Sei sulla pagina 1di 49

USOO9248119B2

(12) United States Patent (10) Patent No.: US 9.248,119 B2


Taneja et al. (45) Date of Patent: Feb. 2, 2016
(54) MODIFIED RELEASE DOSAGE FORMS OF WO OO,25752 5, 2000
XANTHINE OXDOREDUCTASE INHIBITOR W. WO 38: 56 A2 358:
ORXANTHINE OXIDASE INHIBITORS WO 2006056711 6, 2006
8 WO 2007/04O997 4/2007
(75) Inventors: Rajneesh Taneja, Libertyville, IL (US); WO 200703.6671 4/2007
Vijay Gupte, Vernon Hills, IL (US); WO 2008/064.015 A1 5.2008
Majid Vakilynejad, Gurnee, IL (US) WO 2009/143020 A1 11, 2009
WO 2011, 162390 12/2011
(73) Assignee: Takeda Pharmaceuticals U.S.A., Inc., WO 2012/005365 1, 2012
Deerfield, IL (US) OTHER PUBLICATIONS
(*) Notice: Subject to any disclaimer, the term of this Technical information on Eudragit, EUDRAGITOR L 100 and
patent is extended or adjusted under 35 EUDRAGITRS 100 (an Evonik product, Evonik Industries AG, pp.
U.S.C. 154(b) by 0 days. 1-7, Dec. 2012).*
(Sigma-Aldrich Sigma-Aldrich Allopurinol product information, pp.
(21) Appl. No.: 13/160,572 1-2, 2013).*
Khosravan, et al., Pharmacokinetics, pharmacodynamics and safety
(22) Filed: Jun. 15, 2011 offebuxostat, a non-purine selective inhibitor of xanthine oxidase, in
a dose escalation study in healthy subjects. TAP Pharmaceutical
(65) Prior Publication Data Products, Inc., Lake Forest, Illinois, United States, Journal: Clinical
US 2011 FO311620 A1 Dec. 22, 2011 pharmacokinetics, 2006, vol. 45(8), pp. 821-841. Abstract.
Fukunari, et al., Y-7OO 1-3-Cyano-4-(2,2-
dimethylproproxy)phenyl)-1H-pyrazole-4-carboxylic acid; a potent
Related U.S. Application Data Xanthine oxidoreductase inhibitor with hepatic excretion. J.
(60) Provisional application No. 61/355,164, filed on Jun. Pharmacol. Exp. Ther. 2004, vol. 3.11(2), pp. 519-528.
Breithaupt, et al., Kinetics of allopurinol after single intravenous and
16, 2010. oral doses. Noninteraction with benzbromarone and
hydrochlorothiazide. European Journal of Clinical Pharmacology,
(51) Int. Cl. 1982, vol. 22(1), pp. 77-84: Abstract.
A6K9/58 (2006.01) Becker, et al., “Febuxostat, a novel nonpurine selective inhibitor of
A6 IK3I/426 (2006.01) Xanthine oxidase.” Arthritis & Rheumatism, 2005, vol. 52, No. 3, pp.
A6 IK9/00 (2006.01) 916-923.
A6 IK9/20 (2006.01) Guerra, et al., “Bioecuivalence of allopurinol and its metabolite
A61 K9/24 (2006.01) oxipurinol in two tablet formulations,” Journal of Clinical Pharmacy
A61 K9/50 (2006.01) and Therapeutics, 2001, vol. 26, pp. 113-119; Abstract.
(52) U.S. Cl. Graham, et al., “Pharmacodynamics of oxypurinol after administra
tion of allopurinol to healthy subjects.” Br. J. Clin. Pharmacol., 1996,
CPC ............. A6 IK3I/426 (2013.01); A61 K9/0004 vol. 41, pp. 299-304: Abstract.
(2013.01); A61 K9/2027 (2013.01); A61 K Mayer, et al., “Pharmacokinetics and pharmacodynamics of
9/2054 (2013.01); A61 K9/5084 (2013.01); febuxostat, a new non-purine selective inhibitor of Xanthine oxidase
A61 K9/209 (2013.01); A61 K9/5026 (2013.01); in Subjects with renal impairment.” American Journal of Therapeu
A61 K9/5047 (2013.01); A61 K9/5078 tics, 2005, vol. 12, pp. 22-34: Abstract.
(2013.01) International Search Report from International Patent Application
(58) Field of Classification Search Publication No. WO2011/159745, dated Oct. 25, 2011.
None Written Opinion for Application No. PCT/US2011/040418 dated
See application file for complete search history. Oct. 25, 2011 (10 pages).
Notice of Opposition for Colombian Application No. 12-227085 by
(56) References Cited Laboratorio Franco Colombiano S.A. dated Jul. 15, 2013 (with trans
lation).
U.S. PATENT DOCUMENTS Notice of Opposition for Peruvian Application No. 2433-2012 by
Unimed del Peru S.A. dated Aug. 27, 2013 (with translation.
5,614.520 A 3, 1997 Kondo et al. Notice of Opposition for Peruvian Application No. 2433-2012 by
6,730,325 B2 5, 2004 Devane Farmindustria S.A. dated Aug. 27, 2013 (with translation).
2006/0252808 A1 1 1/2006 Joseph-Ridge (Continued)
2008/0299197 A1* 12/2008 Toneguzzo et al. ........... 424/473
2009/0022807 A1 1, 2009 Li
2009.0053308 A1 2/2009 Ishida et al. .................. 424/461
Primary Examiner — Snigdha Maewall
FOREIGN PATENT DOCUMENTS (74) Attorney, Agent, or Firm — Foley & Lardner LLP
CN 1713897 A 12/2005
CN 101474 175 T 2009 (57) ABSTRACT
CN 1016.58505 3, 2010
CN 101711743 5, 2010
CN 101711751 5, 2010 The present disclosure relates to novel dosage forms of Xan
CN 101773498 T 2010 thine oxidoreductase inhibitors.
EC SP-99-328O 1, 2001
EP O779074 6, 1997
WO OOf 23055 4/2000 30 Claims, 12 Drawing Sheets
US 9.248,119 B2
Page 2

(56) References Cited European Patent Office Action Search Report for Application No.
11796331 dated Nov. 7, 2013.
Notice of Opposition for Ecuardian Patent Application No. SP2013
OTHER PUBLICATIONS 12394 by Asociacion de Laboratorios Farmaceuticos (ALAFAR)
dated Apr. 23, 2014 (with translation).
Beara-Lasic, L. et al., “Advances in the management of gout: critical Notice of Opposition for Chilean Patent Application No. 3548-2012
appraisal offebuxostat in the control of hyperuricemia.” Int. J. Neph. by Asociacion Industrial de Laboratorios Farmaceuticos AG
Renovasc. Dis. (2010) 3: 1-10. (ASILFA) dated Aug. 12, 2014 (translation).
Becker, M.A. et al., “Febuxostat (TMX-67) a novel, non-purine, Notice of Opposition for Chilean Patent Application No. 3548-2012
Selective inhibitor of Xanthine oxidase, is safe and decreases serum by Laboratorios Recalcine S.A. dated Aug. 12, 2014 (translation).
urate in healthy volunteers.” Neucleosides, Nucleotides & Nucleic
Acids (2004) 23(8-9): 1111-1116. * cited by examiner
U.S. Patent Feb. 2, 2016 Sheet 1 of 12 US 9.248,119 B2

Fig. 1
MEAN PLASMA CONCENTRATIONS OF FEBUXOSTAT (LINEAR TOP AND
LOG-LINEAR BOTTOM) FOLLOWING ADMINISTRATION OF ASINGLE ORAL
DOSE OF 80 mg FEBUXOSTAT RELEASED IN VARIOUSLOCATIONS
THE G1 TRACT IN 12 SUBJECTS
3500

3000

2500
-

S. --- IMMEDIATE RELEASE (REF)


2000 A PROXIMAL RELEASE
O - V -- DISTAL RELEASE
is 1500 ---- COLON RELEASE
H

9O 1000
CD
500

O O O O w W

10 12 14 16 18 20 22 24 26 28 30
TIME (h)

10000

1000
-
S
2
a 100
H

H
2

2 10
O
O

8 10 12 14 16 18 20 22 24 26 28 30
TIME (h)
U.S. Patent Feb. 2, 2016 Sheet 2 of 12 US 9.248,119 B2

Fig. 2
ESTIMATED PLASMAPROFILE FORA3-PULSE 80 mg DOSE OF
FEBUXOSTAT, PULSE 1 (D.O.3, t-0) IMMEDIATE RELEASE + PULSE 2
(D.O.3, t-5h)5 HOUR DELAYED RELEASE+PULSE 3 (DO45'04, t-1Oh)
10 HOUR DELAYED RELEASE

10

n 0.1

0.01
O 5 10 15 20 25
TIME
U.S. Patent Feb. 2, 2016 Sheet 3 of 12 US 9.248,119 B2

Fig. 3
ESTIMATED PLASMAPROFILE FORA2PULSE 80 mg DOSE OF
FEBUXOSTAT, PULSE 1 (D.O.2, t=0) IMMEDIATE RELEASE +PULSE
2 (D.O.75, t-5h)5HOURRELEASE+5% DOSE RELEASE IN COLON
(D'O450.05, t- 10 h)
1000

100

0.10

O.01
TIME
U.S. Patent Feb. 2, 2016 Sheet 4 of 12 US 9.248,119 B2

Fig. 4
ESTIMATED PLASMAPROFILE FORAN 80 mg EXTENDED RELEASE (ER)
FORMULATION OF FEBUXOSTAT WHEREIN 90% OF ABSORPTION
COMPLETED IN6HOURS WITHKa=0.462h1, AND 10% OF DOSE WAS
ABSORBED FROM THE COLON WITH FRELEO45.

1.00

O 10

0.01
TIME
U.S. Patent Feb. 2, 2016 Sheet 5 of 12 US 9.248,119 B2

-61-I
G
U.S. Patent Feb. 2, 2016 Sheet 6 of 12 US 9.248,119 B2

Fig. 6
DISSOLUTION PROFILES OF FEBUXOSTAT MODIFIED RELEASE MATRIX
TABLETS (50mg) IN pH 6.8, 0.5MPHOSPHATE BUFFER

- 7 - 69622-001-A1
- A - 69622-001-B1
- A - 69622-001-C1
-A 69622-001-D1
- A 69622-001-E1
- A - 69622-001-F1
-A-69622-001-G1
--A- 69622-001-H1

TIME (hr)
U.S. Patent Feb. 2, 2016 Sheet 7 of 12 US 9.248,119 B2

Fig. 7
PLASMA CONCENTRATIONVERSUS TIME PROFILES IN DOGS FOR THE
FEBUXOSTAT FORMULATIONS DESCRIBED IN EXAMPLE 10

ULORICXR FORMULATIONDOG PLASMA CONC. (MEAN) VS TIME


6000

5000

4000 -- FORMULATION 1
H FORMULATION 2
-A- FORMULATION 3
-- FORMULATION 4
3000 ->e FORMULATION 5
2 -O- FORMULATION 6
CS -H FORMULATION 7
2, 2000 - FORMULATION 8
C

1000

TIME, HR
U.S. Patent Feb. 2, 2016 Sheet 8 of 12 US 9.248,119 B2

Figure 8A
2400
es 2200 Formulation A
2000 Y -.......... Formulation B
g 1800-
se
Formulation C
--- Formulation D
3 1600 - - FOrmulation E
1400
1200
3 1000
is 800
3S. 600
D

LL
400
200
0 - R FEas
O 10 20 30 40 50
Time (hr)

Extended-Release Formulation B
Tmax(a) Cmax AUC(0-tlqc) AUC(0-inf) T112 CLF WZF
(hr) (mg/mL) ng.hr/mL) ng.hr/mL) (h) (L/hr) (L)
N 34 34 34 27 27 27 27
Mean 3.0 1186.6 6749.3 71332 9.7 12.52 1711
SD 0.5-6.0 407.31 2198.84 2497.88 3.65 4.333 78.2
CV 34 33 35 38 35 46
U.S. Patent Feb. 2, 2016 Sheet 9 of 12 US 9.248,119 B2

Figure 8B

Formulation A
Formulation B
Formulation C
1000- H-- Formulation D
Rs. - - FOrmulation E

Time (hr)

Extended-Release Formulation B
Tmax(a) Cmax AUC(0-tiqc) AUC(0-inf) T1/2 CLF WZF
(hr) (mg/mL) ng.hr/mL) ng.hr/mL) (h) (LIhr) (L)
N 34 34 34 27 27 27 27
Mean 3.0 1186.6 6749.3 7133.2 9.7 12.52 1711
SD 0.5-6.0 407.31 2198.84 2497.88 3.65 4.333 78.2
%CW 34 33 35 38 35 46
U.S. Patent Feb. 2, 2016 Sheet 11 of 12 US 9.248,119 B2

-o-CORE #2 W, IR, TABLET #15,


11.02%CWt
-e-CORE #2 W, IR, TABLET #41
10.26%CWt
-A-CORE #2, TABLET #1, 11.02%Cwt
A CORE #2, TABLET #42, 10.63%cwt
U.S. Patent Feb. 2, 2016 Sheet 12 of 12 US 9.248,119 B2

Fig.11
6.7% AQUACOAT CWT OVER 19% CWT DISINTEGRANT
12.9% AQUACOAT CWTOVER 19% CWT DISINTEGRANT
18.5% AQUACOAT CWTOVER 19% CWT DISINTEGRANT
17.6% AQUACOAT CWTOVER 35% CWT DISINTEGRANT
25% AQUACOAT CWT OVER 35% CWT DISINTEGRANT
31% AQUACOAT CWT OVER 35% CWT DISINTEGRANT
DLON CELPHERE 507

100

8O

6O

4O
US 9.248,119 B2
1. 2
MODIFIED RELEASE DOSAGE FORMS OF Xanthine oxidoreductase, thus reducing uric acid levels. In
XANTHINE OXDOREDUCTASE INHIBITOR contrast, it has been found that the extent of protection against
ORXANTHINE OXIDASE INHIBITORS renal injury and hypertension in Subjects Suffering hyperuri
cemia is lower in Subjects treated with the uricoSuric agent
RELATED APPLICATION INFORMATION 5 benziodarone. BenZiodarone does not inhibit xanthine oxi
doreductase activity, but instead reduces plasma uric acid
This application claims priority to U.S. Provisional Appli levels by increasing the excretion of uric acid in the kidney
cation No. 61/355,164 filed on Jun. 16, 2010, the contents of (See, Mazzali M., et al., “Elevated uric acid increases blood
which are herein incorporated by reference in their entirety. pressure in the rat by a novel crystal-independent mecha
10 nism.” Hypertension, 38:1101-1106 (2001) and Mazzali M,
FIELD OF THE INVENTION et al., “Hyperuricemia induces a primary renal arteriolopathy
in rats by a blood pressure-independent mechanism. Am. J.
The present disclosure relates to novel dosage forms com Physiol Renal Physiol., 282:F991-F997 (2002)). Therefore,
prising at least one Xanthine oxidoreductase inhibitor or Xan there is a need in the art for new dosage forms that not only
thine oxidase inhibitor. In addition, the present disclosure 15 reduce uric acid levels in Subjects Suffering hyperuricemia,
also relates to methods of treating certain diseases using the but are also capable of maintaining a high level of (namely, at
novel dosage forms of the present disclosure. least 80%) inhibition of xanthine oxidoreductase activity in a
Subject in order to protect Subjects receiving these dosage
BACKGROUND OF THE INVENTION forms throughout their treatment regimen (i.e., dosing inter
Val which is typically twenty-four hours) against increasing
2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiaz concentrations of oxygen free radicals.
ole-5-carboxylic acid (also known as “febuxostat” and As referenced above, another treatment for hyperuricemia
“TMX-67) is a potent, non-purine selective inhibitor of xan is with the compound febuXostat. Extensive pharmacokinetic
thine oxidoreductase. Febuxostat 40 and 80 mg once daily and pharmacodynamic data have established that maintaining
(QD) is approved in the United States for the chronic man 25 a concentration of febuXostat in plasma over a prolonged
agement of hyperuricemia in patients with gout. Gout is a period of time provides similar efficacy to treatment with high
disease that results from the deposition of urate crystals in doses of the drug. Generally, these studies have shown that
synovial fluid and other tissues when there is an oversatura maintaining a febuXostat plasma concentration of 100 ng/ml
tion of urate in the blood. Febuxostat is a potent selective is required to provide 95% or greater inhibition of Xanthine
inhibitor of the xanthine oxidoreductase enzyme (or xanthine 30 oxidase. Currently, there the only commercially available
oxidoreductase inhibitor) that is required for the synthesis of formulations of febuxostat are immediate release formula
uric acid. tions. There are no currently commercially available
The Xanthine oxidoreductase enzyme can be present in two extended or delayed release formulations of febuxostat.
different forms (See, Enroth C, et al., “Crystal structures of Therefore, a formulation offebuxostat that maintains the drug
bovine milk Xanthine dehydrogenase A and Xanthine oxidase: 35 concentration above the critical concentration of 100 ng/ml
structure-based mechanism of conversion.” Proc. Natl. Acad. for an extended period of time is expected to result in higher
Sci. USA,97(20): 10723-8 (Sep. 26, 2000)). In one form, the efficacy of the drug, and would be a desirable treatment
Xanthine oxidoreductase enzyme is synthesized as Xanthine option for the control of hyperuricemia, gout, and many other
dehydrogenase. This form of the enzyme exhibits a very low disease states.
reactivity with oxygen. However, under stress or disease con 40
ditions, such as ischemia reperfusion injury and congestive SUMMARY OF THE INVENTION
heart failure, Xanthine dehydrogenase can undergo the for
mation of intramolecular disulfide bonds or proteolytic cleav In one embodiment, the present disclosure relates to modi
age, which converts the enzyme to the second form, Xanthine fied release dosage forms. The modified release dosage forms
oxidase. Xanthine oxidase exhibits a high reactivity with 45 can comprise at least one Xanthine oxidoreductase inhibitor
oxygen. Therefore, the synthesis of uric acid from Xanthine or at least one Xanthine oxidase inhibitor.
and hypoxanthine by the Xanthine oxidoreductase enzyme in In another embodiment, the modified release dosage forms
the form of Xanthine oxidase is associated with the generation of the present disclosure comprise: a Xanthine oxidoreductase
of oxygen free radicals, such as Superoxide anion and hydro inhibitor or a pharmaceutically acceptable salt thereof,
gen peroxide. These free radicals are capable of causing a 50 wherein said dosage form, after oral administration to a Sub
variety of toxic activities in the body such as inactivation of ject in need of treatment thereof exhibits at least one of the
proteins, DNA breakdown, lipid peroxidation (which causes following:
cell membrane disruption) and increasing pro-inflammatory (a) maintains in the Subject a plasma concentration of Xan
cytokines. thine oxidoreductase inhibitor or pharmaceutically accept
A number of disease conditions are associated with 55 able salt thereof greater than about 0.1 g/mL for a period of
elevated Xanthine oxidoreductase activity, especially, from about 5 hours to about 24 hours; and
elevated xanthine oxidase activity. Such diseases include, but (b) produces in the Subject a maximum plasma concentra
are not limited to, hyperuricemia, hypertension, metabolic tion (C) of a Xanthine oxidoreductase inhibitor or a phar
syndrome, diabetes, myocardial ischemia, atherosclerosis, maceutically acceptable salt thereof in an amount between
stroke, congestive heart failure, inflammatory bowl disease, 60 about 2.5 g/mL to about 0.5ug/mL.
renal disease progression, prostatitis, sleep apnea and autoim Alternatively, the modified release dosage form, after oral
mune diseases. Hyperuricemia is also associated with a num administration to a subject in need of treatment thereof,
ber of disease conditions, such as renal injury and hyperten exhibits the following:
Sion. (a) maintains in the Subject a plasma concentration of Xan
Allopurinol is used in the treatment of hyperuricemia. 65 thine oxidoreductase inhibitor or pharmaceutically accept
Allopurinol has been shown to prevent the renal injury and able salt thereof greater than about 0.1 g/mL for a period of
hypertension associated with hyperuricemia by inhibiting from about 5 hours to about 24 hours; and
US 9.248,119 B2
3 4
(b) produces in the Subject a maximum plasma concentra (b) produces in the Subject a maximum plasma concentra
tion (C) of a xanthine oxidoreductase inhibitor or a phar tion (C) of a Xanthine oxidoreductase inhibitor or a phar
maceutically acceptable salt thereof of between about 2.0 maceutically acceptable salt thereof of between about 2.0
ug/mL to about 1.0 ug/mL. ug/mL to about 0.075 ug/mL.
Still further alternatively, the modified release dosage 5 Still further alternatively, the modified release dosage
form, after oral administration to a Subject in need of treat form, after oral administration to a Subject in need of treat
ment thereof, can exhibit each of the following: ment thereof, can exhibit each of the following:
(a) maintains in the Subject a plasma concentration of Xan thine (a) maintains in the Subject a plasma concentration of Xan
thine oxidoreductase inhibitor or pharmaceutically accept 10 able salt oxidoreductase inhibitor or pharmaceutically accept
able salt thereof greater than about 0.1 g/mL for a period of from about thereof greater than about 0.1 g/mL for a period of
from about 5 hours to about 24 hours; and 5 hours to about 16 hours; and
(b) produces in the Subject a maximum plasma concentra tion(b)(C) produces in the Subject a maximum plasma concentra
tion (C) of a Xanthine oxidoreductase inhibitor or a phar maceuticallyofacceptable a Xanthine oxidoreductase inhibitor or a phar
salt thereof of between about 2.5
maceutically acceptable salt thereof of between about 2.5 15 ug/mL to about 0.050 g/mL.
ug/mL to about 0.5 ug/mL. The modified release dosage forms of the present disclo
In one aspect, the modified release dosage forms of the Sure can contain from about 40 to about 240 mg of at least one
present disclosure can contain from about 5 to about 240 mg Xanthine oxidoreductase inhibitor or a pharmaceutically
of at least one Xanthine oxidoreductase inhibitor or a phar acceptable salt thereof.
maceutically acceptable salt thereof. In another aspect, the When administered orally to a subject in need of treatment
the modified release dosage forms of the present disclosure thereof, the modified release dosage forms of the present
can contain from about 40 to about 240 mg of at least one disclosure can produce in the Subject a C of a Xanthine
Xanthine oxidoreductase inhibitor or a pharmaceutically oxidoreductase inhibitor or a pharmaceutically acceptable
acceptable salt thereof. salt thereof in an amount of about 2.5 Lig/mL, about 2.4
When administered orally to a subject in need of treatment 25 g/mL, about 2.3 Lig/mL, about 2.2 Lug/mL, about 2.1 Lug/mL,
thereof, the modified release dosage forms of the present 2.0 g/mL about 1.9 ug/mL, about 1.8 Lug/mL, about 1.7
disclosure can produce in the Subject a C of a Xanthine ug/mL, about 1.6 ug/mL, about 1.5 ug/mL, about 1.4 ug/mL,
oxidoreductase inhibitor or a pharmaceutically acceptable about 1.3 g/mL, about 1.2 Lug/mL, about 1.1 ug/mL, about
salt thereof in an amount of about 2.5 Lig/mL, about 2.4 1.0 g/mL, about 0.9 g/mL, about 0.8 ug/mL, about 0.7
g/mL, about 2.3 Lig/mL, about 2.2 Lug/mL, about 2.1 Lug/mL, 30 ug/mL, about 0.6 ug/mL, about 0.5ug/mL, about 0.4 ug/mL,
2.0 ug/mL about 1.9 ug/mL, about 1.8 Lug/mL, about 1.7 about 0.3 g/mL, about 0.2 Lug/mL, about 0.1 ug/mL, about
g/mL, about 1.6 ug/mL, about 1.5ug/mL, about 1.4 Lig/mL, 0.099 g/mL, about 0.098 g/mL, about 0.097 ug/mL, about
about 1.3 g/mL, about 1.2 Lug/mL, about 1.1 ug/mL, about 0.096 g/mL, about 0.095 ug/mL, about 0.094 ug/mL, about
1.0 g/mL, about 0.9 g/mL, about 0.8 ug/mL, about 0.7 0.093 ug/mL, about 0.092 lug/mL, about 0.091 ug/mL, about
ug/mL, about 0.6 ug/mL or about 0.5ug/mL. Specifically, the 35 0.090 ug/mL, about 0.089 ug/mL, about 0.088 ug/mL, about
modified release dosage forms of the present disclosure, 0.087 g/mL, about 0.086 ug/mL, about 0.085ug/mL, about
when administered orally to a subject in need of treatment 0.084 g/mL, about 0.083 lug/mL, about 0.082 ug/mL, about
thereof, can produce in that Subject a C of a Xanthine 0.081 g/mL, about 0.080 ug/mL, about 0.079 lug/mL, about
oxidoreductase inhibitor or a pharmaceutically acceptable 0.078 mg/mL, about 0.077 ug/mL, about 0.076 g/mL, about
salt thereof in a range of about 2.5ug/mL to about 1.0 ug/mL. 40 0.075 ug/mL, about 0.074 ug/mL, about 0.073 ug/mL, about
Even more specifically, the modified release dosage forms of 0.072 ug/mL, about 0.071 mg/mL, about 0.070 g/mL, about
the present disclosure, when administered orally to a subject 0.069 g/mL, about 0.068 ug/mL, about 0.067 ug/mL, about
in need of treatment thereof, can produce in that Subject a 0.066 ug/mL, about 0.065 lug/mL, about 0.064 ug/mL, about
C of a Xanthine oxidoreductase inhibitor or a pharmaceu 0.063 g/mL, about 0.062 ug/mL, about 0.061 ug/mL, about
tically acceptable salt thereof in a range of about 2.0 g/mL to 45 0.060 ug/mL, about 0.059 ug/mL, about 0.058 ug/mL, about
about 1.5 mg/mL. 0.057 g/mL, about 0.056 ug/mL, about 0.055ug/mL, about
In yet another embodiment, the modified release dosage 0.054 LG/mL, about 0.053 ug/mL, about 0.052 ug/mL, about
forms of the present disclosure comprise: a Xanthine oxi 0.051 ug/mL or about 0.050 ug/mL. Specifically, the modi
doreductase inhibitor or a pharmaceutically acceptable salt fied release dosage forms of the present disclosure, when
thereof, wherein said dosage form, after oral administration 50 administered orally to a subject in need of treatment thereof,
to a subject in need of treatment thereof exhibits at least one can produce in that Subject a C of a Xanthine oxidoreduc
of the following: tase inhibitor or a pharmaceutically acceptable salt thereof in
(a) maintains in the Subject a plasma concentration of Xan a range of about 2.5 g/mL to about 0.050 ug/mL. Even more
thine oxidoreductase inhibitor or pharmaceutically accept specifically, the modified release dosage forms of the present
able salt thereof greater than about 0.1 g/mL for a period of 55 disclosure, when administered orally to a subject in need of
from about 5 hours to about 16 hours; and treatment thereof, can produce in that subject a C of a
(b) produces in the Subject a maximum plasma concentra Xanthine oxidoreductase inhibitor or a pharmaceutically
tion (C) of a Xanthine oxidoreductase inhibitor or a phar acceptable salt thereof in a range of about 2.0 ug/mL to about
maceutically acceptable salt thereof in an amount between 0.075 ug/mL.
about 2.5 g/mL to about 0.050 ug/mL. 60 In yet another embodiment, the modified release dosage
Alternatively, the modified release dosage form, after oral forms of the present disclosure comprise: a Xanthine oxi
administration to a Subject in need of treatment thereof, doreductase inhibitor or a pharmaceutically acceptable salt
exhibits the following: thereof, wherein said dosage form, after oral administration
(a) maintains in the Subject a plasma concentration of Xan to a subject in need of treatment thereof exhibits at least one
thine oxidoreductase inhibitor or pharmaceutically accept- 65 of the following:
able salt thereof greater than about 0.1 g/mL for a period of (a) maintains in the Subject a plasma concentration of Xan
from about 5 hours to about 16 hours; and thine oxidoreductase inhibitor or pharmaceutically accept
US 9.248,119 B2
5
able salt thereof greater than about 0.1 g/mL for a period of
from about 5 hours to about 14 hours; and
(b) produces in the Subject a maximum plasma concentra
tion (C) of a xanthine oxidoreductase inhibitor or a phar 5 R R4
maceutically acceptable salt thereof in an amount between
about 2.5 g/mL to about 0.090 ug/mL.
Alternatively, the modified release dosage form, after oral wherein R and R are each independently a hydrogen, a
administration to a Subject in need of treatment thereof, hydroxyl group, a COOH group, an unsubstituted or Substi
exhibits the following: 10 tuted C-Co alkyl group, an unsubstituted or Substituted
(a) maintains in the Subject a plasma concentration of Xan C-C alkoxy, an unsubstituted or substituted hydroxy
thine oxidoreductase inhibitor or pharmaceutically accept alkoxy, a phenylsulfinyl group or a cyano ( CN) group;
able salt thereof greater than about 0.1 g/mL for a period of wherein R and Rare each independently a hydrogen or A,
from about 5 hours to about 14 hours; and B, C or Das shown below:
15
(b) produces in the Subject a maximum plasma concentra
tion (C) of a Xanthine oxidoreductase inhibitor or a phar A
maceutically acceptable salt thereof of between about 2.0 T S
ug/mL to about 0.095 ug/mL.
Still further alternatively, the modified release dosage N
Y-R,
form, after oral administration to a Subject in need of treat R6
ment thereof, can exhibit each of the following: B
(a) maintains in the Subject a plasma concentration of Xan T
NN
thine oxidoreductase inhibitor or pharmaceutically accept 25
R7
able salt thereof greater than about 0.1 g/mL for a period of NS
from about 5 hours to about 14 hours; and
(b) produces in the Subject a maximum plasma concentra Rs
C
tion (C) of a xanthine oxidoreductase inhibitor or a phar 30 H
maceutically acceptable salt thereof of between about 2.5 T
ug/mL to about 0.090 g/mL. NS
N

