and stirred for 10 minutes. The reaction mixture was then extracted with ethyl acetate (2 x 40 mL). The combined organics were washed with water (50 mL) and brine (50 mL) then dried (MgSO4) and concentrated in vacuo. The crude mixture was purified by column chromatography eluting with 5% methanol (1:1 ethyl acetate/dichloromethane) to give the product. The 1H-NMR showed minor impurities still present (impurities not observed by LCMS). Various further purifications were attempted but failed to remove these impurities.
1H-NMR (300MHz, CDC13) 6 8.04 (1H, d), 7.65 (1H, d), 7.44 ¨ 7.10 (14H, m), 6.95 (2H, m), 5.33 (4H, m), 4.09 (2H, t), 3.06 (4H, m), 2.65 (4H, m), 2.50 (2H, t), 1.90 (2H, m), 1.74 (2H, m). [M+H] ' 707.8.

7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)quinolin-2-y1 dibenzyl phosphate (Example 19, Compound-322) To a solution of dibenzyl phosphate (2.48 g, 8.91 mmol) in dichloromethane (25 ml) was added dimethyl formamide (1 drop) followed by oxalyl chloride (0.75 mL, 8.91 mmol). After 2 hours the reaction mixture was concentrated in vacuo. The residue was dissolved in 2-methyltetrahydrfuran (5 mL) and added to a suspension of dehydro-aripiprazole (1.66 g, 3.71 mmol) and potassium t-butoxide (0.92 g, 8.17 mmol) in 2-methyltetrahydrfuran (35 mL) at 0 C under Ar (g) then allowed to gradually warm to room temperature. After stirring overnight the reaction was quenched with water (25 mL) and 28% aq NH3 (15 mL) and stirred for 10 minutes. The reaction mixture was then extracted with ethyl acetate (2 x 40 mL). The combined organics were washed with water (50 mL) and brine (50 mL) then dried (MgSO4) and concentrated in vacuo. The crude mixture was purified by column chromatography eluting with 5% methanol (1:1 ethyl acetate/dichloromethane) to give the product. The 1H-NMR showed minor impurities still present (impurities not observed by LCMS). Various further purifications were attempted but failed to remove these impurities.
1H-NMR (300MHz, CDC13) 6 8.04 (1H, d), 7.65 (1H, d), 7.44 ¨7.10 (14H, m), 6.95 (2H, m), 5.33 (4H, m), 4.09 (2H, t), 3.06 (4H, m), 2.65 (4H, m), 2.50 (2H, t), 1.90 (2H, m), 1.74 (2H, m). [M+H]+ 707.8.
7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)quinolin-2-ylpivalate (Example 20, Compound-316) To a solution of dehydro-aripiprazole (1.2 g, 2.7mmol) in dichloromethane (30 mL) was added pyridine (1 mL), followed by pivaloyl chloride (0.66 mL, 5.4 mmol). The reaction mixture was stirred for 20 hours, then washed with water and dried over MgSO4.
After evaporation the residue was co-evaporated with toluene and then further purified on silica eluting with ethyl acetate. After evaporation of the product containing fraction, Compound-316 (0.53g) was obtained as a colourless oil.
1H-NMR (300MHz, CDC13) 6 8.12(d, 1H), 7.70 (d, 1H), 7.34(d, 1H), 7.20-7.11 (m, 3H), 7.00-6.92 (m, 2H), 4.12 (t, 2H), 3.18-3.02 (m, 4H), 2.77-2.45 (m, 6H), 1.95-1.85 (m, 2H), 1.83-1.72 (m, 2H), 1.42 (s, 9H). [M+H] 530.1.
Acetoxymethyl 7-(4-(4-(2,3-dichlorophenybpiperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (Example 21, Compound-87) CI
Sc' N

The reactions were carried out in 2 x 1 g batches side by side.
To a suspension of aripiprazole (2 g, 4.46 mmol) in 2-methyltetrahydrofuran (40 mL) was added NaH (357 mg, 8.92 mmol). After 20 minutes, further 2-methyltetrahydrofuran (20 mL) was added to aid stirring. Chloromethyl chloroformate (1.19 mL, 13.38 mmol) was then added and the reaction stirred for 2 days. The reactions were combined for work up. The reaction was cooled to 0 C, diluted with ethyl acetate (50 mL) and quenched with aqueous saturated NaHCO3 solution (50 mL). The reaction was extracted with ethyl acetate (3 x 50 mL) and the combined organics washed with brine (100 mL), then dried (MgSO4) and concentrated to give chloromethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (crude yield 2.59g) as 3:2 mixture of product/aripiprazole. The product was not purified due to instability during silica chromatography and so taken onto the next step crude.
Synthesis of Compound 87 To a solution of chloromethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (1.20 g, 2.22 mmol) in dimethyl formamide (20 mL) was added cesium acetate (639 mg, 3.33 mmol). The reaction was then stirred at ambient temperature overnight. The reaction was diluted with ethyl acetate (50 mL) and quenched with water/brine (1:1, 50 mL). The reaction was extracted with ethyl acetate (3 x 50 mL). The combined organics were washed with water (50 mL) then brine (50 mL) and dried (Mg504) and concentrated. The crude product was purified by column chromatography on silica eluting with 0-10% tetrahydrofuran/dichloromethane with 1% Et3N to give the product which required further purification. The product was purified by column chromatography on silica eluting with 0.5% Et3N/ethyl acetate to give acetoxymethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate, Compound-87 (243 mg, 19% over 2 steps).