The modified release dosage forms of the present disclo


Sure can contain from about 40 to about 240 mg of at least one
Xanthine oxidoreductase inhibitor or a pharmaceutically
acceptable salt thereof.
When administered orally to a subject in need of treatment
35
Y-st O
D
R10
thereof, the modified release dosage forms of the present
disclosure can produce in the subject a C of a Xanthine
oxidoreductase inhibitor or a pharmaceutically acceptable
salt thereof in an amount of about 2.5 Lig/mL, about 2.4
g/mL, about 2.3 Lig/mL, about 2.2 Lug/mL, about 2.1 Lug/mL,
2.0 ug/mL about 1.9 ug/mL, about 1.8 Lug/mL, about 1.7
40
-3. N-A-N

wherein T connects A, B, C or D to the aromatic ring shown


ug/mL, about 1.6 ug/mL, about 1.5 ug/mL, about 1.4 ug/mL, 45 above at R. R. R. or R.
about 1.3 g/mL, about 1.2 Lug/mL, about 1.1 ug/mL, about wherein Rs and R are each independently a hydrogen, a
1.0 g/mL, about 0.9 g/mL, about 0.8 ug/mL, about 0.7 hydroxyl group, a COOH group, an unsubstituted or Substi
ug/mL, about 0.6 ug/mL, about 0.5ug/mL, about 0.4 ug/mL, tuted C-Co alkyl group, an unsubstituted or Substituted
about 0.3 g/mL, about 0.2 Lug/mL, about 0.1 ug/mL, about C-C alkoxy, an unsubstituted or substituted hydroxy
0.099 ug/mL, about 0.098 ug/mL, about 0.097 ug/mL, about 50 alkoxy, COO-Glucoronide or COO-Sulfate;
0.096 ug/mL, about 0.095 ug/mL, about 0.094 ug/mL, about wherein R, and Rs are each independently a hydrogen, a
0.093 ug/mL, about 0.092 lug/mL or about 0.091 lug/mL. hydroxyl group, a COOH group, an unsubstituted or Substi
Specifically, the modified release dosage forms of the present tuted C-Co alkyl group, an unsubstituted or Substituted
disclosure, when administered orally to a subject in need of C-C alkoxy, an unsubstituted or substituted hydroxy
treatment thereof, can produce in that subject a C of a 55 alkoxy, COO-Glucoronide or COO-Sulfate;
Xanthine oxidoreductase inhibitor or a pharmaceutically wherein R is an unsubstituted pyridyl group or a substi
acceptable salt thereof in a range of about 2.5 g/mL to about tuted pyridyl group; and
0.090 ug/mL. Even more specifically, the modified release wherein Rio is a hydrogen or a lower alkyl group, a lower
dosage forms of the present disclosure, when administered alkyl group Substituted with a pivaloyloxy group and in each
orally to a subject in need of treatment thereof, can produce in 60 case, Ro bonds to one of the nitrogen atoms in the 1,2,4-
that Subject a C of a Xanthine oxidoreductase inhibitor or a triazole ring shown above.
pharmaceutically acceptable salt thereof in a range of about Examples of compounds having the above formula are: (a)
2.0 ug/mL to about 0.095 ug/mL. 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
An example of a Xanthine oxidoreductase inhibitor that can 5-carboxylic acid or a pharmaceutically acceptable salt
be used in the modified release dosage forms of the present 65 thereof; (b) 2-3-cyano-4-(3-hydroxy-2-methylpropoxy)phe
disclosure are Xanthine oxidoreductase inhibitors that com nyl-4-methyl-5-thiazolecarboxylic acid or a pharmaceuti
prise the formula: cally acceptable salt thereof; (c) 2-3-cyano-4-(2-hydroxy-2-
US 9.248,119 B2
7 8
methylpropoxy)phenyl]-4-methyl-5-thiazolecarboxylic acid effective amount of above described modified release dosage
or a pharmaceutically acceptable salt thereof; (d) 2-(3-cyano form comprising at least one Xanthine oxidoreductase inhibi
4-hydroxyphenyl)-4-methyl-5-thiazolecarboxylic acid or a tor or at least one Xanthine oxidase inhibitor.
pharmaceutically acceptable salt thereof; (e) 2-4-(2-carbox In a further embodiment, the present disclosure relates to a
ypropoxy)-3-cyanophenyl-4-methyl-5-thiazolecarboxylic pharmaceutical composition that comprises a Xanthine oxi
acid or a pharmaceutically acceptable salt thereof (f) 1-3- doreductase inhibitor or a pharmaceutically acceptable salt
cyano-4-(2,2-dimethylpropoxy)phenyl)-1H-pyrazole-4-car thereof or at least one Xanthine oxidase inhibitor or a phar
boxylic acidora pharmaceutically acceptable salt thereof (g) maceutically acceptable salt thereof and at least one pharma
pyrazolo 1.5-a-1,3,5-triazin-4-(1H)-one, 8-3-methoxy-4- 10
ceutically acceptable polymer, wherein the pharmaceutical
(phenylsulfinyl)phenyl-Sodium salt (it); and (h) 3-(2-me composition includes at least one of the following: an imme
thyl-4-pyridyl)-5-cyano-4-isobutoxyphenyl)-1,2,4-triazole diate release component, a delayed release component, and/
or a pharmaceutically acceptable salt thereof. or a controlled release component. Examples of Xanthine
Another example of at least one Xanthine oxidoreductase oxidoreductase inhibitors that may be incorporated in the
inhibitor that can be used in the modified release dosage 15 pharmaceutical composition include all those previously
forms of the present disclosure are Xanthine oxidoreductase cited. An example of a Xanthine oxidase inhibitor is oxypu
inhibitors that comprise the formula: rinol or allopurinol. In addition, the immediate release com
ponent, delayed release component, and controlled release
component may comprise one or more beads capable of vari
B ous release profiles. Immediate release beads release the Xan
8 R13 thine oxidoreductase inhibitor immediately after ingestion,
R 14 the delayed release beads release the xanthine oxidoreductase
RsOCO-A-Z
l R12 inhibitor upon exposure to internal environments with speci
fied pH levels, and the controlled release beads release the
25 xanthine oxidoreductase inhibitor over an extended period of
time compared to the immediate release bead. The various
wherein R and R2 are each independently a hydrogen, a beads comprise an inert core coated with a Xanthine oxi
substituted or unsubstituted lower alkyl group, a substituted doreductase inhibitor compound and one or more layers of a
or unsubstituted phenyl, or R and R may together form a pharmaceutically acceptable polymer.
four- to eight-membered carbon ring together with the carbon 30 In an additional embodiment, the current disclosure
atom to which they are attached; encompasses a single pharmaceutical composition that incor
wherein R is a hydrogen or a substituted or unsubstituted porates both immediate release beads and delayed release
lower alkyl group; beads with solubility at pH levels greater than or equal to 6.8.
wherein R is one or two radicals selected from a group The pharmaceutical composition of this embodiment com
consisting of a hydrogen, a halogen, a nitro group, a Substi 35 prises immediate releaseXanthine oxidoreductase inhibitor or
tuted or unsubstituted lower alkyl, a substituted or unsubsti Xanthine oxidase inhibitor beads in an amount ranging from
tuted phenyl, —OR and —SONRR7, wherein R is a approximately 20% to approximately 40% (w/w) of the total
hydrogen, a Substituted or unsubstituted lower alkyl, a phe composition weight delayed release Xanthine oxidoreductase
nyl-substituted lower alkyl, a carboxymethyl orester thereof, inhibitor or Xanthine oxidase inhibitor beads that release at a
a hydroxyethyl or ether thereof, or an allyl; R, and R7 are 40 pH of 6.8 in an amount ranging from approximately 60% to
each independently a hydrogen or a Substituted or unsubsti approximately 80% (w/w) of the total composition weight.
tuted lower alkyl: For example, in one aspect, the pharmaceutical composition
wherein Rs is a hydrogen or a pharmaceutically active comprises comprises immediate release Xanthine oxi
ester-forming group; doreductase inhibitor or Xanthine oxidase inhibitor beads in
wherein A is a straight or branched hydrocarbon radical 45 an amount of approximately 20% (w/w) of the total compo
having one to five carbon atoms; sition weight delayed release Xanthine oxidoreductase inhibi
wherein B is a halogen, an oxygen, or a ethylenedithio; tor or xanthine oxidase inhibitor beads that release at a pH of
wherein Y is an oxygen, a Sulfur, a nitrogen or a substituted 6.8 in an amount of approximately 80% (w/w) of the total
nitrogen; composition weight. In yet another aspect, the pharmaceuti
wherein Z is an oxygen, a nitrogen or a Substituted nitro 50 cal composition comprises immediate release Xanthine oxi
gen; and doreductase inhibitor or Xanthine oxidase inhibitor beads in
the dotted line refers to either a single bond, a double bond, an amount of approximately 25% (w/w) of the total compo
or two single bonds. sition weight delayed release Xanthine oxidoreductase inhibi
In another embodiment, the present disclosure relates to a tor or xanthine oxidase inhibitor beads that release at a pH of
method of treating a patient Suffering from gout, hyperurice 55 6.8 in an amount of approximately 75% (w/w) of the total
mia, prostatitis, inflammatory bowel disease, QT interval pro composition weight. In still yet another aspect, the pharma
longation, myocardial infarction, cardiac hypertrophy, hyper ceutical composition comprises immediate release Xanthine
tension, nephrolithiasis, renal impairment, chronic kidney oxidoreductase inhibitor or Xanthine oxidase inhibitor beads
disease, metabolic syndrome, diabetes, diabetic nephropathy in an amount of approximately 30% (w/w) of the total com
or congestive heart failure and in need of treatment thereof. 60 position weight delayed release Xanthine oxidoreductase
The method comprises the step of administering to a subject inhibitor or Xanthine oxidase inhibitor beads that release at a
Suffering from gout, hyperuricemia, prostatitis, inflammatory pH of 6.8 in an amount of approximately 70% (w/w) of the
bowel disease, QT interval prolongation, myocardial infarc total composition weight. In still yet another aspect, the phar
tion, cardiac hypertrophy, hypertension, nephrolithiasis, maceutical composition comprises immediate release Xan
renal impairment, chronic kidney disease, metabolic Syn 65 thine oxidoreductase inhibitor or Xanthine oxidase inhibitor
drome, diabetes, diabetic nephropathy or congestive heart beads in an amount of approximately 40% (w/w) of the total
failure and in need of treatment thereof, a therapeutically composition weight delayed release Xanthine oxidoreductase
US 9.248,119 B2
10
inhibitor or Xanthine oxidase inhibitor beads that release at a Pharmacodynamics and Safety of Febuxostat, a Non-Purine
pH of 6.8 in an amount of approximately 60% (w/w) of the Selective Inhibitor of Xanthine Oxidase, in a Dose Escalation
total composition weight. Study in Healthy Subjects, CLINICAL PHARMACOKINETICS, 2006:
In yet another embodiment of the current disclosure, the 45 (8): 821-841. Specifically, the pharmacokinetic param
pharmaceutical dosage form encompasses a single pharma eters of the study are discussed on page 829 of the article, and
ceutical composition that incorporates immediate release are shown in Table 1 of Example 1, included herein.
beads, delayed release beads with solubility at pH levels In a Phase 1, multiple-dose, randomized, placebo-con
greater than or equal to 6.0, and delayed release beads with trolled, double-blind, single-center, multiple-location dose
solubility at pH levels greater than or equal to 6.8. The phar escalation study involving febuXostat, the pharmacokinetics
maceutical composition of this embodiment comprises 10 and pharmacodynamics offebuXostat was studied in healthy
immediate releaseXanthine oxidoreductase inhibitor beads in Subjects. In this study, oral doses of an immediate release
an amount ranging from approximately 25% to approxi dosage form offebuXostat (a Xanthine oxidoreductase inhibi
mately 35% (w/w) of the total composition weight, pH 6.0 tor) ranged from 10 mg once a day to 240 mg once a day
delayed release xanthine oxidoreductase inhibitor orxanthine (hereinafter "QD”) and as 30 mg twice a day (hereinafter
oxidase inhibitor beads in an amount ranging from approxi 15 “BID). In this study, it was determined that a dose of 30 mg
mately 25% to approximately 35% (w/w) of the total compo febuxostat administered twice daily (for a total dose of 60 mg
sition weight, and pH 6.8 delayed release xanthine oxi daily) was as effective in lowering uric acid levels as a once
doreductase inhibitor or Xanthine oxidase inhibitor beads in daily 120 mg febuxostat dose. Given these findings, it was
an amount ranging from approximately 35% to approxi determined that maintenance of drug levels above a minimum
mately 45% (w/w) of the total composition weight. concentration was critical to improved uric acid lowering.
In a further embodiment of the current disclosure, the Through further research and gathering of pharmacokinetic
pharmaceutical composition encompasses a single pharma data, it was determined that maintenance of in vivo febuXostat
ceutical composition that incorporates immediate release concentrations at or above 100 ng/mL (0.1 ug/mL) resulted in
beads and delayed-controlled release beads, with the delayed 80% or greater inhibition of uric acid levels. In view of this
release beads having solubility at a pH level of at least 6.8 and 25 Surprising determination, the inventors developed an
a controlled release rate of approximately four to six hours. extended release febuxostat formulation effective in maxi
The pharmaceutical composition of this embodiment com mizing time spent above the minimum critical febuXostat
prises immediate releaseXanthine oxidoreductase inhibitor or concentration of 100 ng/mL (0.1 g/mL).
Xanthine oxidase inhibitor beads in an amount ranging from The pharmacokinetic data obtained in the clinical trial
approximately 20% to approximately 40% (w/w) of the total 30 referenced above was subsequently used to develop estimated
composition weight and delayed-controlled release Xanthine plasma profiles for various febuXostat formulations, includ
oxidoreductase inhibitor or Xanthine oxidase inhibitor beads ing extended release matrix tablets, two-pulse febuxostat for
having solubility at pH levels greater than or equal to 6.8 and mulations, and three-pulse febuXostat formulations. The esti
providing prolonged release of Xanthine oxidoreductase mated extended release formulation data was based on a
inhibitor or xanthine oxidase inhibitor over a period of about 35 matrix formulation incorporating one or more polymers, and
4 hours to about 6 hours, in an amount ranging from approxi the estimated two-pulse and three-pulse formulation data was
mately 60% to approximately 80% (w/w) of the total compo based on a formulation incorporating two or more types of
sition weight. beads with differing release profiles. This information and
In still another embodiment of the current disclosure, the methodology is discussed in Example 2. Additionally, as part
pharmaceutical composition encompasses a single pharma 40 of the development process for an extended release febuxostat
ceutical composition that incorporates immediate release formulation, various sites of absorption were investigated to
beads and controlled release beads capable of active release determine the optimal physiologic sites of drug absorption for
over approximately ten to approximately twelve hours. The an extended release febuxostat formulation. In early pre
pharmaceutical composition of this embodiment generally clinical studies, absorption of febuxostat from various
comprises immediate releaseXanthine oxidoreductase inhibi 45 regions of the gastro-intestinal tract was studied in rats. The
tor or Xanthine oxidase inhibitor beads in an amount ranging testing in rat models showed that absorption offebuxostat
from approximately 10% to approximately 30% (w/w) of the was very poor from the colonic region. To guide development
total composition weight and controlled release Xanthine oxi of a dosage form that provided the desired plasma concentra
doreductase inhibitor orxanthine oxidase inhibitor beads pro tion-time profile, a site of absorption study was conducted in
viding prolonged release of Xanthine oxidoreductase inhibi 50 humans. This data and the methodology for gathering the
tor or xanthine oxidase inhibitor over a period of about 10 information are included in Example 3. The site of absorption
hours to about 12 hours, in an amount ranging from approxi data surprisingly showed that absorption offebuxostat in the
mately 70% to approximately 90% (w/w) of the total compo colon was only approximately 40% compared to the absorp
sition weight. tion profiles witnessed with immediate release, proximal
The development of the previously described embodi 55 intestine, and distal intestine formulations, higher than what
ments resulted from a long drug development process. Ini would be expected from the rat data.
tially, a dose-escalation, placebo-controlled, double blind In view of this Surprising test data, the inventors began
study was performed on twelve healthy Subjects, designed to development of an extended release febuxostat formulation
assess the safety and maximum tolerated dose offebuXostat that minimized febuXostat exposure in the colon, and maxi
by oral administration. The study was also designed to assess 60 mized febuXostat exposure in other areas, including the stom
the pharmacokinetic and pharmacodynamic profiles of mul ach, proximal intestine, and distal intestine. Novel febuxostat
tiple daily oral doses over a range of doses and ranges, includ formulations were developed by producing febuxostatformu
ing both once-daily and twice-daily administration. The lations with immediate release components, delayed release
results of this study established valuable pharmacokinetic components based on pH levels, and continuous release com
and pharmacodynamic information pertaining to febuXostat 65 ponents based on release profile over an extended time period.
bioavailability in vivo. The results of this study were pub Specific formulations are described in Examples 4-9. The
lished in the article: Reza Khosravanet al., Pharmcokinetics, novel febuxostat formulations were then tested in a dog
US 9.248,119 B2
11 12
model, as described in Example 10. The results of testing in Formulation A (reference): Febuxostat (Uloric) IR 80 mg
the dog model were expected given the well-recognized limi tablet.
tations of pharmacokinetic testing in dog models. Despite the Formulation B (test): Two-pulsatile prototype (80 mg)
limitations regarding the length of the dog gastrointestinal febuxostat capsule (TMX-67 XRFormulation B).
tract, the delayed release (i.e., pH dependent) formulations Formulation C (test): Three-pulsatile prototype (80 mg)
exhibited improved pharmacokinetic parameters compared febuxostat capsule (TMX-67 XRFormulation C).
to the reference immediate release febuxostat formulation. Formulation D (test): Combination of pulsatile and con
These formulations were then tested in humans in a single tinuous release (80 mg) febuxostat capsule (TMX-67
dose study as described in Example 11. Formulation D).
The specific parameters and scope of the extended release 10
Formulation E (test): Continuous release (80 mg) proto
febuxostat formulations are described in greater detail in the type febuxostat capsule (TMX-67 XR Formulation E).
Detailed Description. FIG. 9 shows how the dissolution profile of the formula
BRIEF DESCRIPTION OF THE FIGURES tions described in Example 12 can be varied pending on the
15 ratio of cellulose acetate to polyethylene glycol (PEG).
FIG. 1 shows the mean febuXostat plasma concentration— FIG. 10 shows how a formulation described in Example 12
time profile for multiple 80 mg febuxostat formulations can be overcoated with an immediate release layer of the drug
designed to release febuxostat in different portions of the (febuXostat) to overcome a time-lag.
gastrointestinal tract. Specifically, FIG. 1 illustrates the aver FIG. 11 shows how multiparticulate formulations
age plasma febuXostat concentration over time for a dosage described in Example 12 can be prepared to have desired
form designed to release febuXostat immediately in the stom release characteristics by varying the amount of ethylcellu
ach, in the proximal Small intestine, in the distal Small intes lose coating contained on said formulations.
tine, and in the colon.
FIG. 2 shows the simulated febuxostat plasma concentra DETAILED DESCRIPTION OF THE
tion-time profile for a dosage form comprising an 80 mg 25 DISCLOSURE
3-pulse febuxostat formulation, in which 30% of the febux
ostat dose is released immediately (at time–0 hours) (i.e., I. Definitions
pulse 1), 30% of the febuxostat dose is released after 5 hours
(i.e., pulse 2), and 40% of the febuxostat formulation is Section headings as used in this section and the entire
released after 10 hours (i.e., pulse 3). The simulated data was 30 disclosure herein are not intended to be limiting.
calculated using parameters obtained from the site of absorp As used herein, the singular forms “a,” “an and “the
tion data referenced and discussed in Example 3. include plural referents unless the context clearly dictates
FIG. 3 shows the simulated febuxostat plasma concentra otherwise. For the recitation of numeric ranges herein, each
tion-time profile for a dosage form comprising an 80 mg intervening number there between with the same degree of
2-pulse febuxostat formulation, in which 20% of the febux 35 precision is explicitly contemplated. For example, for the
ostat dose is released immediately (at time–0 hours) (i.e., range 6-9, the numbers 7 and 8 are contemplated in addition
pulse 1), 75% of the febuxostat dose is released after 5 hours, to 6 and 9, and for the range 6.0–7.0, the numbers 6.0, 6.1, 6.2,
and 5% of the febuxostat formulation is released in the colon 6.3, 6.4., 6.5, 6.6, 6.7, 6.8, 6.9 and 7.0 are explicitly contem
after 10 hours (the 5 hour and 10 hour releases collectively plated.
comprising pulse 2). The simulated data was calculated using 40 As used herein, the term “about is used synonymously
parameters obtained from the site of absorption data refer with the term “approximately.” Illustratively, the use of the
enced and discussed in Example 3. term “about indicates that values slightly outside the cited
FIG. 4 shows the simulated febuxostat plasma concentra values, namely, plus or minus 10%. Such dosages are thus
tion-time profile for a dosage form comprising an 80 mg encompassed by the scope of the claims reciting the terms
extended release (ER) febuxostat formulation, in which 90% 45 “about' and “approximately.”
of the febuxostat dose is absorbed within 6 hours after dosing As used herein, the term “AUC” refers to the area under the
and the remaining 10% of the febuxostat dose is absorbed by plasma concentration time curve of the active agent and
the colon. The simulated data was calculated using param which is calculated using the trapezoidal rule. The term
eters obtained from the site of absorption data referenced “AUC” means the area under the plasma concentration time
discussed in Example 3. 50 curve from time 0 to 120 hours after administration in units of
FIG. 5 shows a table listing the compositions of eight ngth/mL as determined using the trapezoidal rule. The term
febuxostat modified release matrix tablet formulations. “AUCoo’ means the area under the plasma concentration time
FIG. 6 illustrates the dissolution profiles for the eight dis curve from time 0 to infinite time. AUCOO is calculated as
tinct febuxostat modified release matrix tablet formulations AUC+LMT/(-3), where “LMT is the last measurable
over time. Specifically, the dissolution profiles were obtained 55 plasma concentration and B is the terminal phase elimination
by dissolving 50 mg modified release matrix tablet formula rate constant. Unless otherwise noted herein, the reported
tions a solution with a pH of 6.8 and in the presence of a 0.5 value for the AUC is the central value of the AUC. The
M phosphate buffer. “central value” of the AUC is the mean AUC-tstandard devia
FIG. 7 illustrates the plasma febuxostat concentration-time tion.
profile for multiple modified release dosage forms, as 60 The terms “administer”, “administering”, “administered
described in Example 10, as tested in a dog model. or “administration” refer to any manner of providing a drug
FIGS. 8A and 8B show the mean febuxostat plasma con (such as, a Xanthine oxidoreductase inhibitor or a pharmaceu
centration-time profiles (Linear and cLogarithmic) following tically acceptable salt thereof) to a subject or patient. Routes
administration of a single 80 mg oral dose of 4 extended of administration can be accomplished through any means
release and IR febuxostat formulations as described in 65 known by those skilled in the art. Such means include, but are
Example 11. In FIGS. 8A and 8B the formulations are as not limited to, oral, buccal, intravenous, Subcutaneous, intra
follows: muscular, transdermal, by inhalation and the like.
US 9.248,119 B2
13 14
The term “active agent as used herein refers to (1) a As used herein, the term “extended release' refers to a drug
Xanthine oxidoreductase inhibitor or a pharmaceutically formulation that provides for the gradual release of a drug
acceptable salt thereof or (2) a xanthine oxidase inhibitor or a over an extended period of time. The term “controlled
pharmaceutically acceptable salt thereof. The term “active release refers to a type of extended release formulation where
agent” and “drug are used interchangeably herein. The solid 5 the gradual release of the drug is controlled or manipulated
state form of the active agent used in preparing the dosage over a certain extended period of time.
forms of the present disclosure is not critical. For example, The term “immediate release' is used in its conventional
active agent used in preparing the modified release dosage
forms of the present disclosure can be amorphous or crystal sense to refer to a dosage form that provides for release of the
line. The final dosage form contains at least a detectable 10 active agent immediately after drug administration.
amount of crystalline active agent. The crystalline nature of As used herein, the term “modified’ refers to a drug con
the active agent can be detected using powder X-ray diffrac taining formulation in which release of the drug is not imme
tion analysis, by differential scanning calorimetry or any diate (See, for example, Guidance for Industry SUPAC-MR:
other techniques known in the art. Modified Release Solid Oral Dosage Forms, Scale-Up and
The term “C.” refers to the maximum observed plasma 15 Postapproval Changes. Chemistry, Manufacturing, and Con
concentration of a Xanthine oxidoreductase inhibitor or salt trols. In Vitro Dissolution, Testing and In Vivo Bioequivalence
thereof produced by the ingestion of the dosage forms of the Documentation, U.S. Department of Health and Human Ser
present disclosure. Unless otherwise noted herein, the vices, Food and Drug Administration, Center for Drug Evalu
reported value for the C is the central value of the C. ation and Research (“CDER), September 1997 CMC8, page
The "central value of the C is the mean Cistandard 34, herein incorporated by reference). In a modified formu
deviation.
As used herein, the term “delayed release' refers to a type lation, modified release dosage form or modified dosage
of modified release wherein a drug dosage form exhibits a form, administration of said formulation or dosage form does
time delay between oral administration of the drug dosage not result in immediate release of the drug or active agent into
form and the release of the drug from said dosage form. 25 an absorption pool. The term is used interchangeably with
Pulsed release systems (also known as “pulsatile drug “nonimmediate release' as defined in Remington. The Sci
release') and the use of enteric coatings, which are well ence and Practice of Pharmacy, Nineteenth Ed. (Easton, Pa...:
known to those skilled in the art, are examples of delayed Mack Publishing Company, 1995). As used herein, the term
release mechanisms. Generally, delayed release dosage forms "modified release' includes extended or controlled release,
release little or no active compound for a predetermined time 30 delayed release and delayed-controlled release formulations.
or until a predetermined condition is met, Such as exposure to By “pharmaceutically acceptable.” such as in the recitation
a certain pH level, then release of the active compound occurs of a “pharmaceutically acceptable excipient, or a “pharma
immediately thereafter. ceutically acceptable additive is meant a material that is not
As used herein, the term “delayed-controlled release' biologically or otherwise undesirable, i.e., the material may
refers to a type of modified release wherein a drug dosage 35
be incorporated into a pharmaceutical composition adminis
form exhibits a prolonged release of the drug over a set time tered to a patient without causing any undesirable biological
period, with release not initiated until after a certain time effects.
delay post ingestion of the dosage form. Generally, a
“delayed-controlled release' dosage form releases little or no The term “subject” refers to an animal, preferably a mam
active compound, for a predetermined time or until a prede 40 mal, including a human or non-human. The terms patient and
termined condition is met, such as exposure to a certain pH subject may be used interchangeably herein. The terms “treat
range, then release of the active compound occurs over an ing and “treatment” refer to reduction in severity and/or
additional prolonged period of time. frequency of symptoms, elimination of symptoms and/or
The term “dosage form” refers to any solid object, semi underlying cause, prevention of the occurrence of symptoms
Solid, or liquid composition designed to contain a specific 45 and/or their underlying cause, and improvement or remedia
pre-determined amount (i.e., dose) of a certain active agent. tion of damage. Thus, for example, “treating a patient
Suitable dosage forms may be pharmaceutical drug delivery involves prevention of a particular disorder or adverse physi
systems, including those for oral administration, buccal ological event in a Susceptible individual as well as treatment
administration, rectal administration, topical or mucosal of a clinically symptomatic individual by inhibiting or caus
delivery or Subcutaneous implants, or other implanted drug 50
ing regression of a disorder or disease.
delivery systems and the like. Preferably, the dosage forms of
the present disclosure are considered to be solid, however, As used herein, the term "Xanthine oxidoreductase' refers
they may contain liquid or semi-solid components. More to at least one form of Xanthine oxidoreductase enzyme,
preferably, the dosage form is an orally administered system namely Xanthine oxidase and/or Xanthine dehydrogenase.
for delivering an active agent to the gastrointestinal tract of a 55 As used herein, the phrase "Xanthine oxidoreductase
subject. The dosage form of the present disclosure exhibit inhibitor” refers to any compound that (1) is an inhibitor of a
modified release of the active agent. Xanthine oxidoreductase, Such as, but not limited to, Xanthine
By an “effective amount’ or a “therapeutically effective oxidase; and (2) chemically, does not contain a purine ring in
amount of an active agent is meant a nontoxic but sufficient its structure (i.e. is a “non-purine'). The phrase “xanthine
amount of the active agent to provide the desired effect. The 60 oxidoreductase inhibitor as defined herein also includes
amount of active agent that is “effective' will vary from metabolites, polymorphs, Solvates and prodrugs of the Such
Subject to Subject, depending on the age and general condition compounds, including metabolites, polymorphs, Solvates and
of the individual, the particular active agent or agents, and the prodrugs of the compounds described in Formula I and For
like. Thus, it is not always possible to specify an exact “effec mula II below. Examples of xanthine oxidoreductase inhibi
tive amount.” However, an appropriate “effective amount” in 65 tors include, but are not limited to, 2-4-(2-carboxypropoxy)-
any individual case may be determined by one of ordinary 3-cyanophenyl]-4-methyl-5-thiazolecarboxylic acid and
skill in the art using routine experimentation. compounds having the following Formula I or Formula II:
US 9.248,119 B2
15 16
Compounds of Formula I: Compounds of Formula II:

R R4 RsOCO-A-Z S
10
wherein R and R are each independently a hydrogen, a
hydroxyl group, a COOH group, an unsubstituted or Substi wherein R and R2 are each independently a hydrogen, a
tuted C-Co alkyl group, an unsubstituted or Substituted substituted or unsubstituted lower alkyl group, a substituted
C-C alkoxy, an unsubstituted or substituted hydroxy or unsubstituted phenyl (the substituted phenyl in this For
alkoxy, a phenylsulfinyl group or a cyano ( CN) group; 15
mula II refers to a phenyl substituted with a halogen or lower
wherein RandR are each independently a hydrogen or A, B, alkyl, and the like. Examples include, but are not limited to,
C or Das shown below: p-tolyl and p-chlorophenyl), or R and R may together
form a four- to eight-membered carbon ring together with the
carbon atom to which they are attached;
A
wherein R is a hydrogen or a substituted or unsubstituted
lower alkyl group;
T

N
S
y-R, wherein R is one or two radicals selected from a group
consisting of a hydrogen, a halogen, a nitro group, a Substi
tuted or unsubstituted lower alkyl group, a substituted or
R6 25 unsubstituted phenyl (the substituted phenyl in this Formula
B II refers to a phenyl substituted with a halogen or lower alkyl
T group, and the like. Examples include, but are not limited to,
NN p-tolyl and p-chlorophenyl), —OR and —SONR,R7.
R7 wherein R is a hydrogen, a Substituted or unsubstituted
NS
30 lower alkyl, a phenyl-substituted lower alkyl, a carboxym
R8 ethyl or ester thereof, a hydroxyethyl or ether thereof, or an
C
allyl; R, and R7 are each independently a hydrogen or a
T
H substituted or unsubstituted lower alkyl group;
wherein Rs is a hydrogen or a pharmaceutically active
N 35 ester-forming group;
s h wherein A is a straight or branched hydrocarbon radical
having one to five carbon atoms;
\--- O
D
40
wherein B is a halogen, an oxygen, or an ethylenedithio;
wherein Y is an oxygen, a Sulfur, a nitrogen or a substituted
nitrogen;
wherein Z is an oxygen, a nitrogen or a Substituted nitro
R10 gen; and
C)
-Q's, 45
the dotted line refers to either a single bond, a double bond,
or two single bonds (for example, when B is ethylenedithio.
the dotted line shown in the ring structure can be two single
bonds).
As used herein, the term “lower alkyl(s)' group refers to a
wherein T connects or attaches A, B, C or D to the aromatic C-C, alkyl group, including, but not limited to, including
ring shown above at R. R. R. or R. methyl, ethyl, n-propyl, isopropyl, butyl, isobutyl, sec-butyl,
wherein Rs and R are each independently a hydrogen, a 50 tert-butyl, pentyl, isopentyl, hexyl, heptal and the like.
hydroxyl group, a COOH group, an unsubstituted or Substi As used herein, the term “lower alkoxy' refers to those
tuted C-Co alkyl group, an unsubstituted or Substituted groups formed by the bonding of a lower alkyl group to an
C-C alkoxy, an unsubstituted or substituted hydroxy oxygen atom, including, but not limited to, methoxy, ethoxy,
propoxy, isopropoxy, butoxy, isobutoxy, pentoxy, hexoxy,
alkoxy, COO-Glucoronide or COO-Sulfate; 55 heptoxy and the like.
wherein R, and Rs are each independently a hydrogen, a As used herein, the term “lower alkylthio group' refers to
hydroxyl group, a COOH group, an unsubstituted or Substi those groups formed by the bonding of a lower alkyl to a
tuted C-Co alkyl group, an unsubstituted or Substituted Sulfur atom.
C-C alkoxy, an unsubstituted or substituted hydroxy As used herein, the term “halogen refers to fluorine, chlo
alkoxy, COO-Glucoronide or COO-Sulfate; 60 rine, bromine and iodine.
wherein Ro is an unsubstituted pyridyl group or a Substi As used herein, the term “substituted pyridyl refers to a
tuted pyridyl group; and pyridyl group that can be substituted with a halogen, a cyano
group, a lower alkyl, a lower alkoxy or a lower alkylthio
wherein Rio is a hydrogen or a lower alkyl group, a lower group.
alkyl group Substituted with a pivaloyloxy group and in each 65 As used herein, the term “four- to eight-membered carbon
case, Ro bonds to one of the nitrogen atoms in the 1,2,4- ring refers to cyclobutyl, cyclopentyl, cyclohexyl, cyclohep
triazole ring shown above in Formula I. tyl, cyclooctyl and the like.
US 9.248,119 B2
17 18
As used herein, the phrase “pharmaceutically active ester base esterification catalyst (e.g., NaOH, HSO). Ethyl ester
forming group' refers to a group which binds to a carboxyl prodrugs are prepared in similar fashion using ethanol in
group through an ester bond. Such ester-forming groups can place of methanol.
be selected from carboxy-protecting groups commonly used Examples of compounds having the above Formula I are:
for the preparation of pharmaceutically active Substances, 5 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
especially prodrugs. For the purpose of the present disclo 5-carboxylic acid (also known as “febuxostat”), 2-3-cyano
Sure, said group should be selected from those capable of 4-(3-hydroxy-2-methylpropoxy)phenyl]-4-methyl-5-thiaz
binding to compounds having Formula II wherein Rs is olecarboxylic acid, 2-3-cyano-4-(2-hydroxy-2-
hydrogen through an ester bond. Resultantesters are effective methylpropoxy)phenyl]-4-methyl-5-thiazolecarboxylic
to increase the stability, Solubility, and absorption in gas 10 acid, 2-(3-cyano-4-hydroxyphenyl)-4-methyl-5-thiazolecar
trointestinal tract of the corresponding non-esterified forms boxylic acid, 2-4-(2-carboxypropoxy)-3-cyanophenyl-4-
methyl-5-thiazolecarboxylic acid, 1-(3-cyano-4-(2,2-dim
of said compounds having Formula II, and also prolong the ethylpropoxy)phenyl)-1H-pyrazole-4-carboxylic acid, 1-3-
effective blood-level of it. Additionally, the ester bond can be cyano-4-(2,2-dimethylpropoxy)phenyl)-1H-pyrazole-4-
cleaved easily at the pH of body fluid or by enzymatic actions 15 carboxylic acid, pyrazolo 1.5-a-1,3,5-triazin-4-(1H)-one,
in vivo to provide a biologically active form of the compound 8-3-methoxy-4-(phenylsulfinyl)phenyl-sodium salt (+) or
having Formula II. Preferred pharmaceutically active ester 3-(2-methyl-4-pyridyl)-5-cyano-4-isobutoxyphenyl)-1,2,4-
forming groups include, but are not limited to, 1-(oxygen triazole.
Substituted)-C to Cs alkyl groups, for example, a straight, Preferred compounds having the above Formula I are:
branched, ringed, or partially ringed alkanoyloxyalkyl 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
groups, such as acetoxymethyl, acetoxyethyl, propiony 5-carboxylic acid, 2-3-cyano-4-(3-hydroxy-2-methylpro
loxymethyl, pivaloyloxymethyl, pivaloyloxyethyl, cyclo poxy)phenyl-4-methyl-5-thiazolecarboxylic acid, 2-3-cy
hexaneacetoxyethyl, cyclohexanecarbonyloxycyclo ano-4-(2-hydroxy-2-methylpropoxy)phenyl-4-methyl-5-
hexylmethyl, and the like, C to Cs alkoxycarbonyloxyalkyl thiazolecarboxylic acid, 2-(3-cyano-4-hydroxyphenyl)-4-
groups, such as ethoxycarbonyloxyethyl, isopropoxycarbo 25 methyl-5-thiazolecarboxylic acid, 2-4-(2-carboxypropoxy)-
nyloxyethyl, isopropoxycarbonyloxypropyl, t-butoxycarbo 3-cyanophenyl]-4-methyl-5-thiazolecarboxylic acid. These
nyloxyethyl, isopentyloxycarbonyloxypropyl, cyclohexy preferred compounds have also been found not have an effect
loxycarbonyloxyethyl, cyclohexyl at a therapeutically effective amount in a subject on the activ
methoxycarbonyloxyethyl, bornyloxycarbonyloxyisopropyl. ity of any of the following enzymes involved in purine and
and the like, C. to Calkoxyalkyls, such as methoxy methyl, 30 pyrimidine metabolism: guanine deaminase, hypoxanthine
methoxyethyl, and the like, C to Cs 2-oxacycloalkyls such guanine phosphoribosyltransferse, purine nucleotide phos
as, tetrahydropyranyl, tetrahydrofuranyl, and the like, substi phorylase, orotate phosphoribosyltransferase or orotidine-5-
tuted Cs to Caralkyls, for example, phenacyl, phthalidyl, monophosphate decarboxylase (i.e., meaning that it is
and the like, C to Caryl, for example, phenyl Xylyl, inda “selective' for none of these enzymes which are involved in
nyl, and the like, C. to Calkenyl, for example, allyl. (2-oxo 35 purine and pyrimidine metabolism). Assays for determining
1,3-dioxolyl)methyl, and the like, and 4,5-dihydro-4-oxo the activity for each of the above-described enzymes is
1H-pyrazolo 3,4-dpyrimidin-1-yl)methyl, and the like. described in Yasuhiro Takano, et al., Life Sciences, 76:1835
In R in Formula II, the term “ester as used in the phrase 1847 (2005). These preferred compounds have also been
“the ester of carboxymethyl refers to a lower alkyl ester, such referred to in the literature as nonpurine, selective inhibitors
as methyl or ethyl ester; and the term “ether used in the 40 of xathine oxidase (NP/SIXO).
phrase “the ether of hydroxyethyl means an ether which is Examples of compounds having the above Formula II are
formed by substitution of the hydrogen atom of hydroxyl described in U.S. Pat. No. 5,268,386 and EPO 415 566 A1,
group in the hydroxyethyl group by aliphatic or aromatic and are incorporated, in their entirety, herein.
alkyl group, Such as benzyl. With the exception of pyrazolo 1.5-a-1,3,5-triazin-4-
The carboxy-protecting groups may be substituted in vari 45 (1H)-one, 8-3-methoxy-4-(phenylsulfinyl)phenyl-sodium
ous ways. Examples of Substituents include halogen atom, salt (t), methods for making Xanthine oxidoreductase inhib
alkyl groups, alkoxy groups, alkylthio groups and carboxy iting compounds of Formulas I and II for use in the methods
groups. of the present disclosure are known in the art and are
As used herein, the term “straight or branched hydrocarbon described, for example, in U.S. Pat. Nos. 5.268,386, 5,614,
radical” in the definition of A in Formula II above refers to 50 520, 6,225,474, 7,074,816 and EPO 415 566A1 and in the
methylene, ethylene, propylene, methylmethylene, or isopro publications Ishibuchi, S. et al., Bioorg. Med. Chem. Lett.,
pylene. 11:879-882 (2001) and which are each herein incorporated by
As used herein, the substituent of the “substituted nitro reference. Other xanthine oxidoreductase inhibiting com
gen’ in the definition of Y and Z in Formula II above are pounds can be found using Xanthine oxidoreductase and Xan
hydrogen, lower alkyl, or acyl. 55 thine in assays to determine if such candidate compounds
As used herein, the term “phenyl-substituted lower alkyl inhibit conversion of Xanthine into uric acid. Such assays are
refers to a lower alkyl group Substituted with phenyl, such as well known in the art.
benzyl, phenethyl or phenylpropyl. As used herein, the term Pyrazolo 1.5-a-1,3,5-triazin-4-(1H)-one, 8-3-methoxy
“prodrug” refers to a derivative of the compounds shown in 4-(phenylsulfinyl)phenyl-sodium salt (t) is available from
the above-described Formula I and Formula II that have 60 Otsuka Pharmaceutical Co. Ltd. (Tokyo, Japan) and is
chemically or metabolically cleavable groups and become by described in the following publications: Uematsu T., et al.,
Solvolysis or under physiological conditions compounds that "Pharmacokinetic and Pharmacodynamic Properties of a
are pharmaceutically active in vivo. Esters of carboxylic acids Novel Xanthine Oxidase Inhibitor, BOF-4272, in Healthy
are an example of prodrugs that can be used in the dosage Volunteers, J. Pharmacology and Experimental Therapeu
forms of the present disclosure. Methyl ester prodrugs may be 65 tics, 270:453-459 (August 1994), Sato, S., A Novel Xanthine
prepared by reaction of a compound having the above-de Deydrogenase Inhibitor (BOF-4272). In Purine and Pyrimi
scribed formula in a medium Such as methanol with an acid or dine Metabolism in Man, Vol. VII, Part A. ed. By P. A. Hark
US 9.248,119 B2
19 20
ness, pp. 135-138, Plenum Press, New York. Pyrazolo 1.5-a- acceptable salt thereof greater than about 0.1 g/mL for a
1,3,5-triazin-4-(1H)-one, 8-3-methoxy-4-(phenylsulfinyl) period of about 5 hours to about 24 hours; or (b) produce in the
phenyl-Sodium salt (t) can be made using routine techniques Subject a maximum plasma concentration (C) of a Xan
known in the art. thine oxidoreductase inhibitor or a pharmaceutically accept
able salt thereof of between about 2.0 ug/mL to about 1.0
II. Dosage Forms ug/mL. In still yet another embodiment, the modified release
dosage forms of the present disclosure after oral administra
The present disclosure relates to modified release solid tion to a subject in need of treatment thereof exhibit each of
dosage forms comprising at least one active agent. Specifi the following: (a) maintain in the Subject a plasma concen
cally, the at least one active agent contained in the modified 10 tration of Xanthine oxidoreductase inhibitor or pharmaceuti
release solid dosage forms of the present disclosure is at least cally acceptable salt thereof greater than 0.1 ug/mL for a
one Xanthine oxidoreductase inhibitor or at least one Xanthine period of about 5 hours to about 24 hours; and (b) produce in
oxidase inhibitor. the Subjecta about maximum plasma concentration (C) of
The modified release dosage forms of the present disclo a Xanthine oxidoreductase inhibitor or a pharmaceutically
Sure can achieve any one of a number of objects. First, the 15 acceptable salt thereof in an amount of between about 2.5
modified release dosage forms of the present disclosure, ug/mL to about 0.5 ug/mL.
when administered to a subject in need of treatment thereof, As mentioned previously herein, the modified dosage
provide a high percentage of Xanthine oxidoreductase inhibi forms of the present disclosure, after oral administration to a
tion or Xanthine oxidase inhibition at a maximum observed Subject in need of treatment thereof, can maintain in the
plasma concentration (namely C) that is significantly Subject, a plasma concentration of Xanthine oxidoreductase
lower than that provided by an immediate release dosage inhibitor or pharmaceutically acceptable salt thereof greater
form containing at least one Xanthine oxidoreductase inhibi than about 0.1 g/mL for a period of from about 5 to about 24
tor (for example, an immediate release dosage form contain hours. More specifically, the modified dosage forms of the
ing 40 mg, 80 mg, 120 mg or 240 mg offebuXostat which is present disclosure after oral administration to a Subject in
administered to a Subject once a day) similar to or lower than 25 need of treatment thereof, can maintain in the Subject, a
the highest doses Xanthine oxidoreductase inhibitors cur plasma concentration of Xanthine oxidoreductase inhibitor or
rently available (namely, currently available dosage (e.g., 80 pharmaceutically acceptable salt thereof greater than about
mg (US) or 120 mg (Europe)) of 2-3-cyano-4-(2-methylpro 0.1 ug/mL for a period for about 5.0 hours, for about 6.0
poxy)phenyl]-4-methylthiazole-5-carboxylic acid (which is hours, for about 7.0 hours, for about 8.0 hours, for about 9.0
also known as “febuxostat')) or at least one Xanthine oxidase 30 hours, for about 10.0 hours, for about 11.0 hours, for about
inhibitor (for example, an immediate release dosage form 12.0 hours, for about 13.0 hours, for about 14.0 hours, for
containing 300 mg of allopurinol which is administered to a about 15.0 hours, for about 16.0 hours, for about 17.0 hours,
Subject once a day). Second, because the dosage forms of the for about 18.0 hours, for about 19.0 hours, for about 20.0
present disclosure provide for Xanthine oxidoreductase inhi hours, for about 21.0 hours, for about 22.0 hours, for about
bition or xanthine oxidase inhibition, for extended time (dos 35 23.0 hours or for about 24.0 hours.
ing) periods, these solid dosage forms can be used to treat a As also mentioned previously herein, the modified dosage
variety of different conditions or diseases, such as, but not forms of the present disclosure after oral administration to a
limited to, gout, hyperuricemia, prostatitis, inflammatory Subject in need of treatment thereof, can produce, in the
bowel disease, QT interval prolongation, myocardial infarc Subject, a maximum plasma concentration (C) of a Xan
tion, cardiac hypertrophy, hypertension, nephrolithiasis, 40 thine oxidoreductase inhibitor or a pharmaceutically accept
renal impairment, chronic kidney disease, metabolic Syn able salt thereof in an amount of between about 2.5ug/mL to
drome, diabetes, diabetic nephropathy, congestive heart fail about 0.5 ug/mL (as well as any combination of ranges in
ure and other disorders. Third, the modified release dosage between, Such as, for example, about 2.5ug/mL to about 0.6
forms of the present disclosure protect Subjects receiving ug/mL, about 2.5ug/mL to about 0.7 ug/mL, about 2.5ug/mL
these dosage forms throughout their treatment regimen 45 to about 0.8 g/mL, about 2.4 ug/mL to about 0.5 ug/mL,
against increasing concentrations of oxygen free radicals. about 2.4 ug/mL to about 0.6 ug/mL, about 2.3 ug/mL to
In order to obtain these benefits, the modified release dos about 0.5ug/mL, about 2.2 Lug/mL to about 0.5ug/mL, about
age forms of the present disclosure must achieve a certain 2.1 ug/mL to about 0.5 g/mL, about 2.0 ug/mL to about 0.5
pharmacokinetic profile when compared to immediate ug/mL, about 2.0 ug/mL to about 1.0 ug/mL, about 1.9Lug/mL
release Xanthine oxidoreductase inhibitor or Xanthine oxidase 50 to about 0.5 g/mL, about 1.9 Lig/mL to about 1.0 ug/mL,
inhibitor dosage forms. about 1.8 Lig/mL to about 0.5 ug/mL, about 1.8 ug/mL to
In one embodiment, the modified release dosage forms of about 1.0 ug/mL, about 1.7 Lig/mL to about 0.5ug/mL, about
the present disclosure containing at least one Xanthine oxi 1.7 ug/mL to about 0.6 g/mL, about 1.7 ug/mL to about 0.7
doreductase inhibitor after oral administration to a subject in ug/mL, about 1.7 ug/mL to about 0.8 ug/mL, about 1.7 ug/mL
need of treatment thereof exhibit at least two of the following: 55 to about 1.0 g/mL, about 1.6 ug/mL to about 0.5 ug/mL,
(a) maintain in the Subject a plasma concentration of Xanthine about 1.5ug/mL to about 1.0 g/mL etc.). More specifically,
oxidoreductase inhibitor or pharmaceutically acceptable salt the modified dosage forms of the present disclosure can, after
thereof greater than about 0.1 lug/mL for a period of about 5 oral administration to a Subject in need of treatment thereof,
hours to about 24 hours; or (b) produce in the subject a produce in the Subject, a C of a Xanthine oxidoreductase
maximum plasma concentration (C) of a Xanthine oxi 60 inhibitor or a pharmaceutically acceptable salt thereof in an
doreductase inhibitor or a pharmaceutically acceptable salt amount of about 2.5 ug/mL, about 2.4 ug/mL, about 2.3
thereof in an amount of between about 2.5ug/mL to about 0.5 ug/mL, about 2.2 Lug/mL, about 2.1 ug/mL, 2.0 Lig/mL about
ug/mL. In another embodiment, the modified release dosage 1.9 g/mL, about 1.8 Lug/mL, about 1.7 Lig/mL, about 1.6
forms of the present disclosure after oral administration to a ug/mL, about 1.5ug/mL, about 1.4 ug/mL, about 1.3 ug/mL,
subject in need of treatment thereof exhibit at least two of the 65 about 1.2 g/mL, about 1.1 ug/mL, about 1.0 ug/mL, about
following: (a) maintain in the Subject a plasma concentration 0.9 g/mL, about 0.8 ug/mL, about 0.7 ug/mL, about 0.6
of xanthine oxidoreductase inhibitor or pharmaceutically ug/mL or about 0.5ug/mL.
US 9.248,119 B2
21 22
The dosage forms of the present disclosure can contain ug/mL, about 2.5 ug/mL to about 0.07 ug/mL, about 2.5
from about 5 mg to about 240 mg of at least one Xanthine ug/mL to about 0.08 ug/mL, about 2.5 lug/mL to about 0.09
oxidoreductase inhibitor. More specifically, the dosage form ug/mL, about 2.5ug/mL to about 0.1 ug/mL, about 2.5ug/mL
can containabout 5 mg, about 6.25 mg, about 10 mg, about 20 to about 0.2 g/mL, about 2.5 Lig/mL to about 0.3 ug/mL,
mg, about 25 mg, about 30 mg, about 40 mg, about 50 mg. about 2.5 Lig/mL to about 0.40 g/mL, about 2.5 ug/mL to
about 60 mg, about 70 mg, about 75 mg, about 80 mg, about about 0.5ug/mL about 2.5ug/mL to about 0.6 g/mL, about
90 mg, about 100 mg, about 110 mg, about 120 mg, about 130 2.5 lug/mL to about 0.7 g/mL, about 2.5 ug/mL to about 0.8
mg, about 140 mg, about 150 mg, about 160 mg, about 170 ug/mL, about 2.5ug/mL to about 0.91g/mL, about 2.5ug/mL
mg, about 180 mg, about 190 mg, about 200 mg, about 210 to about 1.0 ug/mL, 2.4 g/mL to about 0.05 ug/mL, 2.4
mg, about 220 mg, about 230 mg or about 240 mg of at least 10 ug/mL to about 0.06 ug/mL, about 2.4 ug/mL to about 0.07
one Xanthine oxidoreductase inhibitor. ug/mL, about 2.4 ug/mL to about 0.08 Lig/mL, about 2.4
In another embodiment, the modified release dosage forms ug/mL to about 0.09 g/mL, about 2.4 g/mL to about 0.1
of the present disclosure containing at least one Xanthine ug/mL, about 2.4 ug/mL to about 0.2 ug/mL, about 2.4 ug/mL
oxidoreductase inhibitor after oral administration to a subject to about 0.3 ug/mL, about 2.4 g/mL to about 0.40 ug/mL,
in need of treatment thereof exhibit at least two of the follow 15 about 2.4 ug/mL to about 0.5 g/mL about 2.4 g/mL to about
ing: (a) maintain in the Subject a plasma concentration of 0.6 ug/mL, about 2.4 g/mL to about 0.7 ug/mL, about 2.4
Xanthine oxidoreductase inhibitor or pharmaceutically ug/mL to about 0.8 ug/mL, about 2.4 g/mL to about 0.9
acceptable salt thereof greater than about 0.1 g/mL for a ug/mL, 2.4 ug/mL to about 1.0 ug/mL, 2.3 ug/mL to about
period of about 5 hours to about 16 hours; or (b) produce in the 0.06 ug/mL, about 2.3 ug/mL to about 0.07 g/mL, about 2.3
Subject a maximum plasma concentration (C) of a Xan ug/mL to about 0.08 ug/mL, about 2.3 ug/mL to about 0.09
thine oxidoreductase inhibitor or a pharmaceutically accept ug/mL, about 2.3 ug/mL to about 0.1 ug/mL, about 2.3 ug/mL
able salt thereof in an amount of between about 2.5ug/mL to to about 0.2 g/mL, about 2.3 ug/mL to about 0.3 ug/mL,
about 0.05 g/mL. In yet another embodiment, the modified about 2.3 ug/mL to about 0.40 g/mL, about 2.3 ug/mL to
release dosage forms of the present disclosure after oral about 0.5ug/mL about 2.3 ug/mL to about 0.6 g/mL, about
administration to a subject in need of treatment thereof 25 2.3 ug/mL to about 0.7 g/mL, about 2.3 ug/mL to about 0.8
exhibit at least two of the following: (a) maintain in the ug/mL, about 2.3 ug/mL to about 0.91g/mL, about 2.3 ug/mL
Subject a plasma concentration of Xanthine oxidoreductase to about 1.0 ug/mL, 2.2 g/mL to about 0.05 ug/mL, 2.2
inhibitor or pharmaceutically acceptable salt thereof greater ug/mL to about 0.06 ug/mL, about 2.2 lug/mL to about 0.07
than about 0.1 ug/mL for a period of about 5 hours to about 16 ug/mL, about 2.2 Lug/mL to about 0.08 Lig/mL, about 2.2
hours; or (b) produce in the Subject a maximum plasma con 30 ug/mL to about 0.09 g/mL, about 2.2 g/mL to about 0.1
centration (C) of a Xanthine oxidoreductase inhibitor or a ug/mL, about 2.2 Lug/mL to about 0.2 ug/mL, about 2.2 Lug/mL
pharmaceutically acceptable salt thereof of between about to about 0.3 ug/mL, about 2.2 Lig/mL to about 0.40 g/mL,
2.0 ug/mL to about 0.075ug/mL. In still yet another embodi about 2.2 Lug/mL to about 0.5 g/mL about 2.2 g/mL to about
ment, the modified release dosage forms of the present dis 0.6 ug/mL, about 2.4 g/mL to about 0.7 ug/mL, about 2.2
closure after oral administration to a subject in need of treat 35 ug/mL to about 0.8 ug/mL, about 2.2 g/mL to about 0.9
ment thereof exhibit each of the following: (a) maintain in the ug/mL, about 2.2 g/mL to about 1.0 g/mL, 2.1 g/mL to