1H-NMR (300 MHz, CDC13) 6 7.17-7.08 (3H, m), 6.99-6.89 (1H, m), 6.66-6.58 (2H, m), 5.95 (2H, s), 3.96 (2H, t), 3.12-3.01 (4H, m), 2.89-2.85 (2H, m), 2.69-2.65 (6H, m), 2.50-2.45 (2H, m), 2.17 (3H, s), 1.86-1.59 (4H, m). [M+H] ' = 564.17.
Butyryloxymethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (Example 22, Compound-88) CI
Sc' N
N 0 el To a solution of chloromethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (1.20 g, 2.22 mmol) in dimethyl formamide (20 mL) was added butyric acid (0.30 mL, 3.33 mmol) followed by cesium carbonate (542 mg, 1.67 mmol). The reaction was then stirred at ambient temperature overnight. The reaction was diluted with ethyl acetate (50 mL) and quenched with water/brine (1:1, 50 mL).
The reaction was extracted with ethyl acetate (3 x 50 mL). The combined organics were washed with water (50 mL) then brine (50 mL) and dried (MgSO4) and concentrated. The crude product was purified by column chromatography on silica eluting with 010%
tetrahydrofuran/dichloromethane with 1% Et3N to give the product which required further purification. The product was purified by column chromatography on silica eluting with 0.5-1.5% Et3N/ethyl acetate to give butyryloxymethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin- 1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate, Compound-88 (348 mg, 26%
over 2 steps).
1H-NMR (300 MHz, CDC13) 6 7.17-7.07 (3H, m), 7.01-6.89 (1H, m), 6.66-6.58 (2H, m), 5.97-5.92 (2H, m), 3.96 (2H, t), 3.07-2.99 (4H, m), 2.89-2.84 (2H, m), 2.70-2.58 (6H, m), 2.51-2.37 (4H, m), 1.86-1.61 (6H, m), 0.96 (3H, m). [M+H] = 592.28.
Palmitoyloxymethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-y1)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (Example 23, Compound-89) CI

N

Synthesis of chloromethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate To a suspension of aripiprazole (1 g, 2.23 mmol) in 2-methyltetrahydrofuran (20 mL) was added NaH (178 mg, 4.46 mmol). After 1 hour, a further of NaH (178 mg) was added. The reaction was stirred at ambient temperature overnight then heated to reflux for 3 hours. The reaction was cooled to room temperature and chloromethyl chloroformate (0.60 mL, 6.69 mmol) was then added and the reaction stirred for 2 hours. The reaction was cooled to 0 C, diluted with ethyl acetate (20 mL) and quenched with aqueous saturated NaHCO3 solution (20 mL). The reaction was extracted with ethyl acetate (3 x 20 mL) and the combined organics washed with brine (50 mL), then dried (Mg504) and concentrated to give chloromethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (crude yield 1.54g) as a 2:1 mixture of product/aripiprazole. The product was taken onto the next step crude.
Synthesis of Compound-89 To a solution of chloromethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (1.16 g, 2.14 mmol) in dimethyl formamide (20 mL) was added palmitic acid (825 mg, 3.22 mmol) and cesium carbonate (524 mg, 1.61 mmol).
The reaction was heated to 60 C for 5 hours then allowed to cool to room temperature. The reaction was diluted with ethyl acetate (20 mL) and washed with 1:1 water/brine (3 x 40 mL).
The organics were filtered to remove a white precipitate then dried (Mg504) and concentrated. The crude product was purified by column chromatography on silica eluting with 0.5% Et3N/ethyl acetate to give palmitoyloxymethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-l-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate, Compound-89 (548 mg, 32% over 2 steps). The material was determined to be 83%
pure by LCMS.

1H-NMR (300 MHz, CDC13) 6 7.17-7.07 (3H, m), 6.99-6.93 (1H, m), 6.67-6.59 (2H, m), 5.96-5.91 (2H, m), 3.96 (2H, t), 3.11-3.03 (4H, m), 2.92-2.83 (2H, m), 2.70-2.65 (6H, m), 2.52-2.38 (4H, m), 1.83-1.57 (6H, m), 1.35-1.15 (24H, m), 0.87 (3H, t). [M+H]
' = 760.48.
1-(butyryloxy)ethyl 7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate) (Example 24, Compound-90) CI
Sc' N
N 0 el )00 Synthesis of 1-chloroethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate To a suspension of aripiprazole (2.0 g, 4.46 mmol) in 2-methyltetrahydrofuran (50 mL) was added NaH (357 mg, 8.92 mmol). The reaction was stirred overnight at room temperature then chloroethyl chloroformate (1.46 mL, 13.38 mmol) was added. Further 2-methyltetrahydrofuran (10 mL) was added to aid stirring. The reaction was stirred overnight at room temperature. The reaction was cooled to 0 C, diluted with ethyl acetate (50 mL) and quenched with aqueous saturated NaHCO3 (50 mL). The reaction was extracted with ethyl acetate (3 x 50 mL). The combined organics were dried (Mg504) and concentrated to give 1-chloroethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (crude yield 2.22 g) as a 3:2 mixture of aripiprazole/product. The product was taken onto the next step without purification.
Synthesis of Compound-90 1-chloroethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (2.0 g, 3.60 mmol) was stirred with butyric acid (1.0 mL, 10.81 mmol) and ethyldiisopropylamine (0.94 mL, 5.41 mmol) at 50 C for 48 hours. The reaction was diluted with diethyl ether (20 mL) and quenched with aqueous saturated NaHCO3 (40 mL). The reaction was extracted with ethyl acetate (2 x 50 mL) and the combined organics were washed with water (50 mL), brine (50 mL) then dried (Mg504). The crude product was purified by column chromatography on silica eluting with 0.5% Et3N/ethyl acetate to give 1-(butyryloxy)ethyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate, Compound-90 (628 mg, 23% over 2 steps).
1H-NMR (300 MHz, CDC13) 6 7.18-6.89 (5H, m), 6.67-6.58 (2H, m), 4.01-3.93 (2H, m), 3.11-2.99 (4H, m), 2.94-2.83 (2H, m), 2.69-2.57 (6H, m), 2.53-2.47 (2H, m), 2.38-2.27 (3H, m), 1.91-1.60 (9H, m). [M+H] = 606.5.
1-(butyryloxy)ethyl 7-(4-(4-(2,3-dichlorophenybpiperazin-1-y1)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (Example 25, Compound-91) CI
Sc' N

Synthesis of chloro(phenyl)methyl carbonochloridate To a solution of benzaldehyde (2.67 mL, 26.29 mmol) in diethyl ether (40 mL) at -20 C
under argon was added triphosgene (8.61 g, 28.92 mmol) followed by pyridine (90.21 mL, 2.63 mmol). The reaction was allowed to gradually warm to room temperature and stirred for 2 hours. The reaction was filtered through celite and concentrated. The residue was co-evaporated with diethylether (3 x 20 mL) to give chloro(phenyl)methyl carbonochloridate (5.44 g). This was used without further purification (contains ¨11%
benzaldehyde).
Synthesis of chloro(phenyl)methyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate To a suspension of aripiprazole (1 g, 2.23 mmol) in 2-methyltetrahydrofuran (20 mL) was added NaH (178 mg, 4.46 mmol). The reaction was heated to reflux for 1.5 hours then cooled to room temperature. Chloro(phenyl)methyl carbonochloridate (1.37 g, 6.69 mmol) was then added and the reaction stirred overnight at room temperature. The reaction was cooled to 0 C, diluted with ethyl acetate (20 mL) and quenched with aqueous saturated NaHCO3 (20 mL) solution. The reaction was extracted with ethyl acetate (3 x 20 mL) and the combined organics washed with brine (50 mL), then dried (MgSO4) and concentrated to give chloro(phenyl)methyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (crude yield 2.10 g) as a 1:3 mixture of product/aripiprazole. The product was taken onto the next step crude.
Synthesis of Compound-91 To a solution of chloro(phenyl)methyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate (4.11 g, 6.66 mmol) in dimethyl formamide (40 mL) was added butyric acid (0.91 mL, 9.99 mmol) and cesium carbonate (1.63 g, 4.99 mmol). The reaction was stirred at room temperature for 20 hours then diluted with ethyl acetate (40 mL) and washed with 1:1 water/brine (3 x 50 mL). The organics were dried (Mg504) and concentrated. The crude product was purified by column chromatography on silica eluting with 0.5% Et3N/ethyl acetate to give butyryloxy(phenyl)methyl 7-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butoxy)-2-oxo-3,4-dihydroquinoline-1(2H)-carboxylate, Compound-91 (1.12 g). The material was determined to be 67% pure by LCMS.
1H-NMR (300 MHz, CDC13) 6 7.66-7.57 (2H, m), 7.49-7.38 (3H, m), 7.18-7.13 (2H, m), 7.07-7.04 (1H, m), 6.98-6.92 (1H, m), 6.73-6.61 (2H, m), 3.96-3.83 (2H, m), 3.11-3.00 (4H, m), 2.89-2.85 (2H, m), 2.74-2.63 (6H, m), 2.57-2.51 (2H, m), 2.48-2.41 (2H, m), 1.81-1.60 (6H, m), 0.96 (3H, dt).
[M+H] ' = 668.48.
Pharmacokinetic Evaluation in Rats Pharmacokinetic Evaluation of Prodrugs in Rats Following Oral Administration (PO) Animals: Male Sprague¨Dawley rats (Charles River Laboratories, Wilmington, MA) were obtained. Approximately 24 rats were used in each study. Rats were approximately 350-375 g at time of arrival. Rats were housed 2 per cage with ad libitum chow and water.
Environmental conditions in the housing room: 64-67 F, 30% to 70% relative humidity, and 12:12-h light: dark cycle. All experiments were approved by the institutional animal care and use committee.
Pharmacokinetics study: Rats were dosed 10mg/kg (3.3mL suspension) of the test compound (see Table E). Blood samples were collected from the jugular vein after brief anesthesia with Isoflurane. A 271/2G needle and lcc syringe without an anticoagulant was used for the blood collection. Approximately 2504 of whole blood was collected at each sampling time-point of 15 and 30 minutes, and 1, 2, 4, 7 and 24 hours after administration.
Once collected, whole blood was immediately transferred prechilled tubes containing EDTA, inverted 10-15 times and immediately placed on ice. The tubes were centrifuged for 2 minutes at >14,000 g's (11500 RPMs using Eppendorf Centrifuge) at room temperature to separate plasma. Plasma samples were transferred to labeled plain tubes (MICROTAINERt) and stored frozen at < -70 C.
Data Analysis: Drug concentrations in plasma samples were analyzed by liquid chromatography¨mass spectroscopy using appropriate parameters for each compound. Half-life, volume of distribution, clearance, maximal concentration, and AUC were calculated by using WinNonlin Version 5.2 software (Pharsight, St. Louis, MO). Results are shown in The Figure.

Claims (10)

What is claimed is:
1. A compound having the formula:
wherein wherein ~ represents a single or double bond;
R1 is -C(O)OR20, -C(O)R20, or -C(O)NR2OR21;
wherein each R20 is independently C7-C24 alkyl, C7-C24 substituted alkyl, C7-C24 alkenyl, C7-C24 substituted alkenyl, C7-C24 alkynyl, or C7-C24 substituted alkynyl;
R21 is hydrogen, aliphatic, substituted aliphatic, aryl or substituted aryl;
each R3, R4 and R5 is independently absent, hydrogen, halogen, -0R10, -SR10, -NR10R11, -C(O)R10, optionally substituted aliphatic, optionally substituted aryl or optionally substituted heterocyclyl;
wherein, A is absent, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, -S-, -S(O)-, -S(O)2-, -S[C(R30)(R31)]u-, -S(O)[C(R30)(R31)b-, -S(O)2[C(R30)(R31)]u-, -0[C(R30)(R31)]u-, -N(R30)-, -N(R30)[C(R31)(R32)]u-, -[C(R30)(R31)]u, or -C(O)[C(R30)(R31)]u-;
each u is independently 1, 2, 3, 4, 5, 6 or 7;
D is absent, -O-, -NR33-, -C(R34)( R35)-, -S-, -S(O)-, -S(O)2-, or -C(O)-;
m and q are independently 0, 1, or 2;
each R30, R31, R32 R33, R34, and R35 is independently absent, hydrogen, halogen, -0R10, -SR10, -NR10R11, -C(O)R10, optionally substituted aliphatic, optionally substituted aryl or optionally substituted heterocyclyl;