Subject a plasma concentration of Xanthine oxidoreductase about 0.05ug/mL, 2.1 ug/mL to about 0.06 g/mL, about 2.1
inhibitor or pharmaceutically acceptable salt thereof greater ug/mL to about 0.07 ug/mL, about 2.1 ug/mL to about 0.08
than 0.1 ug/mL for a period of about 5 hours to about 16 ug/mL, about 2.1 ug/mL to about 0.09 ug/mL, about 2.1
hours; and (b) produce in the Subject a about maximum 40 ug/mL to about 0.1 ug/mL, about 2.1 g/mL to about 0.2
plasma concentration (C) of a Xanthine oxidoreductase ug/mL, about 2.1 ug/mL to about 0.3 ug/mL, about 2.1 ug/mL
inhibitor or a pharmaceutically acceptable salt thereof in an to about 0.40 ug/mL, about 2.1 ug/mL to about 0.5 ug/mL
amount of between about 2.5 lug/mL to about 0.05ug/mL about 2.1 ug/mL to about 0.6 ug/mL, about 2.1 ug/mL to
As mentioned previously herein, the modified dosage about 0.7 ug/mL, about 2.1 ug/mL to about 0.8 ug/mL, about
forms of the present disclosure, after oral administration to a 45 2.1 ug/mL to about 0.9 g/mL, about 2.1 ug/mL to about 1.0
Subject in need of treatment thereof, can maintain in the ug/mL, 2.0 ug/mL to about 0.05ug/mL, 2.0 g/mL to about
Subject, a plasma concentration of Xanthine oxidoreductase 0.06 ug/mL, about 2.0 ug/mL to about 0.07 g/mL, about 2.0
inhibitor or pharmaceutically acceptable salt thereof greater ug/mL to about 0.08 ug/mL, about 2.0 ug/mL to about 0.09
than about 0.1 lug/mL for a period of from about 5 to about 16 ug/mL, about 2.0 ug/mL to about 0.1 ug/mL, about 2.0Lug/mL
hours. More specifically, the modified dosage forms of the 50 to about 0.2 g/mL, about 2.0 ug/mL to about 0.3 ug/mL,
present disclosure after oral administration to a Subject in about 2.0 ug/mL to about 0.40 g/mL, about 2.0 ug/mL to
need of treatment thereof, can maintain in the Subject, a about 0.5ug/mL about 2.0 ug/mL to about 0.6 g/mL, about
plasma concentration of Xanthine oxidoreductase inhibitor or 2.0 ug/mL to about 0.7 g/mL, about 2.0 ug/mL to about 0.8
pharmaceutically acceptable salt thereof greater than about ug/mL, about 2.0 ug/mL to about 0.91g/mL, about 2.0Lug/mL
0.1 ug/mL for a period for about 5.0 hours, for about 6.0 55 to about 1.0 ug/mL, 1.9 g/mL to about 0.05 ug/mL, 1.9
hours, for about 7.0 hours, for about 8.0 hours, for about 9.0 ug/mL to about 0.06 ug/mL, about 1.9 ug/mL to about 0.07
hours, for about 10.0 hours, for about 11.0 hours, for about ug/mL, about 1.9 ug/mL to about 0.08 Lig/mL, about 1.9
12.0 hours, for about 13.0 hours, for about 14.0 hours, for ug/mL to about 0.09 g/mL, about 1.9 ug/mL to about 0.1
about 15.0 hours, or for about 16.0 hours. ug/mL, about 1.9Lug/mL to about 0.2 ug/mL, about 1.9Lug/mL
As also mentioned previously herein, the modified dosage 60 to about 0.3 ug/mL, about 1.9 g/mL to about 0.40 ug/mL,
forms of the present disclosure after oral administration to a about 1.9Lug/mL to about 0.5 g/mL about 1.9 g/mL to about
Subject in need of treatment thereof, can produce, in the 0.6 ug/mL, about 1.9 g/mL to about 0.7 ug/mL, about 1.9
Subject, a maximum plasma concentration (C) of a Xan ug/mL to about 0.8 ug/mL, about 1.9 g/mL to about 0.9
thine oxidoreductase inhibitor or a pharmaceutically accept ug/mL, about 1.9 g/mL to about 1.0 g/mL, 1.8 g/mL to
able salt thereofinan amount of between about 2.5 mg/mL to 65 about 0.05ug/mL, 1.8 ug/mL to about 0.06 g/mL, about 1.8
about 0.05 ug/mL (as well as any combination of ranges in ug/mL to about 0.07 ug/mL, about 1.8 ug/mL to about 0.08
between, such as, for example, about 2.5ug/mL to about 0.06 ug/mL, about 1.8 Lug/mL to about 0.09 ug/mL, about 1.8
US 9.248,119 B2
23 24
ug/mL to about 0.1 ug/mL, about 1.8 Lig/mL to about 0.2 mg, about 25 mg, about 30 mg, about 40 mg, about 50 mg.
ug/mL, about 1.8g/mL to about 0.3 ug/mL, about 1.8Lug/mL about 60 mg, about 70 mg, about 75 mg, about 80 mg, about
to about 0.40 g/mL, about 1.8 g/mL to about 0.5 ug/mL 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130
about 1.8 Lig/mL to about 0.6 g/mL, about 1.8 ug/mL to mg, about 140 mg, about 150 mg, about 160 mg, about 170
about 0.7 g/mL, about 1.8 Lig/mL to about 0.8 ug/mL, about mg, about 180 mg, about 190 mg, about 200 mg, about 210
1.8 ug/mL to about 0.9 g/mL, about 1.8 g/mL to about 1.0 mg, about 220 mg, about 230 mg or about 240 mg of at least
ug/mL, 1.7 ug/mL to about 0.05 g/mL, 1.7 Lig/mL to about one Xanthine oxidoreductase inhibitor.
0.06 ug/mL, about 1.7 ug/mL to about 0.07 g/mL, about 1.7 In yet another embodiment, the modified release dosage
ug/mL to about 0.08 ug/mL, about 1.7 ug/mL to about 0.09 forms of the present disclosure containing at least one Xan
ug/mL, about 1.7 g/mL to about 0.1 ug/mL, about 1.7Lug/mL 10 thine oxidoreductase inhibitor after oral administration to a
to about 0.2 g/mL, about 1.7 Lig/mL to about 0.3 ug/mL, subject in need of treatment thereof exhibit at least two of the
about 1.7 g/mL to about 0.40 g/mL, about 1.7 ug/mL to following: (a) maintain in the Subject a plasma concentration
about 0.5ug/mL about 1.7 ug/mL to about 0.6 g/mL, about of xanthine oxidoreductase inhibitor or pharmaceutically
1.7 ug/mL to about 0.7 g/mL, about 1.7 g/mL to about 0.8 acceptable salt thereof greater than about 0.1 g/mL for a
ug/mL, about 1.7 g/mL to about 0.91g/mL, about 1.7Lug/mL 15 period of about 5 hours to about 14 hours; or (b) produce in the
to about 1.0 ug/mL, 1.6 ug/mL to about 0.05 ug/mL, 1.6 Subject a maximum plasma concentration (C) of a Xan
ug/mL to about 0.06 ug/mL, about 1.6 ug/mL to about 0.07 thine oxidoreductase inhibitor or a pharmaceutically accept
ug/mL, about 1.6 g/mL to about 0.08 Lig/mL, about 1.6 able salt thereof in an amount of between about 2.5ug/mL to
ug/mL to about 0.09 g/mL, about 1.6 g/mL to about 0.1 about 0.090 ug/mL. In yet another embodiment, the modified
ug/mL, about 1.6 g/mL to about 0.2 ug/mL, about 1.6Lug/mL release dosage forms of the present disclosure after oral
to about 0.3 ug/mL, about 1.6 g/mL to about 0.40 ug/mL, administration to a subject in need of treatment thereof
about 1.6 ug/mL to about 0.5 g/mL about 1.6 ug/mL to about exhibit at least two of the following: (a) maintain in the
0.6 ug/mL, about 1.6 g/mL to about 0.7 ug/mL, about 1.6 Subject a plasma concentration of Xanthine oxidoreductase
ug/mL to about 0.8 ug/mL, about 1.6 ug/mL to about 0.9 inhibitor or pharmaceutically acceptable salt thereof greater
ug/mL, about 1.6 g/mL to about 1.0 g/mL, 1.5 g/mL to 25 than about 0.1 ug/mL for a period of about 5 hours to about 14
about 0.05ug/mL, 1.5ug/mL to about 0.06 g/mL, about 1.5 hours; or (b) produce in the Subject a maximum plasma con
ug/mL to about 0.07 ug/mL, about 1.5 lug/mL to about 0.08 centration (C) of a Xanthine oxidoreductase inhibitor or a
ug/mL, about 1.5 g/mL to about 0.09 ug/mL, about 1.5 pharmaceutically acceptable salt thereof of between about
ug/mL to about 0.1 ug/mL, about 1.5 ug/mL to about 0.2 2.0 ug/mL to about 0.095 ug/mL. In still yet another embodi
ug/mL, about 1.5 g/mL to about 0.3 ug/mL, about 1.5ug/mL 30 ment, the modified release dosage forms of the present dis
to about 0.40 g/mL, about 1.5 g/mL to about 0.5 ug/mL closure after oral administration to a subject in need of treat
about 1.5 Lig/mL to about 0.6 g/mL, about 1.5 ug/mL to ment thereof exhibit each of the following: (a) maintain in the
about 0.7 g/mL, about 1.5ug/mL to about 0.8 ug/mL, about Subject a plasma concentration of Xanthine oxidoreductase
1.5ug/mL to about 0.9 ug/mL or about 1.5ug/mL to about 1.0 inhibitor or pharmaceutically acceptable salt thereof greater
ug/mL. More specifically, the modified dosage forms of the 35 than 0.1 g/mL for a period of about 5 hours to about 14
present disclosure can, after oral administration to a subject in hours; and (b) produce in the Subject a about maximum
need of treatment thereof, produce in the Subject, a C of a plasma concentration (C) of a Xanthine oxidoreductase
Xanthine oxidoreductase inhibitor or a pharmaceutically inhibitor or a pharmaceutically acceptable salt thereof in an
acceptable salt thereof in an amount of 2.5 g/mL, about 2.4 amount of between about 2.5 g/mL to about 0.090 ug/mL.
g/mL, about 2.3 Lig/mL, about 2.2 Lug/mL, about 2.1 Lug/mL, 40 As mentioned previously herein, the modified dosage
2.0 ug/mL about 1.9 ug/mL, about 1.8 Lug/mL, about 1.7 forms of the present disclosure, after oral administration to a
ug/mL, about 1.6 ug/mL, about 1.5 ug/mL, about 1.4 ug/mL, Subject in need of treatment thereof, can maintain in the
about 1.3 g/mL, about 1.2 Lug/mL, about 1.1 ug/mL, about Subject, a plasma concentration of Xanthine oxidoreductase
1.0 g/mL, about 0.9 g/mL, about 0.8 ug/mL, about 0.7 inhibitor or pharmaceutically acceptable salt thereof greater
ug/mL, about 0.6 ug/mL, about 0.5ug/mL, about 0.4 ug/mL, 45 than about 0.1 g/mL for a period of from about 5 to about 14
about 0.3 g/mL, about 0.2 Lug/mL, about 0.1 ug/mL, about hours. More specifically, the modified dosage forms of the
0.099 ug/mL, about 0.098 ug/mL, about 0.097 ug/mL, about present disclosure after oral administration to a Subject in
0.096 ug/mL, about 0.095 ug/mL, about 0.094 ug/mL, about need of treatment thereof, can maintain in the Subject, a
0.093 ug/mL, about 0.092 lug/mL, about 0.091 ug/mL, about plasma concentration of Xanthine oxidoreductase inhibitor or
0.090 ug/mL, about 0.089 ug/mL, about 0.088 ug/mL, about 50 pharmaceutically acceptable salt thereof greater than about
0.087 ug/mL, about 0.086 ug/mL, about 0.085ug/mL, about 0.1 ug/mL for a period for about 5.0 hours, for about 6.0
0.084 ug/mL, about 0.083 lug/mL, about 0.082 ug/mL, about hours, for about 7.0 hours, for about 8.0 hours, for about 9.0
0.081 ug/mL, about 0.080 ug/mL, about 0.079 lug/mL, about hours, for about 10.0 hours, for about 11.0 hours, for about
0.078 ug/mL, about 0.077 ug/mL, about 0.076 ug/mL, about 12.0 hours, for about 13.0 hours or for about 14.0 hours.
0.075 ug/mL, about 0.074 ug/mL, about 0.073 ug/mL, about 55 As also mentioned previously herein, the modified dosage
0.072 ug/mL, about 0.071 ug/mL, about 0.070 ug/mL, about forms of the present disclosure after oral administration to a
0.069 ug/mL, about 0.068 ug/mL, about 0.067 ug/mL, about Subject in need of treatment thereof, can produce, in the
0.066 ug/mL, about 0.065 lug/mL, about 0.064 ug/mL, about Subject, a maximum plasma concentration (C) of a Xan
0.063 ug/mL, about 0.062 ug/mL, about 0.061 ug/mL, about thine oxidoreductase inhibitor or a pharmaceutically accept
0.060 ug/mL, about 0.059 ug/mL, about 0.058 ug/mL, about 60 able salt thereof in an amount of between about 2.5ug/mL to
0.057 ug/mL, about 0.056 ug/mL, about 0.055ug/mL, about about 0.090 ug/mL (as well as any combination of ranges in
0.054 ug/mL, about 0.053 ug/mL, about 0.052 ug/mL, about between, Such as, for example, about 2.5ug/mL to about 0.1
0.051 ug/mL or about 0.050 ug/mL. ug/mL, about 2.5ug/mL to about 0.2 ug/mL, about 2.5ug/mL
The dosage forms of the present disclosure can contain to about 0.3 ug/mL, about 2.5 g/mL to about 0.40 ug/mL,
from about 5 mg to about 240 mg of at least one Xanthine 65 about 2.5ug/mL to about 0.5 g/mL about 2.5 g/mL to about
oxidoreductase inhibitor. More specifically, the dosage form 0.6 ug/mL, about 2.5 g/mL to about 0.7 ug/mL, about 2.5
can containabout 5 mg, about 6.25 mg, about 10 mg, about 20 ug/mL to about 0.8 ug/mL, about 2.5 g/mL to about 0.9
US 9.248,119 B2
25 26
ug/mL, about 2.5 g/mL to about 1.0 ug/mL, about 2.4 ug/mL the Subject, a C of a Xanthine oxidoreductase inhibitor or a
to about 0.1 g/mL, about 2.4 ug/mL to about 0.1 ug/mL, pharmaceutically acceptable salt thereof in an amount of 2.5
about 2.4 ug/mL to about 0.2 g/mL, about 2.4 ug/mL to g/mL, about 2.4 Lig/mL, about 2.3 Lig/mL, about 2.2 Lug/mL,
about 0.3 ug/mL, about 2.4 g/mL to about 0.40 ug/mL, about about 2.1 g/mL, 2.0 ug/mL about 1.9 Lig/mL, about 1.8
2.4 ug/mL to about 0.5/mL about 2.4 g/mL to about 0.6 ug/mL, about 1.7 Lig/mL, about 1.6 ug/mL, about 1.5 Lig/mL,
ug/mL, about 2.4 g/mL to about 0.7 ug/mL, about 2.4 ug/mL about 1.4 g/mL, about 1.3 ug/mL, about 1.2 Lug/mL, about
to about 0.8 ug/mL, about 2.4 g/mL to about 0.9 ug/mL, 2.4 1.1 g/mL, about 1.0 ug/mL, about 0.9 ug/mL, about 0.8
ug/mL to about 1.0 ug/mL, about 2.3 ug/mL to about 0.1 ug/mL, about 0.7 Lig/mL, about 0.6 ug/mL, about 0.5ug/mL,
ug/mL, about 2.3 g/mL to about 0.2 ug/mL, about 2.3 ug/mL about 0.4 g/mL, about 0.3 ug/mL, about 0.2 Lug/mL, about
to about 0.3 ug/mL, about 2.3 g/mL to about 0.40 ug/mL, 10
0.1 ug/mL, about 0.099 ug/mL, about 0.098 g/mL, about
about 2.3 ug/mL to about 0.5 g/mL about 2.3 ug/mL to about 0.097 ug/mL, about 0.096 mg/mL, about 0.095 g/mL, about
0.6 ug/mL, about 2.3 g/mL to about 0.7 ug/mL, about 2.3 0.094 g/mL, about 0.093 lug/mL, about 0.092 lug/mL, about
ug/mL to about 0.8 ug/mL, about 2.3 ug/mL to about 0.9 0.091 lug/mL or about 0.090 ug/mL.
ug/mL, about 2.3 g/mL to about 1.0 ug/mL, about 2.2 Lug/mL
to about 0.1 g/mL, about 2.2 Lug/mL to about 0.2 Lug/mL, 15 The dosage forms of the present disclosure can contain
about 2.2 Lug/mL to about 0.3 g/mL, about 2.2 Lug/mL to from about 5 mg to about 240 mg of at least one Xanthine
about 0.40 ug/mL, about 2.2 Lug/mL to about 0.5ug/mL about oxidoreductase inhibitor. More specifically, the dosage form
2.2 Lug/mL to about 0.6 g/mL, about 2.4 g/mL to about 0.7 can contain about 5 mg, about 6.25 mg, about 10 mg, about 20
ug/mL, about 2.2 g/mL to about 0.8 ug/mL, about 2.2 Lug/mL mg, about 25 mg, about 30 mg, about 40 mg, about 50 mg.
to about 0.9 g/mL, about 2.2 Lug/mL to about 1.0 ug/mL, about 60 mg, about 70 mg, about 75 mg, about 80 mg, about
about 2.1 ug/mL to about 0.1 g/mL, about 2.1 ug/mL to 90 mg, about 100 mg, about 110 mg, about 120 mg, about 130
about 0.2 g/mL, about 2.1 ug/mL to about 0.3 ug/mL, about mg, about 140 mg, about 150 mg, about 160 mg, about 170
2.1 g/mL to about 0.40 g/mL, about 2.1 g/mL to about 0.5 mg, about 180 mg, about 190 mg, about 200 mg, about 210
ug/mL about 2.1 g/mL to about 0.6 ug/mL, about 2.1 ug/mL mg, about 220 mg, about 230 mg or about 240 mg of at least
to about 0.7 g/mL, about 2.1 ug/mL to about 0.8 ug/mL, 25 one Xanthine oxidoreductase inhibitor.
about 2.1 ug/mL to about 0.9 g/mL, about 2.1 ug/mL to Methods for determining the C of xanthine oxidoreduc
about 1.0 g/mL, about 2.0 ug/mL to about 0.1 ug/mL, about tase inhibitors and the plasma concentration of Xanthine oxi
2.0 ug/mL to about 0.2 g/mL, about 2.0 g/mL to about 0.3 doreductase inhibitors are well known in the art. In order to
ug/mL, about 2.0 g/mL to about 0.40 ug/mL, about 2.0 determine the percentage of inhibition of Xanthine oxi
ug/mL to about 0.5 g/mL about 2.0 ug/mL to about 0.6 30
doreductase exhibited by a dosage form, the following equa
ug/mL, about 2.0 g/mL to about 0.7 ug/mL, about 2.0Lug/mL tion can be used:
to about 0.8 g/mL, about 2.0 ug/mL to about 0.9 g/mL, Percent Inhibition ("/6 Inhibition') of Xanthine oxi
about 2.0 ug/mL to about 1.0 g/mL, about 1.9 ug/mL to doreductase activity:
about 0.1 g/mL, about 1.9 ug/mL to about 0.2 ug/mL, about
1.9 g/mL to about 0.3 ug/mL, about 1.9 ug/mL to about 0.40 35
ug/mL, about 1.9Lug/mL to about 0.5ug/mL about 1.9Lug/mL C. f.
to about 0.6 g/mL, about 1.9 Lig/mL to about 0.7 Lig/mL, %. Inhibition of xanthine oxidoreductase = 100
C. f... + K.
about 1.9 ug/mL to about 0.8 g/mL, about 1.9 ug/mL to
about 0.9 g/mL, about 1.9 ug/mL to about 1.0 ug/mL, about
1.8 ug/mL to about 0.1 g/mL, about 1.8 g/mL to about 0.2 40 where C plasma concentration of Xanthine oxidoreductase
ug/mL, about 1.8g/mL to about 0.3 ug/mL, about 1.8Lug/mL inhibitor (XORI) in plasma of a subject, f, the free fraction
to about 0.40 g/mL, about 1.8 g/mL to about 0.5 ug/mL of XORI in plasma and K, the xanthine oxidoreductase
about 1.8 Lig/mL to about 0.6 g/mL, about 1.8 ug/mL to inhibitory constant of XORI.
about 0.7 g/mL, about 1.8 Lig/mL to about 0.8 ug/mL, about The plasma concentration of XORI can be determined
1.8 ug/mL to about 0.9 g/mL, about 1.8 g/mL to about 1.0 45
using techniques known in the art such as high performance
ug/mL, 1.7 ug/mL to about 0.05 g/mL, about 1.7 g/mL to liquid chromatography with fluorescence detection or vali
about 0.1 g/mL, about 1.7 Lig/mL to about 0.2 ug/mL, about dated high performance liquid chromatography tandem mass
1.7 g/mL to about 0.3 ug/mL, about 1.7 ug/mL to about 0.40 spectrometry (See Mayer, M. et al., American Journal of
ug/mL, about 1.7 Lig/mL to about 0.5ug/mL about 1.7 ug/mL
to about 0.6 g/mL, about 1.7 Lig/mL to about 0.7 Lig/mL, 50 Therapeutics, 12:22-34 (2005)). The f, can be determined
about 1.7 Lig/mL to about 0.8 g/mL, about 1.7 ug/mL to using the in-vitro binding of ''C XORI at a nominal concen
about 0.9 ug/mL, about 1.7 Lig/mL to about 1.0 g/mL, 1.6 tration of 1 Jug/mL using an equilibrium dialysis technique,
ug/mL to about 0.1 ug/mL, about 1.6 ug/mL to about 0.2 which is well known in the art. For example, thef, for aXORI
ug/mL, about 1.6 g/mL to about 0.3 ug/mL, about 1.6Lug/mL such as febuxostat has been calculated to be 0.9+0.2 in normal
to about 0.40 g/mL, about 1.6 g/mL to about 0.5 ug/mL 55 patients and 1.2-0.2 in patients with severe renal impairment
about 1.6 ug/mL to about 0.6 g/mL, about 1.6 ug/mL to (See Mayer, M. et al., American Journal of Therapeutics,
about 0.7 g/mL, about 1.6 ug/mL to about 0.8 ug/mL, about 12:22-34 (2005)). In another study with a larger number of
1.6 ug/mL to about 0.9 g/mL, about 1.6 g/mL to about 1.0 Subjects, the percent free fraction offebuXostatin plasma was
ug/mL, about 1.5 g/mL to about 0.1 ug/mL, about 1.5ug/mL calculated to be 0.7+0.1 in male, female, younger, and an
to about 0.2 g/mL, about 1.5 Lig/mL to about 0.3 ug/mL, 60 elderly group of subjects (See Khosravan R., et al. Clinic.
about 1.5 g/mL to about 0.40 g/mL, about 1.5 ug/mL to Pharmacology & Therapeutics, P50 (2005)).
about 0.5ug/mL about 1.5ug/mL to about 0.6 g/mL, about The K, for XORIs can be determined using routine tech
1.5 lug/mL to about 0.7 g/mL, about 1.5 g/mL to about 0.8 niques known in the art. For example, the Ki for a XORI such
ug/mL, about 1.5 ug/mL to about 0.9 g/mL or about 1.5 as febuXostat has been determined using a Xanthine oxidase
ug/mL to about 1.0 g/mL. More specifically, the modified 65 assay Such as that described in Osada Y., et al., European J.
dosage forms of the present disclosure can, after oral admin Pharmacology, 241:183-188 (1993). More specifically, the
istration to a subject in need of treatment thereof, produce in K, for febuxostat has been determined to be 0.7 nM and 0.6
US 9.248,119 B2
27 28
nM, respectively (See, Osada Y., et al., European J. Pharma as, matrix systems, osmotic pumps or membrane controlled
cology, 241:183-188 (1993) and Takano, Y., et al., Life Sci technology (also referred to as reservoir system), are
ences, 76:1835-1847 (2005)). described in greater detail below. A detailed discussion of
In still yet another embodiment, the modified release dos these dosage forms may also be found in: (i) Handbook of
age forms of the present disclosure contain at least one Xan 5 pharmaceutical controlled release technology, ed. D. L.
thine oxidase inhibitor. These modified release dosage forms Wise, Marcel Dekker, Inc. New York, N.Y. (2000), and (ii).
containing at least one Xanthine oxidase inhibitor, after oral Treatise on controlled drug delivery, fundamentals, optimi
administration to a Subject, are expected to maintain critical Zation, and applications, ed. A. Kydonieus, Marcel Dekker,
concentration in plasma for longer durations compared to Inc. New York, N.Y. (1992), the contents of each which is
immediate release formulations containing allopurinol 10 hereby incorporated by reference. However, although these
thereby inhibiting target enzyme for a prolonged period of three oral polymeric modified release dosage forms are
time. Thus, these modified release dosage forms will be described in greater detail, other modified release dosage
advantageous over immediate release tablets since these forms known to those skilled in the art are contemplated to be
modified release dosage forms would reduce inter-patient within the scope of the present disclosure.
variability due to variation of half life of oxypurinol and 15 Matrix Systems
allopurinol, thereby improving therapeutic outcome. Matrix systems are well known in the art. In a matrix
The dosage forms of the present disclosure can contain, in system, the drug is homogeneously dispersed in a polymer in
addition to a Xanthine oxidoreductase inhibitor or Xanthine association with conventional excipients. This admixture is
oxidase inhibitor, other drugs. These other drugs may be typically compressed underpressure to produce a tablet. Drug
selected from any of the various classes of agents including, is released from this tablet by diffusion and erosion. Matrix
but not limited to, non-steroidal anti-inflammatory agents, systems are described in detail by Wise and Kydonieus, supra.
analgesic agents, anesthetic agents, anti-anginal agents, anti The matrix dosage forms of the present disclosure can
arthritic agents, anti-arrhythmic agents, antiasthmatic agents, comprise a Xanthine oxidoreductase inhibitor or Xanthine
antibacterial agents, anti-BPHagents, anticancer agents, anti oxidase inhibitor and a pharmaceutically acceptable polymer.
cholinergic agents, anticoagulants, anticonvulsants, antide 25 In one aspect, the Xanthine oxidoreductase inhibitor is 2-3-
pressants, antidiabetic agents, antidiarrheals, anti-epileptic cyano-4-2(2-methylpropoxy)phenyl-4-methylthiazole-5-
agents, antifungal agents, antigout agents, antihelminthic carboxylic acid. In another aspect, the Xanthine oxidase
agents, antihistamines, antihypertensive agents, antiinflam inhibitor is allopurinol.
matory agents, antimalarial agents, antimigraine agents, anti The pharmaceutically acceptable polymer is a water
muscarinic agents, antinauseants, antineoplastic agents, anti 30 soluble hydrophilic polymer, or a water insoluble hydropho
obesity agents, antiosteoporosis agents, antiparkinsonism bic polymer (including waxes). Examples of Suitable water
agents, antiprotozoal agents, antipruritics, antipsychotic soluble polymers include polyvinylpyrrolidine, hydroxypro
agents, antipyretics, antispasmodics, antithyroid agents, anti pylcellulose, hydroxypropyl methylcellulose, methyl cellu
tubercular agents, antiulcer agents, anti-urinary incontinence lose, vinyl acetate copolymers, polysaccharides (such as alig
agents, antiviral agents, anxiolytics, appetite Suppressants, 35 nate, Xanthan gum, etc.), polyethylene oxide, methacrylic
attention-deficit disorder (ADD) and attention deficit hyper acid copolymers, maleic anhydride/methyl vinyl ether
activity disorder (ADHD) drugs, calcium channel blockers, copolymers and derivatives and mixtures thereof. Examples
cardiac inotropic agents, beta-blockers, central nervous sys of suitable water insoluble polymers include acrylates, cellu
tem stimulants, cognition enhancers, corticosteroids, COX-2 lose derivatives such ethylcellulose or cellulose acetate, poly
inhibitors, decongestants, diuretics, gastrointestinal agents, 40 ethylene, methacrylates, acrylic acid copolymers and high
genetic materials, drugs used in the management of gout molecular weight polyvinylalcohols. Examples of suitable
(such as colchine; uricoSuric agents such as probenecid, waxes include fatty acids and glycerides.