each R10 and R11 is independently absent, hydrogen, halogen, aliphatic, substituted aliphatic, aryl or substituted aryl; alternatively two R10 and R11 together with the atoms to which they are attached may form an additional optionally substituted, 3, 4, 5, 6 or 7 membered ring;
R2 is absent, hydrogen, halogen, -OR10, -SR10, -NR10R11, optionally substituted aliphatic, optionally substituted aryl or aryl or optionally substituted heterocyclyl;
r is 0, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11;
each G3 and G4 is independently -N-, X20 is ¨C(R10)- or -N-;
p is 0, 1, 2 or 3; and wherein the term "substituted" refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent defined by:
halo, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, thiol, alkylthio, arylthio, alkylthioalkyl, arylthioalkyl, alkylsulfonyl, alkylsulfonylalkyl, arylsulfonylalkyl, alkoxy, aryloxy, aralkoxy, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkyl, amino, trifluoromethyl, cyano, nitro, alkylamino, arylamino, alkylaminoalkyl, arylaminoalkyl, aminoalkylamino, hydroxy, alkoxyalkyl, carboxyalkyl, alkoxycarbonylalkyl, aminocarbonylalkyl, acyl, aralkoxycarbonyl, carboxylic acid, sulfonic acid, sulfonyl, phosphonic acid, aryl, heteroaryl, heterocyclic, or aliphatic.
2. The compound of claim 1 having the formula:
wherein R1, R2, R3, R4, R5, R10, R11, A, D, m, p and q are as defined in claim 1.
3. A compound of claim 1 having the formula:
wherein, R1, R2, R3, R4, R5, A, D, m, p, q, r, R10 and R11 are as defined in claim 1 and D is -S- or -O-.
4. The compound of claim 2 having the formula:
wherein R1 is -C(O)OR20, -C(O)R20, or -C(O)NR20R21;
wherein each R20 and R21 is independently C7-C24 alkyl, C7-C24 substituted alkyl, C7-C24 alkenyl, C27-C24 substituted alkenyl, C7-C24 alkynyl, or C7-C24 substituted alkynyl;
and, w is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10 or 11.
5. A compound of claim 3 having the formula:
6. The compound of claim 1 defined in Table XI-XII:
wherein R1 is as defined in claim 1.
7. The compound of any one of claims 1-6, wherein R1 is:

8. A compound defined by Tables A, B, C, or D:
9. The use of a prodrug compound in the manufacture of a medicament for the sustained delivery of a lactam containing parent drug wherein said prodrug is the compound of any one of claims 1-8.
10. The use according to claim 9, wherein the prodrug compound has decreased solubility compared to the parent drug when measured at room temperature in a pH 7.4 phosphate buffer.
CA2802733A 2010-06-24 2011-06-24 Prodrugs of nh-acidic compounds: ester, carbonate, carbamate and phosphonate derivatives Active CA2802733C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US35834810P 2010-06-24 2010-06-24
US61/358,348 2010-06-24
PCT/US2011/041830 WO2011163594A2 (en) 2010-06-24 2011-06-24 Prodrugs of nh-acidic compounds: ester, carbonate, carbamate and phosphonate derivatives

Publications (2)

Publication Number Publication Date
CA2802733A1 CA2802733A1 (en) 2011-12-29
CA2802733C true CA2802733C (en) 2017-11-21

Family

ID=45353112

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2802733A Active CA2802733C (en) 2010-06-24 2011-06-24 Prodrugs of nh-acidic compounds: ester, carbonate, carbamate and phosphonate derivatives

Country Status (8)

Country Link
US (4) US8592427B2 (en)
EP (1) EP2585066B1 (en)
JP (2) JP5848761B2 (en)
AU (1) AU2011270701B2 (en)
CA (1) CA2802733C (en)
ES (1) ES2691671T3 (en)
NZ (1) NZ604423A (en)
WO (1) WO2011163594A2 (en)

Families Citing this family (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2445502B2 (en) * 2009-06-25 2022-09-28 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
WO2010151711A1 (en) * 2009-06-25 2010-12-29 Alkermes, Inc. Prodrugs of nh-acidic compounds
WO2011084846A1 (en) * 2010-01-07 2011-07-14 Alkermes, Inc. Quaternary ammonium salt prodrugs
JP5848761B2 (en) * 2010-06-24 2016-01-27 アルカーメス ファーマ アイルランド リミテッド Prodrugs of NH acidic compounds: derivatives of esters, carbonates, carbamates and phosphonates
EP3156056B1 (en) 2011-03-18 2023-12-06 Alkermes Pharma Ireland Limited Pharmaceutical compositions comprising sorbitan esters
JO3227B1 (en) 2011-09-08 2018-03-08 Otsuka Pharma Co Ltd Piperazine-substituted benzothiophene deriveatives as antipsychotic agents
GEP201706656B (en) 2011-11-11 2017-04-25 Pfizer 2-thiopyrimidinones
EP2790734B1 (en) 2011-12-15 2019-02-20 Alkermes Pharma Ireland Limited Prodrugs of secondary amine compounds
JP6219918B2 (en) 2012-03-19 2017-10-25 アルカームス ファーマ アイルランド リミテッド Pharmaceutical composition comprising glycerol ester
NZ630643A (en) 2012-03-19 2017-08-25 Alkermes Pharma Ireland Ltd Pharmaceutical compositions comprising fatty acid esters
WO2013142205A1 (en) 2012-03-19 2013-09-26 Alkermes Pharma Ireland Limited Pharmaceutical compositions comprising benzyl alcohol
CA2872978A1 (en) * 2012-05-08 2013-11-14 Cellixbio Private Limited Compositions and methods for suppression of carbonic anhydrase activity
MY169219A (en) * 2012-06-11 2019-03-19 Bristol Myers Squibb Co Phosphoramidic acid prodrugs of 5 - [5 - phenyl- 4 - (pyridin- 2 - ylmethylamino) quinazolin- 2 - yl] pyridine- 3 ? sulfonamide
CN102863436B (en) * 2012-06-19 2018-02-09 重庆医科大学 Tetrahydrothiazole diketone derivatives and its preparation and use
WO2014031645A1 (en) 2012-08-21 2014-02-27 Ortho-Clinical Diagnostics, Inc Antibodies to risperidone haptens and use thereof
US20140057297A1 (en) 2012-08-21 2014-02-27 Janssen Pharmaceutica Nv Antibodies to Paliperidone Haptens and Use Thereof
CN104736567B (en) * 2012-08-21 2019-09-03 詹森药业有限公司 The antibody and application thereof of Aripiprazole haptens
PL3462173T3 (en) 2012-08-21 2021-08-16 Janssen Pharmaceutica Nv Antibodies to risperidone and use thereof
CN104755631B (en) 2012-08-21 2017-09-05 詹森药业有限公司 Antibody of Aripiprazole and application thereof
EP2888234B1 (en) * 2012-08-21 2017-12-06 Janssen Pharmaceutica NV Haptens of aripiprazole and their use in immunoassays
WO2014031630A2 (en) 2012-08-21 2014-02-27 Ortho-Clinical Diagnostics, Inc Antibodies to paliperidone and use thereof
JP6654042B2 (en) 2012-09-19 2020-02-26 アルカームス ファーマ アイルランド リミテッド Pharmaceutical composition with improved storage stability
GB201308363D0 (en) 2013-05-09 2013-06-19 Bagshawe Kenneth D Tumour therapy
AU2014349103B2 (en) * 2013-11-15 2017-01-12 Emergent Biodefense Operations Lansing Llc Morphic forms of hexadecyloxypropyl-phosphonate esters
KR102428252B1 (en) * 2014-02-21 2022-08-02 상하이 미큐알엑스 파마슈티컬 컴퍼니 리미티드 Water-soluble o-carbonyl phosphoramidate prodrugs for therapeutic administration
CA2943213C (en) 2014-03-20 2022-07-05 Alkermes Pharma Ireland Limited Aripiprazole formulations having increased injection speeds
WO2015167620A1 (en) * 2014-04-29 2015-11-05 Biosearch Technologies, Inc. Compounds compositions and methods including thermally labile moieties
JP5938537B1 (en) 2014-10-24 2016-06-22 久光製薬株式会社 Prodrug
WO2016069637A1 (en) * 2014-10-28 2016-05-06 The Regents Of The University Of California Sulfonamides that activate aba receptors
WO2016130800A2 (en) * 2015-02-12 2016-08-18 Neonc Technologies Inc. Pharmaceutical compositions comprising perillyl alcohol derivatives
JP2018515480A (en) 2015-05-05 2018-06-14 ファイザー・インク 2-thiopyrimidinone
CN104892514A (en) * 2015-05-19 2015-09-09 广州诺威生物技术有限公司 Novel imidazole compound
KR20180071247A (en) 2015-07-31 2018-06-27 더 존스 홉킨스 유니버시티 Prodrugs of glutamine analogs
MA42953A (en) 2015-09-22 2018-08-01 Graybug Vision Inc COMPOUNDS AND COMPOSITIONS FOR THE TREATMENT OF EYE DISORDERS
WO2017076213A1 (en) * 2015-11-02 2017-05-11 苏州旺山旺水生物医药有限公司 Benzimidazole compound, preparation method thereof and usage
MX2018005932A (en) 2015-11-12 2019-05-20 Graybug Vision Inc Aggregating microparticles for therapy.
EP3390449A1 (en) 2015-12-17 2018-10-24 Janssen Pharmaceutica N.V. Antibodies to risperidone and use thereof
CA3008909C (en) 2015-12-23 2023-10-03 The University Of British Columbia Lipid-linked prodrugs
CN107353303B (en) * 2016-05-09 2020-09-01 上海奥博生物医药技术有限公司 Preparation method of fosaprepitant phosphate intermediate
BR112019019452A2 (en) 2017-03-23 2020-04-14 Graybug Vision Inc compound, and, use of a compound
RU2019139817A (en) 2017-05-10 2021-06-10 Грейбуг Вижн, Инк. DELAYED RELEASE MICROPARTICLES AND THEIR SUSPENSIONS FOR DRUG THERAPY
CN107217480B (en) * 2017-06-22 2018-10-02 盐城工学院 Finishing agent with antibacterial and flame retardant property and its preparation method and application method
US11485725B2 (en) 2017-12-15 2022-11-01 Auransa Inc. Derivatives of piperlongumine and uses thereof
PE20211456A1 (en) * 2018-02-28 2021-08-05 Hoffmann La Roche 7-SUBSTITUTED SULFONIMIDOYLPURINONE COMPOUNDS AND ITS DERIVATIVES FOR THE TREATMENT AND PREVENTION OF LIVER CANCER
US11273158B2 (en) 2018-03-05 2022-03-15 Alkermes Pharma Ireland Limited Aripiprazole dosing strategy
EP3762386B1 (en) * 2018-03-05 2024-01-24 Teon Therapeutics, Inc. Adenosine receptor antagonists and uses thereof
US11273137B2 (en) 2018-09-04 2022-03-15 Board Of Trustees Of Michigan State University Methods and compositions to prevent and treat disorders associated with mutations in the ODC1 gene
WO2022002040A1 (en) * 2020-06-30 2022-01-06 Suzhou Abogen Biosciences Co., Ltd. Lipid compounds and lipid nanoparticle compositions
CN111909155B (en) * 2020-08-21 2022-10-18 东莞理工学院 Proteolysis targeting chimera, prodrug molecule for improving oral bioavailability of proteolysis targeting chimera and application of prodrug molecule
CN113402547B (en) * 2021-05-27 2023-05-16 兰州大学 Synthesis method of N-acyl phosphoramide compound
CN116891465A (en) * 2022-03-30 2023-10-17 苏州旺山旺水生物医药有限公司 N-substituted quinolinone compounds, preparation method and application thereof