Sulfinpyrazone, benziodarone; Xanthine oxidase inhibitors In one aspect, the polymer is selected from hydroxypropyl
Such as oxypurinol, allopurinol, etc) histamine receptor cellulose, hydroxypropyl methylcellulose, and methyl cellu
antagonists, hormonolytics, hypnotics, hypoglycemic agents, 45 lose. In another aspect, the polymer is hydroxypropyl meth
immunosuppressants, keratolytics, leukotriene inhibitors, ylcellulose. In still yet another aspect, the polymer is a high
lipid-regulating agents, macrollides, mitotic inhibitors, viscosity hydroxypropyl-methyl cellulose with viscosity
muscle relaxants, narcotic antagonists, neuroleptic agents, ranging from about 4,000 cps to about 100,000 cps. The most
nicotine, nutritional oils, Xanthine derivatives (such as, but preferred high viscosity polymer is a hydroxypropyl methyl
not limited to, caffeine and derivatives of caffeine), parasym 50 cellulose with a viscosity of about 15,000 cps, commercially
patholytic agents, sedatives, sex hormones, sympathomi available under the tradename, Methocel(R), from The Dow
metic agents, tranquilizers, vasodilators, vitamins, and com Chemical Company.
binations thereof. Any of the aforementioned drugs may also The amount of the polymer in the dosage form generally
be administered in combination with the Xanthine oxi varies from about 10% to about 70% by weight of the com
doreductase inhibitors or Xanthine oxidase inhibitors used in 55 position.
the dosage forms of the present disclosure. The dosage forms of the present disclosure will typically
The benefits of the present disclosure are not limited to a include pharmaceutically acceptable excipients. AS is well
single type of dosage form having a particular mechanism of known to those skilled in the art, pharmaceutical excipients
drug release. This enhanced pharmacokinetic profile can be are routinely incorporated into Solid dosage forms. This is
obtained with any of oral extended dosage form known in the 60 done to ease the manufacturing process as well as to improve
art, such as, but not limited to, a pulsatile release dosage form, the performance of the dosage form. Common excipients
an extended release dosage form or a delayed release dosage include diluents or bulking agents, lubricants, binders, etc.
form, following the teachings above. Such excipients can be used in the dosage forms of the present
Many different types of oral polymeric modified release disclosure.
dosages forms are known in the art and are contemplated for 65 Diluents, or fillers, can be added in order to increase the
use in the present disclosure. Examples of three different mass of an individual dose to a size suitable for tablet com
types of oral polymeric modified release dosage forms, such pression. Suitable diluents include powdered Sugar, calcium
US 9.248,119 B2
29 30
phosphate, calcium sulfate, microcrystalline cellulose, lac propylene glycol, Sorbitan monoleate, Sorbic acid, fillers such
tose, mannitol, kaolin, Sodium chloride, dry starch, Sorbitol, as titanium dioxide, and a pharmaceutically acceptable dye.
etc. Osmotic Pumps
Lubricants can be incorporated into the dosage form for a In an osmotic pump system, a tablet core is encased by a
variety of reasons. Lubricants reduce friction between the semipermeable membrane having at least one orifice. The
granulation and die wall during compression and ejection. semipermeable membrane is permeable to water, but imper
This prevents the granulate from sticking to the tablet meable to the drug. When the system is exposed to body
punches, facilitates its ejection from the tablet punches, etc. fluids, water will penetrate through the semipermeable mem
Examples of suitable lubricants that can be used include, but brane into the tablet core containing osmotic excipients and
are not limited to, talc, Stearic acid, vegetable oil, calcium 10 the active drug. Osmotic pressure increases within the dosage
Stearate, Zinc Stearate, magnesium Stearate, etc. form and drug is released through the orifice in an attempt to
Glidants can also be incorporated into the dosage form. A equalize pressure.
glidant improves the flow characteristics of the granulation. In more complex pumps, the tablet core contains two inter
Examples of suitable glidant’s include, but are not limited to, nal compartments. The first compartment contains the drug.
talc, silicon dioxide and cornstarch. 15 The second compartment contains a polymer which Swells on
Binders can be incorporated into the dosage form. Binders contact with fluid. After ingestion, this polymer swells into
are typically utilized if the manufacture of the dosage form the drug containing compartmentata predetermined rate and
includes a granulation step. Examples of Suitable binders forces drug from the dosage form at that rate. Such dosage
include, but are not limited to, povidone, polyvinylpyrroli forms are often used when a zero order release profile is
done, Xanthan gum, cellulose gums such as carboxymethyl desired.
cellulose, methylcellulose, hydroxyethyl cellulose, hydrox Osmotic pumps are well known in the art and have been
ypropyl cellulose, hydroxypropyl methylcellulose, described in the literature. U.S. Pat. Nos. 4,088,864; 4,200,
hydroxycellulose, gelatin, starch, and pregelatinized Starch. 098; and 5,573,776: all of which are hereby incorporated by
Other excipients that can be incorporated into the dosage reference, describe osmotic pumps and methods for their
form include, but are not limited to, preservatives, antioxi 25 manufacture.
dants, or any other excipient commonly used in the pharma As a general guideline, the osmotic pumps of the present
ceutical industry, etc. The amount of excipients used in the disclosure can be formed by compressing a tablet of an
dosage form will correspond to that typically used in a matrix osmotically active drug (or an osmotically inactive drug in
system. The total amount of excipients, fillers and extenders, combination with an osmotically active agent or osmagent)
etc. can vary from about 10% to about 70% by weight of the 30 and then coating the tablet with a semipermeable membrane
dosage form. which is permeable to an exterior aqueous-based fluid but
The matrix dosage forms are generally prepared using impermeable to the passage of drug and/or osmagent. One or
standard techniques well known in the art. Typically, they are more delivery orifices may be drilled through the semiperme
prepared by dry blending the polymer, filler, the xanthine able membrane wall. Alternatively, orifice(s) through the wall
oxidoreductase inhibitor, such as, 2-3-cyano-4-2(2-methyl 35 may be formed in situ by incorporating leachable pore form
propoxy)phenyl-4-methylthiazole-5-carboxylic acid, or ing materials in the wall. In operation, the exterior aqueous
Xanthine oxidase inhibitor, Such as allopurinol and oxypu based fluid is imbibed through the semipermeable membrane
rinol, and other excipients followed by granulating the mix wall and contacts the drug and/or salt to form a solution or
ture using an alcohol until proper granulation is obtained. The Suspension of the drug. The drug solution or Suspension is
granulation is done by methods known in the art. The wet 40 then pumped out through the orifice as fresh fluid is imbibed
granules are dried in a fluid bed dryer, sifted and ground to through the semipermeable membrane.
appropriate size. Lubricating agents are mixed with the dried In one embodiment, the tablet contains two distinct com
granulation to obtain the final dosage form. partments. The first compartment contains the drug as
Alternatively, the matrix dosage forms can be made using described above. The second compartment contains an
direct compression of a powdered, crystalline or granular 45 expandable driving member consisting of a layer of a
composition containing the active agent(s), alone or in com swellable hydrophilic polymer, which operates to diminish
bination with one or more carriers, additives, or the like. the volume occupied by the drug, thereby delivering the drug
Methods of direct compression are well known in the art. from the device at a controlled rate over an extended period of
The dosage forms of the present disclosure can be admin time.
istered orally in the form of tablets, pills, or the granulate may 50 Typical materials for the semipermeable membrane
be loose filled into capsules. The tablets can be prepared by include semipermeable polymers known to the art as osmosis
techniques known in the art and contain a therapeutically and reverse osmosis membranes, such as cellulose acylate,
effective amount of the Xanthine oxidoreductase inhibitor or cellulose diacylate, cellulose triacylate, cellulose acetate, cel
Xanthin oxidase inhibitor and Such excipients as are necessary lulose diacetate, cellulose triacetate, agar acetate, amylose
to form the tablet by such techniques. Tablets and pills can 55 triacetate, beta glucanacetate, acetaldehyde dimethylacetate,
additionally be prepared with enteric coatings and other cellulose acetate ethyl carbamate, polyamides, polyure
release-controlling coatings for the purpose of acid protec thanes, Sulfonated polystyrenes, cellulose acetate phthalate,
tion, easing Swallow ability, etc. The coating may be colored cellulose acetate methyl carbamate, cellulose acetate Succi
with a pharmaceutically accepted dye. The amount of dye and nate, cellulose acetate dimethyl aminoacetate, cellulose
other excipients in the coating liquid may vary and will not 60 acetate ethyl carbamate, cellulose acetate chloracetate, cellu
impact the performance of the modified release tablets. The lose dipalmitate, cellulose dioctanoate, cellulose dicaprylate,
coating liquid generally comprises film forming polymers cellulose dipentanlate, cellulose acetate Valerate, cellulose
Such as, but not limited to, hydroxypropyl cellulose, hydrox acetate Succinate, cellulose propionate Succinate, methyl cel
ypropyl methylcellulose, cellulose esters or ethers (such as lulose, cellulose acetate p-toluene Sulfonate, cellulose acetate
cellulose acetate or ethylcellulose), an acrylic polymer or a 65 butyrate, cross-linked selectively semipermeable polymers
mixture of polymers. The coating solution is generally an formed by the coprecipitation of a polyanion and a polycation
aqueous Solution or an organic solvent further comprising as disclosed in U.S. Pat. Nos. 3,173,876; 3,276,586: 3,541,
US 9.248,119 B2
31 32
005; 3,541,006; and 3,546,142, semipermeable polymers as orifice has been drilled therethrough. A detailed description
disclosed by Loeb and Sourirajan in U.S. Pat. No. 3,133,132, of osmotic passageways and the maximum and minimum
lightly cross-linked polystyrene derivatives, cross-linked dimensions for a passageway are disclosed in U.S. Pat. Nos.
poly(sodium styrene Sulfonate), poly(vinylbenzyltrimethyl 3,845,770 and 3,916,899, the disclosures of which are incor
ammonium chloride), cellulose acetate having a degree of 5 porated herein by reference.
substitution up to 1 and an acetyl content up to 50%, cellulose The osmotic pumps of the present disclosure can be manu
diacetate having a degree of Substitution of 1 to 2 and an factured by standard techniques. For example, in one embodi
acetyl content of 21 to 35%, cellulose triacetate having a ment, the drug and other ingredients that may be housed in
degree of substitution of 2 to 3 and an acetyl content of 35 to one area of the compartment adjacent to the passageway, are
44.8%, as disclosed in U.S. Pat. No. 4,160,020. 10 pressed into a solid possessing dimension that corresponds to
The osmotic agent present in the pump, which may be used the internal dimensions of the area of the compartment the
when the drug itself is not osmotically active, are osmotically agent will occupy, or the agent and other ingredients and a
effective compounds soluble in the fluid that enters the solvent are mixed into a solid or semisolid form by conven
device, and exhibits an osmotic pressure gradient across the tional methods such as ballmilling, calendaring, stirring or
semipermeable wall against the exterior fluid. Osmotically 15 rollmilling, and then pressed into a preselected shape. Next, a
effective osmagents useful for the present purpose include, layer of a hydrophilic polymer is placed in contact with the
but are not limited to, magnesium sulfate, calcium Sulfate, layer of agent in a like manner, and the two layers Surrounded
magnesium chloride, sodium chloride, lithium chloride, with a semipermeable wall. The layering of agent formulation
potassium Sulfate, sodium carbonate, sodium sulfite, lithium and hydrophilic polymer can be fabricated by conventional
Sulfate, potassium chloride, sodium sulfate, d-mannitol, urea, two-layer press techniques. The wall can be applied by mold
Sorbitol, inositol, raffinose, Sucrose, glucose, hydrophilic ing, spraying or dipping the pressed shapes into a wall form
polymers such as cellulose polymers, mixtures thereof, and ing material. Another technique that can be use for applying
the like. The osmagent is usually present in an excess amount, the wall is the air Suspension procedure. This procedure con
and it can be in any physical form, Such as particle, powder, sists of Suspending and tumbling the pressed agent and dry
granule, and the like. The osmotic pressure in atmospheres of 25 hydrophilic polymer in a current of air and a wall forming
the osmagents suitable for the disclosure will be greater than composition until the wall is applied to the agent-hydrophilic
Zero and generally up to about 500 atm, or higher. polymer composite. The air Suspension procedure is
The expandable driving member is typically a swellable, described in U.S. Pat. No. 2,799,241; J. Am. Pharm. Assoc.,
hydrophilic polymer which interacts with water and aqueous Vol. 48, pp. 451–459. (1979). Other standard manufacturing
biological fluids and Swells—or expands to an equilibrium 30 procedures are described in Modern Plastics Encyclopedia,
state. The polymers exhibit the ability to swell in water and Vol. 46, pp. 62-70 (1969); and in Pharmaceutical Sciences, by
retain a significant portion of the imbibed water within the Remington, Fourteenth Edition, pp. 1626-1678 (1970), pub
polymer structure. The polymers Swell or expand to a very lished by Mack Publishing Company, Easton, Pa.
high degree, usually exhibiting a 2 to 50 fold Volume increase. Reservoir Polymeric Systems
The polymers can be noncross-linked or cross-linked. The 35 Reservoir systems are well known in the art. This technol
swellable, hydrophilic polymers can be lightly cross-linked, ogy is also commonly referred to as microencapsulation, bead
Such cross-links being formed by covalent ionic bonds or technology or coated tablets. Small particles of the drug are
hydrogen bonds. The polymers can be of plant, animal or encapsulated with pharmaceutically acceptable polymer(s).
synthetic origin. Hydrophilic polymers suitable for use in the This polymer, and its relative quantity, offers a predetermined
present disclosure include, but are not limited to, poly(hy 40 resistance to drug diffusion from the reservoir to the gas
droxyalkyl methacrylate) having a molecular weight of from trointestinal tract. Thus, the drug is gradually released from
30,000 to 5,000,000; kappa carrageenan, polyvinylpyrroli the beads into the gastrointestinal tract and provides the
done having molecular weight of from 10,000 to 360,000; desired controlled release of (1) a xanthine oxidoreductase
anionic and cationic hydrogels; polyelectrolyte complexes; inhibitor, Such as, 2-3-cyano-4-2(2-methylpropoxy)phe
poly(vinyl alcohol) having a low acetate residual, cross 45 nyl-4-methylthiazole-5-carboxylic acid or (2) a xanthine
linked with glyoxal, formaldehyde, or glutaraldehyde and oxidase inhibitor, Such as allopurinol and oxypurinol.
having a degree of polymerization from 200 to 30,000; a These dosage forms are well known in the art. U.S. Pat.
mixture of methylcellulose; cross-linked agar and carboxym Nos. 5.286,497 and 5,737,320, both of which are hereby
ethyl cellulose; a water insoluble, water swellable copolymer incorporated by reference, describe such formulations and
produced by forming a dispersion of finely divided copoly 50 their methods of production. One skilled in the art, taking into
mer of maleic anhydride with styrene, ethylene, propylene, account the teachings in this application as well as those of the
butylene or isobutylene cross-linked with from 0.001 to about 320 and 497 patents, could produce a bead or pellet based
0.5 moles of saturated cross-linking agent per mole of maleic dosage form matching the pharmacokinetic profile described
anhydride in copolymer; water swellable polymers of N-vi above.
nyl lactams, and the like. 55 As a general guideline, however, a bead is formed with an
The expression “orifice' as used herein comprises means inert core sphere and a Xanthine oxidoreductase inhibito—or
and methods Suitable for releasing the drug from the system. Xanthine oxidase inhibitor, optionally in association with
The expression includes one or more apertures or—orifices conventional excipients. The core of said beads may option
which have been bored through the semipermeable mem ally comprise any materials commonly used in pharmaceutics
brane by mechanical procedures. Alternatively, it may be 60 and should be selected on the basis of compatibility with the
formed by incorporating an erodible element, such as a gela active drug and the physicochemical properties of the beads.
tinplug, in the semipermeable membrane. In cases where the Additional components may include, but are not limited to
semipermeable membrane is sufficiently permeable to the binders, disintegration agents, filling agents, Surfactants,
passage of drug, the pores in the membrane may be sufficient solubilizers, stabilizers, and the like. Additionally, this core is
to release the agent/drug in therapeutically effective amounts. 65 then coated with one, or more, pharmaceutically acceptable
In Such cases, the expression “passageway' refers to the pores polymers capable of imparting varied release characteristics.
within the membrane wall even though no bore or other The central core may be prepared by a number of techniques
US 9.248,119 B2
33 34
known in the art. Typically, the Xanthine oxidoreductase (methacrylic acid, methyl methacrylate), poly(methacrylic
inhibitor or Xanthine oxidase inhibitor is bound to an inert acid, ethylacrylate), poly(methyl acrylate, methyl methacry
core with a conventional binding agent. The inert core typi late, methacrylic acid), poly(ethylacrylate, methylmethacry
cally comprises a starch, Sugar or microcrystalline cellulose. late, trimethylammonioethyl methacrylate chloride), poly
One of skill in the art will appreciate that a variety of sugars (ethylacrylate, methyl methacrylate), poly(methacrylic acid,
may be incorporated into the bead core, and that compatibility ethylacrylate), type A methacrylic acid copolymer, type B
issues should be considered when selecting the appropriate methacrylic acid copolymer, type C methacrylic acid copoly
sugar. Before the xanthine oxidoreductase inhibitor or xan mer, methacrylic acid copolymer dispersion, aqueous acrylic
thine oxidase inhibitor is bound to the inert core, it is typically polymer dispersion, (EUDRAGITR) compounds),
blended with conventional excipients to expedite its handling 10 OPADRYR) and the like, and mixtures thereof. In one aspect,
and to improve the properties of the final dosage form. These the immediate release polymer comprises hydroxypropyl
excipients are identical to those described above for the methylcellulose.
matrix systems. The quantity of these excipients can vary The polymeric layer that encapsulates the core becomes
widely, but will generally be used in conventional amounts. soluble and begins releasing the active drug immediately after
The inert core is then produced by utilizing a binding agent to 15 ingestion by the patient. Under certain circumstances it may
attach the powdered xanthine oxidoreductase inhibitor or be beneficial to coat the core with a polymer, Sealing the core
Xanthine oxidase inhibitor blend to the solid carrier. This can material and providing easier coating of the core.
be accomplished by means known in the art for producing The beads of the current disclosure may also comprise an
pharmaceutical beads. Suitable means include utilization of a enteric coating layer that is applied onto the cores with or
conventional coating pan, a fluid-bed processor, extrusion without seal coating by conventional coating techniques,
spheronization, or a rotogranulator. The production of these Such as pan coating or fluid bed coating using solutions of
central cores is described in more detail in Pharmaceutical polymers in water or Suitable organic solvents or by using
Pelletization Technology, ed. I. Ghebre-Sellassie, Marcel aqueous polymer dispersions. All commercially available
Dekker, Inc. New York, N.Y. (1989) which is hereby incor pH-sensitive polymers are included in the scope of the dis
porated by reference. 25 closure. With enteric coating layers, the pharmaceutical
The second major component of the beads is the polymeric active is not released in the acidic stomach environment of
coating. As noted above, the polymeric coating is responsible approximately below pH 4.5, but not limited to this value. The
for giving the beads their extended release characteristics. pharmaceutical active is typically released when the pH-sen
The polymeric coating may be applied to the central core sitive layer dissolves at the greater pH. Suitable examples of
using methods and techniques known in the art. Examples of 30 delayed release enteric polymers include, but are not limited
suitable coating devices include, but are not limited to, fluid to cellulose acetate phthalate, cellulose acetate trimellitate,
bed coaters, pan coaters, etc. The application techniques are hydroxypropyl methylcellulose phthalate, polyvinyl acetate
described in more detail in: 1) Aqueous polymeric coatings phthalate, carboxymethylethylcellulose, co-polymerized
for pharmaceutical dosage forms, ed. J. W. McGinity, Marcel methacrylic acid/methacrylic acid methyl esters such as, for
Dekker, Inc. New York, N.Y. (1997); and 2) Pharmaceutical 35 instance, materials known under the trade name
Dosage Forms: Tablets Vol. 3. ed. H. A. Lieberman, L. Lach EUDRAGITR) L12.5, L100, EUDRAGITR) S12.5, S100, or
man and J. B. Schwartz, Marcel Dekker, Inc. New York, N.Y. similar compounds used to obtain enteric coatings. Co-poly
pp. 77-287. (1990), the contents of which are hereby incor merized methacrylic acid/methacrylic acid methyl esters gen
porated by reference. erally comprise three Subclasses of compound: methacrylic
The polymer may be incorporated into the beads by means 40 acid copolymer type A, methacrylic acid copolymer type B,
of a layer attached to the pharmaceutical active, distal to the and methacrylic acid copolymer type C. The various types of
core and also may be provided in multiple layers, with each copolymers represent compounds with varying ratios of
layer incorporating distinct polymers, providing for varied methacrylic acid to methacrylic acid methyl ester. Accord
release characteristics in each layer. One such polymeric ingly, methacrylic acid copolymer type A has a ratio of meth