Family Cites Families (101)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1594847A1 (en) * 1966-08-09 1971-05-27 Bayer Ag Whitening agents
DE2306374A1 (en) * 1973-02-09 1974-08-15 Bayer Ag 2-Oxo-4-hydroxy tetrahydro quinazoline-4-carboxamides - prepd by reacting isatin-N-carboxamides with amines and useful as (inters for) dyes
US3998815A (en) 1974-06-24 1976-12-21 Interx Research Corporation 1-hydrocarbonoyloxymethyl-3-carbamoyl or 3-carboethoxy-pyridinium salts
US4204065A (en) 1975-09-22 1980-05-20 Interx Research Corporation Soft quaternary surface active agents and method of using same
US4260769A (en) 1977-04-22 1981-04-07 Interx Research Corporation 5,5-Diphenylhydantoins
EP0005828B1 (en) 1978-06-06 1981-03-11 Hoechst Aktiengesellschaft New substituted phenylpiperazine derivatives, pharmaceutical compositions containing them and process for their preparation
FI77852C (en) 1981-02-17 1989-05-10 Otsuka Pharma Co Ltd Process for the preparation of novel, such as cardiac drugs, useful s unsubstituted amide and (saturated heterocycle) carbonyl carbostyril derivatives.
AU532361B2 (en) 1981-09-01 1983-09-29 Otsuka Pharmaceutical Co., Ltd. Carbostyril derivatives
US4428935A (en) 1982-05-24 1984-01-31 Pfizer Inc. Penicillanic acid dioxide prodrug
IT1212743B (en) 1983-05-17 1989-11-30 Dompe Farmaceutici Spa QUATERNARY SALTS OF BENZO DERIVATIVES [1,4] DIAZEPINONICS, PYRID [1,4] BENZODIAZEPINONICS, PRIDO [1,5] BENZODIAZEPINONICS AND THEIR PHARMACOLOGICAL LIFE ACTS
US4760057A (en) 1983-06-23 1988-07-26 Merck & Co., Inc. (Acyloxyalkoxy)carbonyl derivatives as bioreversible prodrug moieties for primary and secondary amine functions in drugs
DK302883D0 (en) * 1983-06-30 1983-06-30 Hans Bundgaard ALLOPURINOL PRODUCTS
GB8513754D0 (en) 1985-05-31 1985-07-03 Jones T R Anti-cancer quinazoline derivatives
DK588486A (en) 1985-12-09 1987-06-10 Otsuka Pharma Co Ltd USE OF A COMPOUND TO TREAT HYPOXY
GB8607683D0 (en) 1986-03-27 1986-04-30 Ici Plc Anti-tumor agents
US4925860A (en) 1987-08-05 1990-05-15 E. I. Du Pont De Nemours And Company Stable pharmaceutical composition of 3-(hydroxymethyl)-5,5-diphenylhydantoin disodium phosphate ester
FR2640266B2 (en) * 1988-07-12 1992-07-10 Synthelabo INDOLONES-2, QUINOLEINONES-2, BENZO (B) AZEPINONES-2 AND BENZIMIDAZOLONES-2 DERIVATIVES OF (HYDROXY-1 PIPERIDINYL-2 ALKYL), THEIR PREPARATION AND THEIR THERAPEUTIC APPLICATION
US5006528A (en) 1988-10-31 1991-04-09 Otsuka Pharmaceutical Co., Ltd. Carbostyril derivatives
JP2608788B2 (en) * 1988-10-31 1997-05-14 大塚製薬 株式会社 Schizophrenia remedy
US5350747A (en) 1989-07-07 1994-09-27 Pfizer Inc Heteroaryl piperazine antipsychotic agents
DK238190D0 (en) * 1990-10-03 1990-10-03 Lundbeck & Co As H DEPOT DERIVATIVES
AU645681B2 (en) 1991-05-02 1994-01-20 John Wyeth & Brother Limited Piperazine derivatives
TW226016B (en) 1991-12-30 1994-07-01 Sterling Winthrop Inc
GB9204565D0 (en) * 1992-03-03 1992-04-15 Smithkline Beecham Plc Pharmaceuticals
US5462934A (en) 1992-03-09 1995-10-31 Takeda Chemical Industries Condensed heterocyclic ketone derivatives and their use
WO1993025197A1 (en) 1992-06-12 1993-12-23 Affymax Technologies N.V. Compositions and methods for enhanced drug delivery
AU4350493A (en) * 1992-06-27 1994-01-24 Smithkline Beecham Plc Medicaments containing 5-ht4 receptor antagonists
EP0688325A1 (en) 1993-03-08 1995-12-27 Eisai Co., Ltd. Phosphonic acid derivatives
US5497763A (en) 1993-05-21 1996-03-12 Aradigm Corporation Disposable package for intrapulmonary delivery of aerosolized formulations
DE4345266C2 (en) 1993-10-04 1996-12-19 Luitpold Pharma Gmbh Heterocyclic carbamates, process for their preparation and pharmaceuticals
US5508269A (en) 1994-10-19 1996-04-16 Pathogenesis Corporation Aminoglycoside formulation for aerosolization
IT1275903B1 (en) * 1995-03-14 1997-10-24 Boehringer Ingelheim Italia ESTERS AND AMIDES OF 1,4-PIPERIDINE DISPLACED
US6083922A (en) 1996-04-02 2000-07-04 Pathogenesis, Corp. Method and a tobramycin aerosol formulation for treatment prevention and containment of tuberculosis
US5767068A (en) 1997-02-13 1998-06-16 Pathogenesis Corporation Pure biologically active colistin, its components and a colistin formulation for treatment of pulmonary infections
KR20010020172A (en) * 1997-05-30 2001-03-15 나가사까 겐지로 2-oxoimidazole derivatives
EP0986403B1 (en) 1997-06-13 2003-11-12 Cydex Inc. Composition with extended shelf-life storage comprising cyclodextrin and drugs or prodrugs that decompose to water-insoluble components
US6180095B1 (en) 1997-12-17 2001-01-30 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
KR20010033811A (en) 1997-12-31 2001-04-25 토마스 안 빅토리아 Water soluble prodrugs of secondary and tertiary amine containing drugs and methods of making thereof
EP1114028B1 (en) 1998-08-26 2006-11-29 Aventis Pharma Limited Aza-bicycles which modulate the inhibition of cell adhesion
FR2783519B1 (en) 1998-09-23 2003-01-24 Sod Conseils Rech Applic NOVEL AMIDINE DERIVATIVES, THEIR PREPARATION, THEIR USE AS MEDICAMENTS AND THE PHARMACEUTICAL COMPOSITIONS CONTAINING THEM
EP1221443B1 (en) * 1999-10-13 2004-09-01 Banyu Pharmaceutical Co., Ltd. Substituted imidazolidinone derivatives
MY128450A (en) * 2000-05-24 2007-02-28 Upjohn Co 1-(pyrrolidin-1-ylmethyl)-3-(pyrrol-2-ylmethylidene)-2-indolinone derivatives
EP1414798A2 (en) 2001-04-03 2004-05-06 Aryx Therapeutics Ultrashort-acting opioids for transdermal application
US6919331B2 (en) * 2001-12-10 2005-07-19 Bristol-Myers Squibb Company Substituted 2-methyl-benzimidazole respiratory syncytial virus antiviral agents
TW200403223A (en) 2002-02-15 2004-03-01 Glaxo Group Ltd Novel compounds
CA2479256A1 (en) * 2002-03-20 2003-10-02 Bristol-Myers Squibb Company Phosphate prodrugs of fluorooxindoles
DE60327225D1 (en) 2002-08-20 2009-05-28 Bristol Myers Squibb Co ARIPIPRAZOL COMPLEX FORMULATION AND METHOD
PL376721A1 (en) * 2002-10-24 2006-01-09 Pfizer Products Inc. Acyl derivatives of 5-(2-(4-(1,2 benzisothiazole-3-yl)-1-piperazinyl)ethyl)-6-chloro-1,3-dihydro-2h-indol-2-one having neuroleptic activity
US20040152769A1 (en) * 2002-11-09 2004-08-05 Ekwuribe Nnochiri Nkem Modified carbamate-containing prodrugs and methods of synthesizing same
DE10303669A1 (en) 2003-01-28 2004-07-29 Hans-Knöll-Institut für Naturstoff-Forschung e.V. New quaternary ammonioalkyl carbonate esters useful for treating bacterial and fungal infections and potentially useful for regulating cholesterol biosynthesis
US7135575B2 (en) * 2003-03-03 2006-11-14 Array Biopharma, Inc. P38 inhibitors and methods of use thereof