layer includes a modified release polymeric layer. The phar 45 acrylic acid to methacrylic acid methyl ester of approxi
maceutical active may be released from the modified release mately 1:1, type B has a ratio of approximately 1:2, and type
polymeric layer, such that the active particles are released as C has a ratio similar to type A, but may incorporate additional
the polymer becomes soluble with the surrounding environ components, such as Surfactants. Aqueous colloidal polymer
ment. Suitable examples of the immediate release polymers dispersions or re-dispersions can be also applied, including,
that may be used for the immediate release polymeric layer 50 for example, the polymers sold under the trade name
include, but are not limited to ethylcellulose, hydroxypropyl EUDRAGITR L 30D-55, EUDRAGITR) L100-55,
methylcellulose, hydroxylpropyl cellulose, cellulose acetate, EUDRAGITR) S100, EUDRAGITR) preparation 4110D
cellulose propionate (lower, medium or higher molecular (Rohm Pharma); EUDAGRITO FS 30D: AQUATERIC(R),
weight), cellulose acetate propionate, cellulose acetate AQUACOATR CPD 30 (FMC); KOLLICOAT MAER. 3OD
butyrate, cellulose acetate phthalate, cellulose triacetate, poly 55 and 30DP (BASF); and EASTACRYL(R) 30D (Eastman
(methyl methacrylate), poly(ethyl methacrylate), poly(butyl Chemical). In one aspect, the delayed release enteric polymer
methacrylate), poly(isobutyl methacrylate), poly(hexyl comprises methacrylic acid copolymer type A. In still yet
methacrylate), poly(isodecyl methacrylate), poly(lauryl another aspect, the delayed release enteric polymer com
methacrylate), poly(phenyl methacrylate), poly(methyl acry prises a mixture of methacrylic acid copolymer type A and
late), poly(isopropyl acrylate), poly(isobutyl acrylate), poly 60 methacrylic acid copolymer type B.
(octadecyl acrylate), poly(ethylene), poly(ethylene) low den One skilled in the art will appreciate that additional com
sity, poly(ethylene) high density, poly(propylene), poly ponents may be added to the delayed release polymers with
(ethylene oxide), poly(ethylene terephthalate), poly(vinyl out departing from the Scope of the disclosure. For instance,
isobutyl ether), poly(vinyl acetate), poly(vinyl chloride), a plasticizer may be added to the delayed release enteric
polyurethane, ethylcellulose aqueous dispersions (AQUA 65 polymers to improve the physical characteristics of the
COATR), SURELEASE(R), poly(butyl methacrylate, (2-dim delayed release polymeric layer. Non-limiting examples of
ethylaminoethyl) methacrylate, methyl methacrylate), poly plasticizers include triethyl citrate, acetyl triethyl citrate,
US 9.248,119 B2
35 36
tributyl citrate, acetyl tributyl citrate, trihexyl citrate, acetyl combination of ethylcellulose and hydroxypropyl methylcel
trihexylcitrate, trioctyl citrate, acetyl trioctyl citrate, butyryl lulose in a ratio of ethylcellulose to hydroxypropyl methyl
trihexylcitrate, acetylbutyryl trihexyl citrate, trimethyl cit cellulose ranging from about 0.1 to about 10, from about 0.2
rate, acetylated monoglycerides, and alkyl Sulphonic acid to about 5, from about 0.5 to about 3, and from about 1 to
phenyl esters. In yet another aspect, the plasticizer comprises about 2. In still yet another aspect, the controlled release
triethylcitrate. polymer comprises a combination of ethylcellulose aqueous
Furthermore, the enteric polymers used in this disclosure dispersion and hydroxypropyl methylcellulose in a ratio of
can be modified by mixing with other known coating products ethylcellulose aqueous dispersion to hydroxypropyl methyl
that are not pH sensitive. Examples of Such coating products cellulose ranging from about 0.1 to about 10, from about 0.1
include the neutral methacrylic acid esters with a small por 10 to about 5, from about 0.5 to about 4, and from about 1.5 to
tion of trimethylammonioethyl methacrylate chloride, sold about 3.
currently under the trade names EUDRAGITR) and The pharmaceutical composition of the current disclosure,
EUDRAGITR) RL: a neutral ester dispersion without any in one aspect, comprises one or more types of bead, can be
functional groups, sold under the trade names EUDRAGITR) incorporated in multiple dosage forms such as capsules, pills,
NE30D and EUDRAGITR NE30; and other pH independent 15 and tablets. Capsules, including hard gelatin capsules, may be
coating products. produced according to methods known in the art. Generally,
It is also within the scope of this disclosure that an addi the capsules may incorporate the beads discussed herein, and
tional modifying layer may be added on top of the enteric may optionally incorporate additional excipients, as previ
coating layer. This modifying layer can include a water pen ously described. The pharmaceutical composition may also
etration barrier layer (semipermeable polymer) which can be be incorporated into a tablet. Generally, this process involves
Successively coated after the enteric coating to reduce the incorporation of the beads into a tablet matrix, as previously
water penetration rate through the enteric coating layer and described. One skilled in the art will appreciate that additional
thus increase the lag time of the drug release. Controlled components may be added to the formulation to impart the
release coatings commonly knownto one skilled in the art can desired physical characteristics, without departing from the
be used for this purpose by conventional coating techniques 25 Scope of the disclosure.
Such as pan coating or fluid bed coating using Solutions of Types of Beads
polymers in water or Suitable organic solvents or by using There are many types ofbead formulations that are encom
aqueous polymer dispersions. For example, the following passed within the scope of the current disclosure, incorporat
non-limiting list of controlled release polymers may be used ing various polymers in accordance with the polymers previ
in the current disclosure: cellulose acetate, cellulose acetate 30 ously described. One skilled in the art may modify bead
butyrate, cellulose acetate propionate, ethylcellulose, formulations to impart specific chemical characteristics. In
hydroxypropyl methylcellulose, cellulose acetate, cellulose one aspect, the current disclosure describes four primary
propionate (lower, medium or higher molecular weight), cel types of beads. Namely, the four types of beads may be
lulose acetate propionate, cellulose acetate butyrate, cellulose described as immediate release beads, delayed release beads,
acetate phthalate, cellulose triacetate, poly(methyl methacry 35 controlled release beads, and delayed-controlled release
late), poly(ethyl methacrylate), poly(butyl methacrylate), beads. The immediate release beads comprise a Xanthine
poly(isobutyl methacrylate), poly(hexyl methacrylate), poly oxidoreductase inhibitor or xanthine oxidase inhibitor lay
(isodecyl methacrylate), poly(lauryl methacrylate), poly ered on an inert core Such as Sugar spheres or microcrystalline
(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl cellulose spheres by means of a suitable polymeric binder.
acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate), 40 The polymeric binder functions to create a sealcoat around
poly(ethylene), poly(ethylene) low density, poly(ethylene) the inert core material, improving the friability of the inert
high density, poly(propylene), poly(ethylene oxide), poly core. The polymeric binder may comprise any of the imme
(ethylene terephthalate), poly(vinyl isobutyl ether), poly(vi diate release polymers previously described. In one aspect,
nyl acetate), poly(vinyl chloride), polyurethane, ethylcellu the polymeric binder comprises hydroxypropyl methylcellu
lose aqueous dispersions such as AQUACOATR) and 45 lose. For the purposes of the various bead compositions
SURELEASE(R), poly(butyl methacrylate, (2-dimethylami described herein, the polymeric binder component will com
noethyl) methacrylate, methyl methacrylate), poly(meth prise the same material as the immediate release polymer, and
acrylic acid, methyl methacrylate), poly(methacrylic acid, the percentage composition of immediate release polymer
ethylacrylate), poly(methyl acrylate, methyl methacrylate, will include the immediate release polymer used in the imme
methacrylic acid), poly(ethylacrylate, methylmethacrylate, 50 diate release layer Surrounding the inert core, as well as the
trimethylammonioethyl methacrylate chloride), poly(ethy polymeric binder providing the sealcoat for the inert core.
lacrylate, methyl methacrylate), poly(methacrylic acid, ethy The immediate release bead typically comprises from
lacrylate), type A methacrylic acid copolymer, type B meth about 5% to about 55% (w/w) xanthine oxidoreductase
acrylic acid copolymer, type C methacrylic acid copolymer, inhibitor or Xanthine oxidase inhibitor, from about 20% to
methacrylic acid copolymer dispersion, aqueous acrylic 55 about 80% (w/w) inert core, and from about 1% to about 40%
polymer dispersion, (EUDRAGITR) compounds), (w/w) immediate release polymer. In one aspect, the imme
OPADRYR), fatty acids and their esters, waxes, Zein, and diate release bead typically comprises from about 25% to
aqueous polymer dispersions such as EUDRAGITR) RS and about 35% (w/w) xanthine oxidoreductase inhibitor or Xan
RL 30D, EUDRAGITR NE 30D, cellulose acetate latex. The thine oxidase inhibitor, from about 40% to about 60% (w/w)
combination of above polymers and hydrophilic polymers 60 inert core, and from about 10% to about 20% (w/w) immedi
Such as hydroxyethyl cellulose, hydroxypropyl cellulose ate release polymer. In another aspect the Xanthine oxi
(KLUCEL(R), Hercules Corp.), hydroxypropyl methylcellu doreductase inhibitor or xanthine oxidase inhibitor comprises
lose (METHOCEL(R), Dow Chemical Corp.), and polyvi from about 29% to about 34% (w/w) of the total composition,
nylpyrrolidone may also be incorporated. In one aspect, the the inert core comprises from about 50% to about 55% (w/w)
controlled release polymer comprises ethylcellulose, hydrox 65 of the total composition, and the immediate release polymer
ypropyl methylcellulose, and combinations thereof. In yet comprises from about 14% to about 18% (w/w) of the total
another aspect, the controlled release polymer comprises a composition.
US 9.248,119 B2
37 38
An additional type of bead that is contemplated by the about 43% (w/w) of an inert core, from about 12% to about
current disclosure is a delayed release bead. Delayed release 14% (w/w) immediate release polymer, and from about 17%
beads are coated beads obtained by coating immediate release to about 19% (w/w) of one or more delayed release enteric
beads with a delayed release enteric polymer either in an polymers. In a further iteration of this example, the delayed
aqueous dispersion or in an organic Solvent. These polymers 5 release pH 6.8 bead further comprises approximately 1% to
have pH dependent solubility depending on the functional approximately 3% (w/w) plasticizer.
groups on the polymer. For a delayed release bead coated with An additional type of bead that is contemplated by the
Suitable amount of delayed release enteric polymer, drug current disclosure is a controlled release bead. Controlled
release will not occur in a medium unless medium pH is above release beads are coated beads obtained by coating immediate
the pH at which the polymer dissolves. The delayed release 10 release beads with a controlled release polymer according to
enteric polymers of the current disclosure generally become methods currently known in the art. Generally, the controlled
soluble when the bead is exposed to a pH level generally less release beads incorporate one or more polymers that decrease
acidic than the environment of the stomach. Specifically, the the release rate of the drug from the bead, so that the drug is
delayed release polymer may become soluble at pH levels released over an extended period of time. Controlled release
greater than or equal to 4.5; 4.6; 4.7; 4.8; 4.9; 5.0; 5.1; 5.2; 5.3: 15 beads differ from delayed release beads in that the release
5.4; 5.5; 5.6; 5.7; 5.8; 5.9; 6.0; 6.1: 6.2; 6.3; 6.4; 6.5; 6.6; 6.7: from controlled release beads is continuous after exposure to
6.8; 6.9; 7.0; 7.1: 7.2: 7.3; 7.4; 7.5; 7.6: 7.7; 7.8; 7.9; 8.0; 8.1; dissolution medium, over an extended period of time,
8.2; 8.3; 8.4; 8.5; 8.6; 8.7; 8.8; 8.9; 9.0; 9.1; 9.2: 9.3; 9.4:9.5; whereas release from delayed release beads is very rapid once
9.6; 9.7; 9.8; 9.9; and 10.0. In one aspect, the delayed release the beads are exposed to a pH above which the delayed release
polymer becomes soluble at pH levels greater than or equal to enteric polymer is soluble. In general, the composition of the
5.5, 6.0, and 6.8. controlled release polymeric layer may be modified such that
The composition of the immediate release component of release is possible over periods of time ranging from approxi
the delayed release bead is the same as previously described, mately 1 hour to approximately 24 hours. Specifically, the
and generally comprises a Xanthine oxidoreductase inhibitor controlled release formulation may release active drug over a
or Xanthine oxidase inhibitor, an inert core, and an immediate 25 period of 1,2,3,4,5,6,7,8,9, 10, 11, 12, 13, 14, 15, 16, 17,
release polymer, as previously described. The delayed release 18, 19, 20, 21, 22, 23, and 24 hours.
bead additionally comprises a pH-sensitive enteric polymer, In one embodiment of the current disclosure, the controlled
as previously described. A non-limiting example of a delayed release beads incorporate a composition capable of releasing
release bead is one in which the bead has a solubility at pH the active compound over a period of time ranging from
levels greater than or equal to 6.0. This delayed release pH 6.0 30 approximately four hours to approximately six hours. The
bead typically comprises a Xanthine oxidoreductase inhibitor controlled release beads of this embodiment generally com
or Xanthine oxidase inhibitor from about 5% to about 50% prise from about 5% to about 40% (w/w) xanthine oxi
(w/w) of the overall delayed release bead, an inert core com doreductase inhibitor or Xanthine oxidase inhibitor, from
prising from about 20% to about 70% (w/w) of the total about 20% to about 50% (w/w) of an inert core, from about
delayed release bead, an immediate release polymer compris 35 5% to about 25% (w/w) immediate release polymer, from
ing from about 1% to about 35% (w/w) of the total delayed about 10% to about 50% (w/w) controlled release polymer. In
release bead, and a delayed release enteric polymer in an one aspect, the controlled release beads comprise from about
amount ranging from about 1% to about 35% (w/w) of the 20% to about 24% (w/w) xanthine oxidoreductase inhibitor
total delayed release bead. In one aspect, the delayed release or Xanthine oxidase inhibitor, from about 30% to about 40%
bead comprises from about 20% to about 30% (w/w)xanthine 40 (w/w) of an inert core, from about 9% to about 13% (w/w)
oxidoreductase inhibitor or Xanthine oxidase inhibitor, from immediate release polymer, from about 25% to about 35%
about 40% to about 50% (w/w) of an inert core, from about (w/w) controlled release polymer. In one aspect, the con
10% to about 16% (w/w) immediate release polymer, and trolled release beads comprise from about 25% to about 35%
from about 13% to about 20% (w/w) delayed release polymer. (w/w) xanthine oxidoreductase inhibitor or xanthine oxidase
In a furtheriteration of this example, the delayed release pH 45 inhibitor, from about 40% to about 60% (w/w) of an inert
6.0 bead further comprises approximately 1% to approxi core, from about 12% to about 18% (w/w) immediate release
mately 3% (w/w) plasticizer. polymer, from about 3% to about 9% (w/w) controlled release
Another non-limiting example of a delayed release bead is polymer.
one in which the bead has a solubility at pH levels greater than In an additional embodiment, the controlled release beads
or equal to 6.8. This delayed release pH 6.8 bead typically 50 incorporate a composition capable of releasing the active
comprises a Xanthine oxidoreductase inhibitor or Xanthine compound over a period of time ranging from approximately
oxidase inhibitor from about 5% to about 50% (w/w) of the ten hours to approximately twelve hours. The controlled
overall delayed release bead, an inert core comprising from release beads of this embodiment generally comprise from
about 20% to about 70% (w/w) of the total delayed release about 10% to about 50% (w/w) xanthine oxidoreductase
bead, an immediate release polymer comprising from about 55 inhibitor or Xanthine oxidase inhibitor, from about 30% to
1% to about 35% (w/w) of the total delayed release bead, and about 70% (w/w) of an inert core, from about 5% to about
a delayed release enteric polymer in an amount ranging from 25% (w/w) immediate release polymer, and from about 1% to
about 1% to about 35% (w/w) of the total delayed release about 15% (w/w) controlled release polymer. In one aspect,
bead. In one aspect, the delayed release pH 6.8 bead com the controlled release beads comprise from about 25% to
prises from about 20% to about 30% (w/w) xanthine oxi 60 about 35% (w/w) xanthine oxidoreductase inhibitor or Xan
doreductase inhibitor or Xanthine oxidase inhibitor, from thine oxidase inhibitor, from about 40% to about 60% (w/w)
about 40% to about 50% (w/w) of an inert core, from about of an inert core, from about 12% to about 18% (w/w) imme
10% to about 16% (w/w) immediate release polymer, and diate release polymer, and from about 3% to about 9% (w/w)
from about 13% to about 20% (w/w) delayed release polymer. controlled release polymer. In one aspect, the controlled
In one aspect, the delayed release pH 6.8 bead comprises from 65 release beads comprise from about 28% to about 31% (w/w)
about 23% to about 27% (w/w) xanthine oxidoreductase Xanthine oxidoreductase inhibitor or Xanthine oxidase inhibi
inhibitor or Xanthine oxidase inhibitor, from about 40.5% to tor, from about 47% to about 51% (w/w) of an inert core, from
US 9.248,119 B2
39 40
about 14% to about 16% (w/w) immediate release polymer, limiting methods of manufacturing the beads include fluid
and from about 5% to about 7% (w/w) controlled release bed processing, centrifugal granulation, extrusion-spheroni
polymer. Zation, high shear granulation, melt extrusion, and solution or
A further type of bead that is contemplated by the current Suspension layering. In a fluid bed process, the immediate
disclosure is a delayed-controlled release bead. The delayed release polymer is dissolved in water and micronized drug is
controlled release bead combines the attributes of the delayed Suspended in the immediate release polymer Solution. This
release beads described above and the controlled release Suspension is then sprayed onto inert spherical Support beads
beads described above, with the goal of delaying drug release Such as Sugar spheres or microcrystalline cellulose spheres.
until the beads are exposed to a pH greater than the pH at Alternately, non-micronized drug can be suspended in the
which the polymer dissolves, and Subsequently prolonging 10 immediate release polymer Solution and the Suspension can
drug release over an extended period of time. To illustrate, a be passed through a mill. In the centrifugal granulation pro
controlled release bead, administered alone, would typically cess, the inert beads are placed in the granulator on a rotating
release the active component over an extended period of time, disc at the bottom of the granulator. Micronized drug is intro
with release beginning immediately after ingestion. Delayed duced into the granulator and a solution of the immediate
release beads, administered alone, would not begin releasing 15 release polymer is sprayed at the same time. Extrusion and
the active component until the environment meets a minimum spheronization is another manufacturing method for imme
pH level. For instance, the pH level in the intestine is higher diate release beads, wherein the drug is mixed with dry
than that of the stomach, therefore, a delayed release bead excipients and wet-massed by addition of a binder Solution
may be designed to release the active component once it and extruded to form spaghetti-like strands. The extrudate is
reaches the pH level found in the intestine, without releasing then chopped and converted to dense spherical beads using a
any active component in regions where the pH level is lower, spheronizer. Another method of producing beads includes
Such as the stomach. However, once the pH level is triggered, high shear granulation. High shear granulation involves dry
drug release is typically rapid. mixing the active component and other components. Then the
In the instant embodiment, the delayed release polymeric mixture is wetted by addition of a binder solution in a high
layer encapsulates the controlled release polymeric layer, 25 shear-granulator/mixer. The granules are kneaded after wet
Such that the prolonged release of active component allowed ting by the combined action of mixing and milling. The
by the controlled release polymer will not begin until the resulting granules or pellets are Subsequently dried and
delayed-controlled release bead is exposed to a minimum pH sieved. An additional method comprises melt-extrusion or
level. Accordingly, one skilled in the art will appreciate that melt-granulation. This process generally involves melting a
the characteristics of the delayed-controlled release bead may 30 normally solid hydrophobic binder material, e.g. a wax or
be modified such that the delayed release polymer does not similar Substance, and incorporating a powdered drug
become soluble until the bead is exposed to pH levels gener therein. To obtain a controlled or extended release dosage
ally ranging from about 5 to about 10, as previously form, additional hydrophobic release materials, e.g. ethylcel
described. Furthermore, the delayed-controlled release beads lulose or a water-insoluble acrylic polymer, may be incorpo
may be designed to release the active component for a period 35 rated into the molten wax hydrophobic binder material. Fur
ranging from about one hour to about twenty-four hours, as ther, solution or Suspension layering involves a process
previously described. whereby an active component Solution or dispersion with or
In one embodiment, of the current disclosure, the delayed without a binder is sprayed onto starting seeds with a certain
controlled release bead incorporates a delayed release enteric particle size in a fluidized bed processor or other suitable
polymer with a solubility at pH levels greater than or equal to 40 equipment. The drug thus is coated on the Surface of the
6.8, and a controlled release polymer allowing for prolonged starting seeds. The drug-loaded pellets are dried for further
delivery of the active compound over four to six hours. The applications.
delayed-controlled release beads of this embodiment gener Pharmaceutical Dosage Forms
ally comprise from about 5% to about 35% (w/w) xanthine It will be understood by one skilled in the art that the
oxidoreductase inhibitor or Xanthine oxidase inhibitor, from 45 various types of beads described herein, with distinct active
about 20% to about 50% (w/w) of an inert core, from about release profiles may be combined in single or multiple phar
5% to about 20% (w/w) immediate release polymer, from maceutical dosage forms to provide pulsed drug delivery of
about 5% to about 20% (w/w) controlled release polymer, and the Xanthine oxidoreductase inhibitor or Xanthine oxidase
from about 5% to about 35% (w/w) delayed release enteric inhibitors described herein. Various combinations of imme
polymer. In one aspect, the delayed-controlled release beads 50 diate release beads, delayed release beads, controlled release
comprise from about 15% to about 25% (w/w) xanthine oxi beads, and delayed-controlled release beads may be used to
doreductase inhibitor or Xanthine oxidase inhibitor, from impart distinct release profiles. It should be understood that
about 30% to about 40% (w/w) of an inert core, from about any potential combination of immediate release, delayed
8% to about 14% (w/w) immediate release polymer, from release, controlled release, and delayed-controlled release
about 8% to about 15% (w/w) controlled release polymer, and 55 beads for the distribution of the Xanthine oxidoreductase
from about 13% to about 22% (w/w) delayed release enteric inhibitor or Xanthine oxidase inhibitors described herein are
polymer. In another aspect, the delayed-controlled release within the scope of the current disclosure.
bead comprises from about 20% to about 23% (w/w)xanthine In one embodiment, the current disclosure encompasses a
oxidoreductase inhibitor or Xanthine oxidase inhibitor, from single pharmaceutical composition that incorporates both
about 34% to about 37% (w/w) of an inert core, from about 60 immediate release beads and delayed release beads with a
10% to about 12% (w/w) immediate release polymer, from solubility at pH levels greater than or equal to 6.8. The phar
about 11% to about 13% (w/w) controlled release polymer, maceutical composition of this embodiment comprises
and from about 17% to about 20% (w/w) delayed release immediate release Xanthine oxidoreductase inhibitor or Xan
enteric polymer. thine oxidase inhibitor beads in an amount ranging from
One skilled in the art will appreciate that the various modi 65 approximately 20% to approximately 40% (w/w) of the total
fied release beads of the current disclosure may be manufac composition weight and pH 6.8 delayed release Xanthine
tured by any means known in the art. Examples of non oxidoreductase inhibitor or Xanthine oxidase inhibitor beads
US 9.248,119 B2
41 42
in an amount ranging from approximately 60% to approxi The delayed release pH 6.0 beads comprise: a) an inert
mately 80% (w/w) of the total composition weight. core in an amount ranging from about 40.5% to about 43%
The immediate release beads comprise: a.) an inert core in (w/w) of the weight of the delayed release pH 6.0 bead; b.) an
an amount ranging from about 50% to about 55% (w/w) of the immediate release layer encapsulating the inert core compris
weight of the immediate release bead; and b.) an immediate ing a mixture of Xanthine oxidoreductase inhibitor or Xan
release layer that encapsulates the inert core comprising a thine oxidase inhibitor and hydroxypropyl methylcellulose in
mixture of xanthine oxidoreductase inhibitor or Xanthine oxi an amount ranging from about 35% to about 40% (w/w) of the
dase inhibitor and a binder such as hydroxypropyl methylcel weight of the delayed release pH 6.0 bead, the ratio of Xan
lulose or hydroxypropyl cellulose in an amount ranging from thine oxidoreductase inhibitor or Xanthine oxidase inhibitor
about 45% to about 50% (w/w) of the weight of the immediate 10 to hydroxypropyl methylcellulose ranging from about 1.5 to
release bead, the ratio of xanthine oxidoreductase inhibitor or about 3; c.) a delayed release pH 6.0 enteric polymer layer
xanthine oxidase inhibitor to hydroxypropyl methylcellulose encapsulating the immediate release layer comprising a
ranging from about 1.5 to about 3. An insoluble disintegrant delayed release enteric polymer in an amount ranging from
such as low substituted hydroxylpropyl cellulose (L-HPC) about 17% to about 19% (w/w) of the delayed release bead,
may be added to speed release of the active. 15 said delayed release pH 6.0 enteric polymer comprising
The pH 6.8 delayed release beads comprisea.) an inert core methacrylic acid copolymer type A, and d.) a plasticizer in an
in an amount ranging from about 40.5% to about 43% (w/w) amount ranging from about 1% to about 3% (w/w) of the
of the weight of the delayed release bead; b.) an immediate weight of the delayed-controlled release bead.
release layer encapsulating the inert core (as described Two types of beads, namely immediate release and pH 6.0
before) in an amount ranging from about 35% to about 40% delayed release beads can be combined together into a single
(w/w) of the weight of the delayed release bead, the ratio of bead for convenience by applying an immediate release coat
Xanthine oxidoreductase inhibitor or Xanthine oxidase inhibi on the top of a pH 6.0 delayed release coat. The resulting
tor to hydroxypropyl methylcellulose ranging from about 1.5 composition contains a.) an inert core in an amount ranging
to about 3; c.) a delayed release enteric polymer layer encap from about 25% to about 35% (w/w) of the weight of the
Sulating the immediate release layer comprising a delayed 25 combined immediate release-delayed release pH 6.0 bead; b.)
release enteric polymer in an amount ranging from about 17% an immediate release layer encapsulating the inert core com
to about 20% (w/w) of the delayed release bead, said delayed prising a mixture of Xanthine oxidoreductase inhibitor or
release enteric polymer comprising a mixture of methacrylic xanthine oxidase inhibitor and hydroxypropyl methylcellu
acid copolymer type A and methacrylic acid copolymer type lose in an amount ranging from about 25% to about 31%
B in a ratio ranging from approximately 0.1 to approximately 30 (w/w) of the weight of the combined bead; c.) a delayed
0.5; and d.) a plasticizer in an amount ranging from about 1% release pH 6.0 enteric polymer layer encapsulating the imme
to about 3% (w/w) of the weight of the delayed-controlled diate release layer comprising a delayed release enteric poly
release bead. In one aspect of the pharmaceutical composi mer in an amount ranging from about 11% to about 15%
tion, the xanthine oxidoreductase inhibitor comprises febux (w/w) of the delayed release bead, said delayed release pH 6.0
ostat and the plasticizer comprises triethylcitrate. In another 35 enteric polymer comprising methacrylic acid copolymer type
aspect, the Xanthine oxidase inhibitor in the pharmaceutical A.; d.) a plasticizer in an amount ranging from about 1% to
composition comprises allopurinol and the plasticizer com about 3% (w/w) of the weight of the delayed-controlled
prises triethyl citrate. release bead; and e.) an immediate release overcoat layer
In yet another embodiment of the current disclosure, the comprising a mixture of Xanthine oxidoreductase inhibitor or
pharmaceutical dosage form encompasses a single pharma 40 xanthine oxidase inhibitor and hydroxypropyl methylcellu
ceutical composition that incorporates immediate release lose in an amount ranging from about 23% to about 29%
beads, delayed release beads with solubility at pH levels (w/w) of the weight of the combined bead.
greater than or equal to 6.0, and delayed release beads with The delayed release pH 6.8 beads comprise: a) an inert
solubility at pH levels greater than or equal to 6.8. The phar core in an amount ranging from about 40.5% to about 43%
maceutical composition of this embodiment comprises 45 (w/w) of the weight of the delayed release bead; b.) an imme
immediate release Xanthine oxidoreductase inhibitor or Xan diate release layer encapsulating the inert core comprising a
thine oxidase inhibitor beads in an amount ranging from mixture of Xanthine oxidoreductase inhibitor or Xanthine oxi
approximately 25% to approximately 35% (w/w) of the total dase inhibitor and hydroxypropyl methylcellulose in an
composition weight, pH 6.0 delayed release Xanthine oxi amount ranging from about 35% to about 40% (w/w) of the
doreductase inhibitor or Xanthine oxidase inhibitor beads in 50 weight of the delayed release bead, the ratio of Xanthine
an amount ranging from approximately 25% to approxi oxidoreductase inhibitor or Xanthine oxidase inhibitor to
mately 35% (w/w) of the total composition weight, and pH hydroxypropyl methylcellulose ranging from about 1.5 to
6.8 delayed release xanthine oxidoreductase inhibitor or xan about 3; c.) a delayed release pH 6.8 enteric polymer layer
thine oxidase inhibitor beads in an amount ranging from encapsulating the immediate release layer comprising a
approximately 35% to approximately 45% (w/w) of the total 55 delayed release enteric polymer in an amount ranging from
composition weight. about 17% to about 20% (w/w) of the delayed release pH 6.8
The immediate release beads comprise: a.) an inert core in bead, said delayed release enteric polymer comprising a mix
an amount ranging from about 50% to about 55% (w/w) of the ture of methacrylic acid copolymer type A and methacrylic
weight of the immediate release bead; and b.) an immediate acid copolymer type B in a ratio ranging from approximately
release layer that encapsulates the inert core comprising a 60 0.1 to approximately 0.5; and d.) a plasticizer in an amount
mixture of xanthine oxidoreductase inhibitor or Xanthine oxi ranging from about 1% to about 3% (w/w) of the weight of the
dase inhibitor and hydroxypropyl methylcellulose in an delayed-controlled release bead, said plasticizer comprising
amount ranging from about 45% to about 50% (w/w) of the triethylcitrate. In another aspect of the pharmaceutical com
weight of the immediate release bead, the ratio of Xanthine position, the Xanthine oxidoreductase inhibitor comprises
oxidoreductase inhibitor or Xanthine oxidase inhibitor to 65 febuXostat, and the plasticizer comprises triethyl citrate. In
hydroxypropyl methylcellulose ranging from about 1.5 to still yet another aspect of this pharmaceutical composition,
about 3. the Xanthine oxidase inhibitor comprises allopurinol, and the
US 9.248,119 B2
43 44
plasticizer comprises triethylcitrate. In a further embodiment oxidase inhibitor to hydroxypropyl methylcellulose ranging
of the current disclosure, the pharmaceutical composition from about 1.5 to about 2.5; c.) a controlled release layer that
encompasses a single pharmaceutical composition that incor encapsulates the immediate release layer comprising a con
porates immediate release beads and delayed-controlled trolled release polymer in an amount ranging from about 10%
release beads, with the delayed release polymer having Solu 5 to about 14% (w/w) of the weight of the delayed-controlled
bility at a pH level of at least 6.8 and a controlled release rate release bead, said controlled release polymer comprising a
of approximately four to six hours. The pharmaceutical com mixture of ethylcellulose aqueous dispersion and hydrox
position of this embodiment comprises immediate release ypropyl methylcellulose, the ratio of ethylcellulose disper
Xanthine oxidoreductase inhibitor or Xanthine oxidase inhibi sion to hydroxypropyl methylcellulose ranging from about
tor beads in an amount ranging from approximately 20% to 10 1.5 to about 3; d.) a delayed release pH 6.8 layer that encap
approximately 40% (w/w) of the total composition weight Sulates the controlled release layer comprising a delayed
and delayed-controlled release Xanthine oxidoreductase release pH 6.8 polymer in an amount ranging from about
inhibitor or xanthine oxidase inhibitor beads having a solu 17.5% to about 20% (w/w) of the weight of the delayed
bility at pH levels greater than or equal to 6.8 and providing controlled release bead, said delayed release pH 6.8 polymer
prolonged release of Xanthine oxidoreductase inhibitor or 15 comprising a mixture of methacrylic acid copolymer type A
xanthine oxidase inhibitor over a period of about 4 hours to and methacrylic acid copolymer type B, the ratio of copoly
about 6 hours, in an amount ranging from approximately 60% mer type A to copolymer type B ranging from about 0.1 to
to approximately 80% (w/w) of the total composition weight. about 0.5; and e.) a plasticizer in an amount ranging from
For example, in one aspect, the pharmaceutical composition about 1% to about 3% (w/w) of the weight of the delayed
comprises immediate releaseXanthine oxidoreductase inhibi controlled release bead, said plasticizer comprising triethyl
tor or Xanthine oxidase inhibitor beads in an amount of citrate. In one aspect of the pharmaceutical composition, the
approximately 20% (w/w) of the total composition weight Xanthine oxidoreductase inhibitor comprises febuXostat, and
delayed release xanthine oxidoreductase inhibitor orxanthine the plasticizer comprises triethylcitrate. In yet another aspect
oxidase inhibitor beads that release at a pH of 6.8 in an of this pharmaceutical composition, the Xanthine oxidase
amount of approximately 80% (w/w) of the total composition 25 inhibitor comprises allopurinol, and the plasticizer comprises
weight. In yet another aspect, the pharmaceutical composi triethyl citrate.
tion comprises immediate release Xanthine oxidoreductase In still another embodiment of the current disclosure, the
inhibitor or Xanthine oxidase inhibitor beads in an amount of pharmaceutical composition encompasses a single pharma
approximately 25% (w/w) of the total composition weight ceutical composition that incorporates immediate release
delayed release xanthine oxidoreductase inhibitor orxanthine 30 beads and controlled release beads capable of active release
oxidase inhibitor beads that release at a pH of 6.8 in an over approximately ten to approximately twelve hours. The
amount of approximately 75% (w/w) of the total composition pharmaceutical composition of this embodiment generally
weight. In still yet another aspect, the pharmaceutical com comprises immediate releaseXanthine oxidoreductase inhibi
position comprises immediate release Xanthine oxidoreduc tor or Xanthine oxidase inhibitor beads in an amount ranging
tase inhibitor or Xanthine oxidase inhibitor beads in an 35 from approximately 10% to approximately 30% (w/w) of the
amount of approximately 30% (w/w) of the total composition total composition weight and controlled release Xanthine oxi
weight delayed release xanthine oxidoreductase inhibitor or doreductase inhibitor orxanthine oxidase inhibitor beads pro
xanthine oxidase inhibitor beads that release at a pH of 6.8 in viding prolonged release of Xanthine oxidoreductase inhibi
an amount of approximately 70% (w/w) of the total compo tor or xanthine oxidase inhibitor over a period of about 10
sition weight. In still yet another aspect, the pharmaceutical 40 hours to about 12 hours, in an amount ranging from approxi
composition comprises immediate release Xanthine oxi mately 70% to approximately 90% (w/w) of the total compo
doreductase inhibitor or Xanthine oxidase inhibitor beads in sition weight.
an amount of approximately 40% (w/w) of the total compo The immediate release beads comprise: a.) an inert core in
sition weight delayed release Xanthine oxidoreductase inhibi an amount ranging from about 50% to about 55% (w/w) of the
tor or xanthine oxidase inhibitor beads that release at a pH of 45 weight of the immediate release bead; and b.) an immediate
6.8 in an amount of approximately 60% (w/w) of the total release layer that encapsulates the inert core comprising a
composition weight. mixture of Xanthine oxidoreductase inhibitor or Xanthine oxi
The immediate release beads comprise: a.) an inert core in dase inhibitor and hydroxypropyl methylcellulose in an
an amount ranging from about 50% to about 55% (w/w) of the amount ranging from about 45% to about 50% (w/w) of the
weight of the immediate release bead; and b.) an immediate 50 weight of the immediate release bead, the ratio of Xanthine
release layer that encapsulates the inert core comprising a oxidoreductase inhibitor or Xanthine oxidase inhibitor to
mixture of xanthine oxidoreductase inhibitor or Xanthine oxi hydroxypropyl methylcellulose ranging from about 1.5 to
dase inhibitor and hydroxypropyl methylcellulose in an about 3.
amount ranging from about 45% to about 50% (w/w) of the The ten to twelve hour controlled release beads comprise:
weight of the immediate release bead, the ratio of Xanthine 55 a.) an inert core in an amount ranging from about 47% to
oxidoreductase inhibitor or Xanthine oxidase inhibitor to about 51% (w/w) of the weight of the controlled release bead;
hydroxypropyl methylcellulose ranging from about 1.5 to b.) an immediate release layer that encapsulates the inert core
about 3. comprising a mixture of Xanthine oxidoreductase inhibitor or
The delayed-controlled release beads comprise: a.) a inert xanthine oxidase inhibitor and hydroxypropyl methylcellu
core in an amount ranging from about 34% to about 37% 60 lose in an amount ranging from about 42% to about 48%
(w/w) of the weight of the delayed-controlled release bead; (w/w) of the weight of the controlled release bead, the ratio of
b.) an immediate release layer that encapsulates the inert core Xanthine oxidoreductase inhibitor or Xanthine oxidase inhibi
comprising a mixture of Xanthine oxidoreductase inhibitor or tor to hydroxypropyl methylcellulose ranging from about 1.5
xanthine oxidase inhibitor and hydroxypropyl methylcellu to about 2.5; and c.) a controlled release layer that encapsu
lose in an amount ranging from about 31% to about 34% 65 lates the immediate release layer comprising a controlled
(w/w) of the weight of the delayed-controlled release bead, release polymer, said controlled release polymer comprising
the ratio of Xanthine oxidoreductase inhibitor or Xanthine a mixture of ethylcellulose and hydroxypropyl methylcellu
US 9.248,119 B2
45 46
lose in an amount ranging from about 4% to about 8% (w/w) effective amount (or therapeutically effective amount) of the
of the weight of the controlled release bead, the ratio of dosage form of the present disclosure to treat said disease
ethylcellulose to hydroxypropyl methylcellulose ranging condition.
from about 1 to about 2. In yet another aspect of the pharma By way of example, and not of limitation, examples of the
ceutical composition, the Xanthine oxidoreductase inhibitor present disclosure will now be given.
comprises febuxoStat. In still yet another aspect of this phar
maceutical composition, the Xanthine oxidase inhibitor com Example 1
prises allopurinol.
One of skill in the art will appreciate that the various Methods for Obtaining a High Level of Xanthine
embodiments and dosage forms described herein may incor 10
Oxidase Inhibition Comparative Example
porate any dosage form known in the art. In one aspect, the
dosage forms including pills, tablets, and capsules. Further In healthy human Subjects, the majority of orally adminis
more, the pharmaceutical compositions may have total com tered febuxostat is absorbed in approximately 1 hour (i.e., the
position weights ranging from approximately 5 mg to t is approximately one hour). Moreover, the oral clearance
approximately 240 mg. In one aspect, the overall pharmaceu 15
offebuxostat from the plasma is approximately 7.3-15.1 L/hr,
tical composition weight (total weight) comprises approxi
mately 60 mg to approximately 100 mg. In another aspect, the with an effective half-life of approximately six (6) hours. In
total pharmaceutical composition weight of approximately fact, the drug is highly bound to albumin in the blood
80 mg. (~99.3%) and appears to have a low to medium apparent
Methods of Treatment volume of distribution of approximately 0.7 L/kg.
The dosage forms of the present disclosure can be used in Despite the relatively short effective half-life offebuxostat,
treating a variety of disease conditions. When the dosage clinical studies have shown that once a day dosing with imme
forms of the present disclosure contain febuXostat, such dos diate release formulations containing as little as 10 mg of
age forms can be used in treating conditions such as, but not febuxostat lowers concentrations of uric acid with minimal
limited to, gout, hyperuricemia, prostatitis, inflammatory 25 fluctuations in the serum uric acid concentrations in healthy
bowel disease, QT interval prolongation, myocardial infarc subjects. This is due to the nature of the pharmacokinetic
tion, cardiac hypertrophy, hypertension, nephrolithiasis, (PK) pharmacodynamic (PD) relationship between the
renal impairment, chronic kidney disease, metabolic Syn plasma concentrations offebuxostat (the PK marker) and the
drome (also referred to as “Syndrome X’ and includes, at serum uric acid concentrations (the PD marker). Once a day
least one of abdominal obesity, atherogenic dyslipidemia, 30 dosing with immediate release dosage forms containing as
insulin resistance, glucose intolerance, a prothrombotic state little as 10 mg febuxostat is expected to effectively decrease
or a proinflammatory state), diabetes, diabetic nephropathy, and maintain serum uric acid concentrations at a therapeutic
congestive heart failure, etc. Subjects Suffering from one of target (i.e. <6 mg/dL) for a gout patient with low serum uric
the above disease conditions and in need of treatment thereof acid concentration (i.e. 7 mg/dL); however, these dosage
can be administered an effective amount (or therapeutically 35 forms fail to maintain a high degree of inhibition, namely at
effective amount) of the dosage form of the present disclosure least 80% inhibition of the xanthine oxidase enzyme during
to treat said disease condition. the dosing interval (namely, 24 hours) even after multiple
When the dosage forms of the present disclosure contain dosings.
allopurinol or oxypurinol, Such dosage forms can be used in Table 1 below shows the results from a Phase 1, multiple
treating conditions such as, but not limited to, gout, hyperu 40 dose, randomized, placebo-controlled, double-blind, single
ricemia, prostatitis, inflammatory bowel disease, QT interval center, multiple-location dose escalation study involving
prolongation, myocardial infarction, cardiac hypertrophy, febuxostat. This study evaluated the pharmacokinetics and
hypertension, nephrolithiasis, renal impairment, chronic kid pharmacodynamics offebuXostat in healthy Subjects. In this
ney disease, metabolic syndrome (also referred to as “Syn study, oral doses of an immediate release dosage form of
drome X’ and includes, at least one of abdominal obesity, 45 febuXostat ranged from 10 mg once a day to 240 mg once a
atherogenic dyslipidemia, insulin resistance, glucose intoler day (hereinafter “OD) and as 30 mg twice a day (hereinafter
ance, a prothrombotic state or a proinflammatory state), dia “BID). Plasma, serum and urine samples were collected for
betes, diabetic nephropathy, congestive heart failure, etc. the determination offebuxostat and metabolites, uric acid,
Subjects suffering from one of the above disease conditions Xanthine and hypoxanthine concentrations. Samples were
and in need of treatment thereof can be administered an analyzed by high performance liquid chromatography.
TABLE 1.