EA012240B1 (en) 2003-04-03 2009-08-28 Семафор Фармасьютикалз, Инк. Pi-3 kinase inhibitor prodrugs
CA2525366A1 (en) * 2003-05-16 2004-11-25 Pfizer Products Inc. Therapeutic combinations of atypical antipsychotics with gaba modulators, anticonvulsants or benzodiazapines
JP4693778B2 (en) * 2003-05-23 2011-06-01 大塚製薬株式会社 Carbostyril derivatives and mood stabilizers for treating mood disorders
US20050032811A1 (en) 2003-08-06 2005-02-10 Josiah Brown Methods for administering aripiprazole
US7160888B2 (en) 2003-08-22 2007-01-09 Warner Lambert Company Llc [1,8]naphthyridin-2-ones and related compounds for the treatment of schizophrenia
EP1701954A1 (en) 2003-12-31 2006-09-20 Warner-Lambert Company LLC N-substituted piperidine and piperazine derivatives
JP2007522200A (en) 2004-02-13 2007-08-09 ファイザー・プロダクツ・インク Therapeutic combination of atypical antipsychotics and corticotropin releasing factor antagonists
US7169803B2 (en) 2004-03-15 2007-01-30 Bristol-Myers Squibb Company N-substituted prodrugs of fluorooxindoles
GB2413795A (en) 2004-05-05 2005-11-09 Cipla Ltd Process for the preparation of rosiglitazone
WO2005115993A1 (en) * 2004-05-31 2005-12-08 Banyu Pharmaceutical Co., Ltd. Quinazoline derivative
WO2006037090A2 (en) 2004-09-28 2006-04-06 Purdue Research Foundation Drug-phosphate conjugates as prodrugs
WO2006054560A1 (en) * 2004-11-17 2006-05-26 Kissei Pharmaceutical Co., Ltd. Aromatic amide derivatives, medicinal compositions containing the same, medical uses of both
WO2006090273A2 (en) 2005-02-22 2006-08-31 Warner-Lambert Company Llc [1,8]naphthyridin-2-ones and related compounds with keto or hydroxyl linkers for the treatment of schizophrenia
JP4315393B2 (en) * 2005-04-14 2009-08-19 大塚製薬株式会社 Heterocyclic compounds
TWI320783B (en) 2005-04-14 2010-02-21 Otsuka Pharma Co Ltd Heterocyclic compound
KR100781704B1 (en) * 2005-04-20 2007-12-03 에스케이케미칼주식회사 Novel pyridine derivatives, process for preparing thereof and pharmaceutical compositions containing them
US7521557B2 (en) * 2005-05-20 2009-04-21 Bristol-Myers Squibb Company Pyrrolopyridine-based inhibitors of dipeptidyl peptidase IV and methods
WO2006130522A2 (en) * 2005-05-31 2006-12-07 Orexigen Therapeutics, Inc. Methods and compositions for managing psychotic disorders
EP2279727A3 (en) 2005-09-15 2011-10-05 Elan Pharma International Limited Nanoparticulate aripiprazole formulations
EP1777222A1 (en) 2005-09-22 2007-04-25 Galantos Pharma GmbH Cholinergic enhancers with improved blood-brain barrier permeability for the treatment of diseases accompanied by cognitive impairment
US20070185069A1 (en) 2005-11-14 2007-08-09 Plum Stacy M Anti-angiogenic activity of 2-methoxyestradiol in combination with anti-cancer agents
BRPI0706623A2 (en) * 2006-01-18 2011-04-12 Schering Corp cannabinoid receptor modulators
WO2007120638A2 (en) * 2006-04-12 2007-10-25 President And Fellows Of Harvard College Methods and compositions for modulating glycosylation
CN101528712B (en) * 2006-08-26 2013-10-30 Abbvie德国有限责任两合公司 Substituted benzimidazolone derivatives, medicaments comprising them and their use
US20090068290A1 (en) 2006-08-31 2009-03-12 Michel Bourin Bifeprunox doses for treating schizophrenia
CA2663189A1 (en) * 2006-09-12 2008-03-20 Pfizer Products Inc. Benzimidazolone derivatives
US20080085888A1 (en) 2006-09-15 2008-04-10 Breining Scott R Therapeutic Combinations
CN101678120A (en) 2006-12-05 2010-03-24 纽罗吉斯克斯公司 Prodrugs and methods of making and using the same
US20080312199A1 (en) 2006-12-15 2008-12-18 Glinsky Gennadi V Treatments of therapy resistant diseases and drug combinations for treating the same
TWI325694B (en) 2006-12-15 2010-06-01 Ind Tech Res Inst All digital delay locked loop
US20080186971A1 (en) 2007-02-02 2008-08-07 Tarari, Inc. Systems and methods for processing access control lists (acls) in network switches using regular expression matching logic
US20080200454A1 (en) * 2007-02-15 2008-08-21 Ameriks Michael K Carbon-linked tetrahydro-pyrazolo-pyridine modulators of cathepsin s
US20080207659A1 (en) * 2007-02-15 2008-08-28 Asit Kumar Chakraborti Inhibitors of phosphodiesterase type 4
US20090053329A1 (en) 2007-03-19 2009-02-26 Acadia Pharmaceuticals, Inc. Combinations of 5-ht2a inverse agonists and antagonists with antipsychotics
PE20090422A1 (en) 2007-04-27 2009-04-18 Purdue Pharma Lp DERIVATIVES OF PIPERIDINE, PIPERAZINE OR TETRAHYDROPYRIDYL AS ANTAGONISTS OF TRPV1
CL2008001563A1 (en) 2007-05-30 2008-10-10 Wyeth6 3 COMPOUNDS DERIVED FROM BENZODIOXANES CONDENSED WITH HETEROCICLES; PREPARATION METHOD; PHARMACEUTICAL COMPOSITION; AND USE IN THE TREATMENT OF DISEASES SUCH AS DEPRESSION, ANXIETY, PANIC CRISIS, AMONG OTHERS.
WO2009052467A1 (en) 2007-10-19 2009-04-23 Board Of Regents Of The University Of Texas System Methods of identifying pi-3-kinase inhibitor resistance
US20090143434A1 (en) 2007-12-03 2009-06-04 Brown Arthur M Methods of using domperidone to terminate acute episodes of cardiac arrhythmia, to restore normal sinus rhythm or heart rate, to prevent recurrence of cardiac arrhythmia and to maintain normal sinus rhythm or heart rate in mammals
JP2011507910A (en) 2007-12-21 2011-03-10 ユニバーシティー オブ ロチェスター Methods for changing the lifetime of eukaryotes
WO2010004390A1 (en) * 2008-06-17 2010-01-14 Glenmark Pharmaceuticals, S.A. Quinazoline dione derivatives as trpa1 modulators
TWI453207B (en) * 2008-09-08 2014-09-21 Signal Pharm Llc Aminotriazolopyridines, compositions thereof, and methods of treatment therewith
EP2445502B2 (en) 2009-06-25 2022-09-28 Alkermes Pharma Ireland Limited Heterocyclic compounds for the treatment of neurological and psychological disorders
WO2010151711A1 (en) 2009-06-25 2010-12-29 Alkermes, Inc. Prodrugs of nh-acidic compounds
US20110166194A1 (en) 2010-01-07 2011-07-07 Alkermes, Inc. Asenapine Prodrugs
US20110166128A1 (en) 2010-01-07 2011-07-07 Alkermes, Inc. Diaryldiazepine Prodrugs for the Treatment of Neurological and Psychological Disorders
ES2768295T3 (en) 2010-01-07 2020-06-22 Alkermes Pharma Ireland Ltd Prodrugs of heteroaromatic compounds
US20110166156A1 (en) 2010-01-07 2011-07-07 Alkermes, Inc. Prodrugs for the Treatment of Schizophrenia and Bipolar Disease
WO2011084846A1 (en) 2010-01-07 2011-07-14 Alkermes, Inc. Quaternary ammonium salt prodrugs
ES2610623T3 (en) * 2010-04-19 2017-04-28 Octeta Therapeutics, Llc New synthesis of thiazolidinedione compounds
JP5848761B2 (en) * 2010-06-24 2016-01-27 アルカーメス ファーマ アイルランド リミテッド Prodrugs of NH acidic compounds: derivatives of esters, carbonates, carbamates and phosphonates

Also Published As

Publication number Publication date
US20150320875A1 (en) 2015-11-12
US9585965B2 (en) 2017-03-07
ES2691671T3 (en) 2018-11-28
CA2802733A1 (en) 2011-12-29
NZ604423A (en) 2015-01-30
JP2016000754A (en) 2016-01-07
US20140094472A1 (en) 2014-04-03
JP5848761B2 (en) 2016-01-27
JP2013541494A (en) 2013-11-14
WO2011163594A2 (en) 2011-12-29
US20110319422A1 (en) 2011-12-29
AU2011270701A1 (en) 2013-01-10
US20170196848A1 (en) 2017-07-13
EP2585066A4 (en) 2014-06-11
US8592427B2 (en) 2013-11-26
WO2011163594A3 (en) 2013-08-01
US9072788B2 (en) 2015-07-07
EP2585066B1 (en) 2018-09-26
EP2585066A2 (en) 2013-05-01
AU2011270701B2 (en) 2015-05-14

Similar Documents

Publication Publication Date Title
CA2802733C (en) Prodrugs of nh-acidic compounds: ester, carbonate, carbamate and phosphonate derivatives
US10723728B2 (en) Prodrugs of Nh-acidic compounds
CA2858787C (en) Prodrugs of tertiary amine compounds
AU2020201426B2 (en) Prodrugs of nh-acidic compounds
CA3035442C (en) Prodrugs of nh-acidic compounds
AU2014265021A1 (en) Prodrugs of nh-acidic compounds
NZ623861B2 (en) Prodrugs of secondary amine compounds

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20121213