Pharmacokinetics and Pharmacodynamics of Febuxostat Administration


at Multiple Doses with Multiple Release Profiles
t Cmax AUC t1/2- V/F CIVF
Dose (h) (Lig/mL) (Ligh/mL) (h) (L) (Lib) C/D AUC/D
10 mg QD
Day 1 O.99 O.3362 O.7269 1.5 (1.3) 38.2 15.12 0.0336 0.0727
Day 14 O.70 O.3995 0.9505 3.0 (2.0) 42.7 11.39 0.0399 0.0950
20 mg QD

Day 1 1.06 11123 2.1816 3.2 (2.6), 29.2 10.00 0.0556 0.1091
Day 14 O.89 O.9342 2.112S 4.7 (3.8) 33.3 10.01 0.0467 0.1056
US 9.248,119 B2
47 48
TABLE 1-continued
Pharmacokinetics and Pharmacodynamics of Febuxostat Administration
at Multiple Doses with Multiple Release Profiles
tnax Cmax AUC t1/2. V/F CI/F
Dose (h) (Ig/mL) (Ig himL) (h) (L) (Lb) C/D AUC/D
30 mg QD
Day O.72 1.1.192 2.5469 9.2 (4.6) 750 12.37
Day 14 O.89 1.2835 2.5681 6.7 (5.7) 62.7 12.19
40 mg QD
Day 144 15282 3.977O 4.2 (3.8) 48.7 12.60
Day 14 1.19 18221 4.2998 0.3 (6.3) 49.S 10.63
50 mg QD
Day O.78 1.9697 4.4O73 5.0 (4.5) 43.2 12.38
Day 14 1.14 1.7917 4.3785 0.1 (6.7) 59.1 12.30
70 mg QD
Day 1.OO 3.08.19 6.9335 5.0 (4.7) 41.6 11.21
Day 14 110 2.6899 6.94.89 2.5 (8.5) S4.1 10.95
90 mg QD
Day O.9S 3.4806 9.0927 9.3 (6.8) 56.7 11.68
Day 14 O.9S 4.0589 9.6467 4.6 (10.0) 63.7 11.17
20 mg QD
Day 1.OO 44720 11.3131 1.4 (9.1) 57.8 11.09
Day 14 1.11 S.3076 11.9599 8.2 (11.9) 55.1 10.47
60 mg QD
Day 0.75 7.2978 20.7463 0.7 (9.8) 40.7 8.22
Day 14 O.80 8.7711 22.2821 1.8 (9.5) 36.1 7.82
80 mg QD
Day 1.07 8.3986 25.5887 23.6 (11.0) 54.6 7.75
Day 14 1.00 8.0488 23.9545 20.8 (15.8) 45.6 8.07
240 mg QD
Day 1.06 8.3858 28.2692 12.7 (10.2) 54.6 9.53
Day 14 O.94 11.2630 34.9763 9.9 (8.1) 31.3 7.28
30 mg BID
Day O.90 13091 2.8169 4.0 (3.8) 34.4 11.30
Day 14 (AM) O.7O 14882 2.9146 4.9 (4.8) 41.1 10.81
Day 14 (PM) 1.75 0.8986 3.3083 11.1 (5.8) 61.0 9.88
AUC refers to AUCAUC24 and AUC2 for Day 1 (QD & BID), Day 14 (QID), and Day 14 (BID), respectively,
Arithmetic Mean (Harmonic Mean)

Furthermore, the percent inhibition of Xanthine oxidase at 45


Research (CDER). Guidance for industry: bioanalytical
12, 16, and 24 hours (post dose) following multiple dosings method validation. May 2001 and Mayer, M. et al., American
with immediate release dosage forms containing 70 mg and Journal of Therapeutics, 12:22-34 (2005), herein each incor
120 mg of febuxostat in healthy subjects were calculated porated by reference). The lower limit of quantitation with a
using Equation 1 below. The results of this calculation were 0.5 mL plasma sample was 0.01 ug/mL for febuxostat.
incorporated into Equation 2, discussed below, and the results 50 The f, can be determined using the in-vitro binding of ''C
of Equation 2 are listed in Table 2 below: febuXostat at a nominal concentration of 1 ug/mL using an
equilibrium dialysis technique, which is well known in the
Percent Inhibition (“% Inhibition') Equation 1 art. For example, the percent f, for febuxostat has been cal
of xanthine oxidase (XOD')
55 culated to be 0.9+0.2 in normal patients and 1.2-0.2 in
patients with severe renal impairment (See Mayer, M. et al.,
%. Inhibition of XOD = 100 C. f. American Journal of Therapeutics, 12:22-34 (2005), herein
C. f... + K. incorporated by reference). In another study with a larger
60
number of subjects, the percent free fraction offebuxostat in
where C plasma concentration of febuXostat in plasma, plasma was calculated to be 0.7+0.1 in male, female, younger
f the free fraction offebuxostat in plasma and K, the xan and an elderly group of Subjects (See Khosravan R., et al.
thine oxidase inhibitory constant offebuxostat. Clinic. Pharmacology & Therapeutics, P50 (2005), herein
The plasma concentration offebuxostat can be determined incorporated by reference).
using a validated high performance liquid chromatography 65 The K, for febuxostat has been determined using a Xan
with fluorescence detection (See Biopharmaceutics Coordi thine oxidase assay Such as that described in Osada Y., et al.,
nating Committee in the Center for Drug Evaluation and European J. Pharmacology, 241:183-188 (1993), herein
US 9.248,119 B2
49 50
incorporated by reference. More specifically, the K, for mg immediate release dosage forms offebuXostat demon
febuxostat has been determined to be 0.7 nM and 0.6 nM, strated that such dosage forms provide a C of about 3.9 and
respectively (See, Osada Y., et al., European J. Pharmacol about 4.2 Lug/mL and exhibited at least 80% inhibition of
ogy, 241:183-188 (1993) and Takano, Y., et al., Life Sciences, xanthine oxidase for about fourteen (14) hours. Further stud
76:1835-1847 (2005)). The K, forfebuxostat is known in the ies involving 240 mg immediate release dosage forms of
art to be 0.6 nM (See, Takano, Y., et al., Life Sciences, febuXostat demonstrated that Such dosage forms provide a
76:1835-1847 (2005), herein incorporated by reference). C of 10.2 ug/mL and exhibit at least 80% inhibition of
xanthine oxidase for about 22 hours. Thereupon, in view of
Additionally, concentrations offebuxostat that exhibited these results, the present inventors conceived of the modified
50%, 60%, 70%, 80%, 90%, 95%, and 99% inhibition of 10
release dosage forms of the present disclosure. These dosage
Xanthine oxidase activity were calculated using the below forms exhibit high levels (at least 80%) inhibition of xanthine
Equation 2 (based on the calculation made above using Equa oxidase for a period of greater than sixteen (16) hours while
tion 1). The result of this calculation for all dosage regimens not compromising patient compliance. The dosage forms of
is listed below in Table 2. 15 the present disclosure maintain high levels of inhibition of the
Xanthine oxidase enzyme at similar or lower total plasma
exposure levels compared to high doses of febuXostat
C %. Inhibition of XOD. K. Equation 2
C (namely, 120 mg and 240 mg QD).
T (100-%. Inhibition of XOD).f,
Example 2
where C plasma concentration of febuXostat in plasma, Estimated Plasma Profiles for Extended Release
f the free fraction offebuxostat in plasma and K, the xan Febuxostat Formulations
thine oxidase inhibitory constant offebuxostat.
25 In view of the pharmacokinetic data pertaining to immedi
TABLE 2 ate release febuxostat formulations included in Example 1,
Percentage Inhibition Following Administration of IR Formulations the inventors developed estimated plasma profiles for various
extended release formulations. Specifically, the inventors
Dosing Time Post Dose developed estimated plasma profiles for three types of formu
30
Regimen 8 hours 12 hours 16 hours 24 hours lations: extended release formulations, two-pulse formula
tions, and three-pulse formulations. The plasma profile infor
10 mgQD 43.6 27.3 O O mation for the extended release formulations is based on a
20 mgQD 55.9 S4O 42.8 O
30 mgQD 62.3 52.5 37.1 35.9 formulation with a polymeric component that provides
40 mgQD 69.8 55.9 43.3 36.2 febuXostat release at a constant rate over time, and may incor
50 mgQD 77.9 64.9 49.1 38.5 35
70 mgQD 81.2 66.3 56.0 35.3 porate technology Such as a matrix formulation. The esti
80 mgQD* 82.7 71.9 60.8 37.0 mated two-pulse formulation data is based on a febuXostat
90 mgQD 85.6 77.7 68.1 S.O.3 formulation having immediate release beads and beads that
120 mgQD 89.0 82.6 75.8 6O.S
160 mgQD 93.9 90.9 81.4 67.6 release febuxostat after approximately 5 hours. The estimated
180 mgQD 94.7 91.7 81.5 68.4 40 three-pulse formulation data is based on a febuxostat formu
240 mgQD 95.7 91.4 86.5 72.4
lation having immediate release beads, beads that release
*The 80 mg data was the result of simulated data used to calculate a simulated percentage
inhibition
febuxostat after approximately 5 hours, and beads that release
febuxostat after approximately 10 hours. The estimated
As shown in Table 2, even when subjects received high 45
plasma profiles for the various formulations are found in
doses offebuxostat (120 mg), at least 80% of inhibition of the Table 3. It should be noted that the plasma profile information
Xanthine oxidase enzyme was not provided for greater than pertaining to the immediate release formulations found in
sixteen (16) hours. Additionally, other studies involving 120 Table 3 represent actual data obtained in clinical testing.
TABLE 3

Estimated Plasma Profiles for Febuxostat Extended Release Formulations

Duration
Time
Conc. -0.1
Dosing Tmax Cmax Cmin AUCoo |g/mL
Formulation Regimen (h) (Lig/mL (Lig/mL) (Ligh/mL) (hours)

IR: 80 QD O.S 3.06 O.O159 7.99 8


SR 80 QD 2.5 O.99 O.O2O 7.32 13.75
2PS-Sh 80 QD 5.75 1.69 O.O2S 7.46 15
3PS-Sh 80 QD 5.5 0.77 O.O28 5.95 17.25

The plasma profiles for the immediate release formulations representactual data, and are notestimated values
US 9.248,119 B2
51 52
Example 3 improper capsule release was replaced by the data from the
corresponding repeated periods using the originally planned
Bioavailability of Febuxostat Released in Various period for that regimen.
Locations of the Gastrointestinal Tract The mean plasma febuXostat concentration-time profiles
(linear and log-linear formats) for each regimen and site of
The inventors tested the relative bioavailability of 80 mg of absorption are depicted in FIG.1. The summary of the mean
febuxostat released in the proximal small intestine, the distal pharmacokinetic parameter estimates for febuXostat follow
Small intestine, and the colon of 12 healthy male Subjects, as ing a single dose offebuXostat 80 mg for all four periods are
compared to the bioavailability of an immediate release dos displayed in Table 5. All febuxostat plasma concentrations
age form. 10 from each subject were used for the pharmacokinetic param
The Subjects were randomly assigned in equal numbers to eter estimations; however, Subject 017-JDF (randomization
one of four regimen sequences, as set forth in Table 4. Sub number 112) had considerably less plasma exposure (C.
and AUC) because of an apparent delayed (approximately 3
jects all received regimens in a crossover fashion according to hours) release of drug product into the distal portion of the
a randomization schedule. Extra periods (up to 2 per Subject) 15 Small intestine (Regimen C) when compared to all others.
were added where Subject dose release issues required repeat Subject 017-JDF distal intestinal release afforded a febux
ing a regimen. There was a washout interval of at least 7 days ostat C value that was 5-13%, and an AUC value that was
between the dose in a period and the next dose in the subse 12-28% that of all other distal releases. Therefore, data from
quent period. the distal intestinal release for this subject was not included in
the statistical analysis.
TABLE 4
TABLE 5
Subiects and Regimen Schedules for FebuXOstat administration
Mean Pharmacokinetic Parameter Estimates for Febuxostat
i
Se- Sub 25
Following a Single Dose of Febuxostat 80 Ing
quence jects Period 1 Period 2 Period 3 Period 4 Tmax Cmax AUC(O-tlqc) AUC(0-inf)
1 3 Regimen A Regimen B Regimen C Regimen D (h) (ngmL) (ng himL) (ng himL)
2 3 Regimen B Regimen A Regimen D Regimen C Reference 80 mg Febuxostat Immediate Release (Regimen A)
3 3 Regimen C Regimen D Regimen A Regimen B
4 3 Regimen D Regimen C Regimen B Regimen A 30 N 12 12 12 12
Mean 1.42 26SO.83 7944.75 8051.70
Regimen A; Febuxostat 80 mg immediate release tablet (reference) SD 1.08 762.84 3265.12 3277.36
RegimenB: InteliSite capsule containing 80 mg offebuxostat drug substance released in the Min O.SO 161O.OO 4394.53 4494.66
proximal small intestine
Regimen C: InteliSite capsule containing 80 mg offebuxostat drug substance released in the Max 4.OO 45SO.OO 13795.70 13873.87
distal small intestine
RegimenD: InteliSite capsule containing 80mg offebuxostat drug substance released in the 80 mg Febuxostat Proximal Intestinal Release (Regimen B)
colon 35
N 12 12 12 12
Blood samples were obtained prior to dosing (0 hour) on Day Mean
SD
O.SO
O.18
3026.67
491.51
7600.62
3O39.19
7703.35
3O42.99
1 of each period, before the investigational product was Min O.25 480.OO 4441.30 4524.33
released from the InteliSite capsule (0 hour preactivation) for Max 1.OO 654O.OO 14797.40 14904.24
Regimens B, C, and D and at 0.25, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 10. 40 80 mg FebuxostatDistal Intestinal Release (Regimen C)
12, 16, 24, 30, and 36 hours after tablet dosing or release of N 12 12 12 12
febuXostat in the selected segment of the gastrointestinal Mean O.73 2S61.83 5964.61 6O72.24
tract. Plasma concentrations offebuxostat were determined SD O.29 OSS.49 2352.75 2352.01
Min O.25 212.OO 1021.8O 1182.78
from EDTA-treated samples using a validated liquid chroma Max 1.OO 4400.OO 9839.35 9967.74
tography assay with mass spectrometric detection using posi 45 80 mg FebuxostatDistal Intestinal Release without
tive ion electrospray ionization at PPD (Richmond, Va.). The Subject 017 (Regimen C)
method utilizes protein precipitation with acetonitrile of a N 11 11 11 11
100-uL aliquot of plasma, with a lower limit of detection of Mean 0.77 2775.45 64 13.95 6516.73
10.0 ng/mL. SD O.26 789.36 1850.34 1864.71
Pharmacokinetic parameters forfebuxostat were estimated 50 Min
Max
O.SO
1.OO
66O.OO
4400.OO
4144.8O
9839.35
4218.01
9967.74
using standard noncompartmental methods. Calculations 80 mg Febuxostat Colon Release (Regimen D)
were performed using the WinNonlin Pro Version 5.2 (Moun
tain View, Calif., USA). The descriptive statistics for phar N 12 12 12 12
macokinetic parameter estimates were computed. Analyses Mean 3.25 382.50 2761.97 2953.24
of variance (ANOVA) were performed on febuxostat T. 55 SD 1.91 173.04 1119.74 1094.88
Min O.SO 1OOOO 1299.93 1455.35
and the natural logarithms of C, AUC(0-tlqc), and AUC Max 6.OO 63.O.OO 5259.23 S411.25
(0-inf) with factors for sequence, subject nested within
sequence, period, and regimen. The effect of the release in the Regimen A: 80 mg febuxostat immediate release reference.
proximal Small intestine, the distal Small intestine, and the Regimen B: 80 mg febuxostat proximal intestinal release.
Regimen C: 80 mg febuxostat distal intestinal release.
colon on the bioavailability offebuxostat was assessed via 60
Regimen D: 80 mg febuxostat colon release.
point estimates and 90% confidence intervals for the ratio of
the central values for febuXostat C and AUCs of Regimen Following administration offebuxostat 80 mg released in the
B to Regimen A. Regimen C to Regimen A, and Regimen D proximal small intestine (Regimen B) or the distal small
to Regimen A, respectively. The Subjects who had improper intestine (Regimen C), mean T values were 65% and 49%
capsule release were allowed to repeat up to two extra periods 65 shorter (4.6% with Subject 017-JDF excluded), respectively,
after their scheduled sequence had ended. For ANOVA pur when compared to that of the immediate release tablet (Regi
poses, the missing or incomplete data from the periods with men A). In the colon (Regimen D), mean T was more than
US 9.248,119 B2
53 54
twice that of the immediate release tablet (Regimen A). When Accordingly, it was observed that following single dose
febuxostat was released in the proximal (Regimen B) or distal oral administrations of febuxostat 80 mg released in the
(Regimen C) small intestine (regardless of Subject 017-JDF), proximal Small intestine, distal Small intestine or colon (Regi
both mean C. values were approximately equal to the mean mens B, C or D) in 12 healthy male subjects (excluding
C. value obtained from the immediate release tablet (Regi Subject 017-JDF for the distal intestinal release), the systemic
men A); whereas, febuxostat mean C. value for the colon exposure offebuxostat was not bioequivalent to that obtained
release (Regimen D) was 14% that of the mean C. value after administration of the febuxostat 80 mg immediate
from the immediate release tablet (Regimen A). Mean febux release tablet (A).
ostat AUC values were generally similar after febuxostat In addition, the inventors used the estimated data included
release in the proximal (Regimen B) intestine and from the 10
immediate release tablet (Regimen A); however, for the distal in Tables 3, 4, and 5 to produce FIGS. 2-4. The inventors
(Regimen C) release, mean AUC values were reduced to 75% calculated estimated pharmacokinetic values for the absorp
(81% when excluding Subject 017-JDF) when compared to tion offebuXostat in various portions of the gastrointestinal
the immediate release tablet (Regimen A) mean AUC values. tract (i.e., the stomach, proximal Small intestine, distal Small
After febuxostat release in the colon (RegimenD), mean AUC intestine, and the colon) and used these parameters to develop
values were approximately 35% of those AUC values from 15 the log-linear graphs of the febuXostat plasma concentration
the immediate release tablet (Regimen A). over time for various dosage forms including an 80 mg
Statistical assessments on the bioavailability offebuxostat 3-Pulse febuxostat formulation, an 80 mg 2-Pulse febuxostat
administered (Regimens, B, C, and D) relative to that febux formulation, and an 80 mg extended release (ER) febuxostat
ostat immediate release (Regimen A) was assessed via point formulation.
estimates and 90% confidence intervals for the ratios of the
central values for C, AUC(0-tlqc), and AUC(0-inf) and are Example 4
summarized in Table 6 (excluding Subject 017-JDF distal
intestinal release data). Modified Release Matrix Tablet Formulations

TABLE 6 25 Modified release dosage forms that continuously release a


Statistical Assessment of the Bioavailability
xanthine oxidoreductase inhibitor, namely febuxostat, were
of Regimens B, C, and D. Compared to Regimen A prepared as matrix tablets containing the ingredients shown in
FIG. 5 (the ingredients are shown in weight percentages).
Point Estimate 90% Confidence More specifically, the tablets were made by wet granulation
of the Relative Interval for the 30 using a Black & Decker Handy Chopper. The order in which
Parameter Bioavailability Point Estimate
the ingredients were added was not critical. A V-blender
Regimen B vs. Regimen A (Blend Master Lab, LC 9292659) was used to manufacture
the final blend. Tablets were compressed on a carver press
Cmax 1.066O (0.7988-14226) into a round shape by utilizing A-2308 tooling or oval shape
AUC(O-tlqc) O.9663 (0.7731-1.2078)
AUC(0-inf) O.9663 (0.7839-1.1911)
35 (A-2253) device. For each of the dosage forms shown in FIG.
Regimen C vs. Regimen A (Subject 017 Excluded) 5, the drug release characteristics were determined and the
resulting dissolution profiles are shown in FIG. 6. More spe
Cmax 1.0555 (0.7810-14267) cifically, with respect to the drug release characteristics and
AUC(O-tlqc) O.84OS (0.6659-1.0609)
AUC(0-inf) O8429 (0.6776-1.0487) dissolution profiles, the oral dosage forms offebuXostat as
Regimen D vs. Regimen A 40 described herein and containing the ingredients shown in
FIG. 5 were evaluated for dissolution in 900 mL of 0.5 M
Cmax O.1356 (0.1015-0.1811) phosphate buffer, pH 6.8, equilibrated at 37° C.ii-0.5° C.
AUC(O-tlqc) O.3475 (0.2777-0.4347)
AUC(0-inf) O.3706 (0.3004-0.4572) using a paddle method (USP Apparatus 2) at 50 rpm. Sample
aliquots were taken at different time intervals and analyzed by
Regimen A: 80 mg febuxostat immediate release reference. 45 high performance liquid chromatography. The compositions
Regimen B: 80 mg febuxostat proximal intestinal release.
Regimen C: 80 mg febuxostat distal intestinal release. shown in FIG. 5 represent extended release febuxostat for
Regimen D: 80 mg febuxostat colon release. mulations.

Based on the data from 12 subjects (11 for the distal intes Example 5
tinal release Regimen C), the 90% confidence intervals for 50
the ratios of the central values when febuxostat 80 mg was Triphasic Dosage Forms
administered in the proximal Small intestine, distal Small
intestine, or colon (Regimens B, C or D, respectively) relative Triphasic dosage forms that release axanthine oxidoreduc
to dosing as an immediate release tablet (Regimen A) were tase inhibitor, Such as febuXostat, can be prepared containing
outside of the bioequivalence limit of 0.80 to 1.25 for febux 55 the ingredients (shown in weight percentages) listed in Table
ostat C, AUC(0-tlqc) or AUC(0-inf). The febuxostat C. 7 below. These dosage forms comprise three (3) sets of gran
central value for the proximal (B) and distal (Regimen C) ules. Granule A is designed to release the active agent in the
release were approximately 7% and 6% greater, respectively, stomach. Granule B is designed to release the active agent in
than the C central value after an immediate release tablet the jejunum. Granule C is designed to release the active agent
(Regimen A). The AUC central values for the proximal 60 in the distal portion of the ileum and ascending colon.
release (Regimen B) or distal release (Regimen C) were More specifically, an active-agent loaded layer can be
reduced by about 3% and 16%, respectively, when compared deposited by spraying an aqueous Suspension of the active
to those after an immediate release tablet (Regimen A). The agent on to a number of neutral cores in order to obtain a
febuxostat C, AUC(0-tlqc) and AUC(0-inf) central values plurality of drug granules. These drug granules are identified
for the colon release (RegimenD) were respective 14%, 35%, 65 as “Granule A'. A first portion of Granule A are removed and
and 37% of those values after an immediate release tablet then coated with a methacrylic acid copolymer dispersion
(Regimen A). (such as Eudragit R. L30D-55 or EudragitR) L100-55) that
US 9.248,119 B2
55 56
contains the active agent. The methacrylic acid copolymer TABLE 8
dispersion can be applied to Granule Aby spraying this aque
ous dispersion directly onto Granule A. This methacrylic acid Composition of Immediate Release FebuXOstat Beads
copolymer coated Granule A is now referred to as “Granule 5 Ingredient % Content
B. A second portion of Granule A is removed and coated with Febuxostat 31.5
an aqueous dispersion which contains a mixture of meth Sugar sphere 52.25
acrylic acid copolymer Type A (such as Eudragit(R) L100 and Hypromellose 16.25
Eudragit(R) L12.5) and Type B (such as Eudragit(R) S100 and
Eudragit(R) S12.5) and the active agent. This aqueous disper 10
sion containing a mixture of methacrylic acid copolymer TABLE 9
Type A and Type B can be applied to Granule Aby spraying
this mixture directly on to Granule A. This coated Granule A Composition of Delayed Release 6.8 Febuxostat Beads
is now referred to as “Granule C. Next, remaining Granules Ingredient % Content
A are then blended together with Granules B and C and filled 15
in hard gelatin size 0 capsules. Febuxostat 25.2
Sugar sphere 4.1.8
Hypromellose 13.0
TABLE 7 Methacrylic Acid Copolymer Type A 4.5
Methacrylic Acid Copolymer Type B 13.7
FebuxostatModified Release Formulations 2O Triethylcitrate 1.8

Components Granule A Granule B Granule C

Febuxostat 2O.O 2O.O 80.0 Example 7


Microcrystalline cellulose sphere 2O.O 2O.O 80.0
Citric Acid 7.0 7.0 1S.O
Sucrose 2S.O 2SO SO.O
25 Composition with 30% Immediate Release
Low-Substituted Hydroxypropyl S.O S.O 2O.O
Febuxostat Beads, 30% Delayed Release 6.0
Cellulose Febuxostat, and 40% Delayed Release Febuxostat
Eudragit L3OD-55 1S.O Beads
Polyethylene Glycol 2.0 30
Titanium Dioxide 1.O The following composition was developed as a three pulse
Talc 2.0 drug delivery system, wherein a single capsule includes three
Methacrylic Acid Copolymer Type B 3O.O types offebuxostat beads. The first pulse consists of 24 mg of
Methacrylic Acid Copolymer Type A 45.O immediate releasefebuxostat beads, wherein the febuxostatis
Triethyl Citrate 6 released immediately after ingestion by the patient. The sec
35
ond pulse consists of 24 mg of delayed release 6.0 febuxostat
FIG. 2 shows the estimated plasma profile for a dosage beads, wherein the febuxostat is released when the beads are
exposed to a pH level of at least 6.0. The third pulse consists
form having three type of granules (3 Pulse), as shown in of 42 mg of delayed release 6.8 febuxostat beads, wherein the
Table 7 above (incorporating all three granules). Specifically, febuxostat is released when the beads are exposed to a pH
FIG. 2 shows the estimated plasma febuxostat concentration level of at least 6.8. The composition of the immediate release
time profile following multiple dosings with a febuxostat febuxostat beads and the delayed release 6.8 febuxostat beads
controlled release (3-Bead, IR=24 mg. CR=24 mg released at are listed in Tables 8 and 9 above, respectively. The compo
5.0 hours, CR-32 mg released at 10 hours) dosage form using sition of the delayed release 6.0 beads is listed in Table 10
the human pharmacokinetic data from Examples 1-3 and 45 below.
other studies and a two-compartmental model with first order
absorption, and comparing the estimated plasma profile to an TABLE 10
80 mg immediate release formulation. Composition of Delayed Release 6.0 Beads
Example 6 50 Ingredient % Content
Febuxostat 25.2
Sugar sphere 4.1.8
Composition with 30% Immediate Release Hypromellose 13.0
Febuxostat Beads and 70% Delayed Release 6.8 Methacrylic Acid Copolymer Type A 18.0
55 Triethylcitrate 2.0
Febuxostat Beads

The following composition was developed as a two pulse Example 8


drug delivery system, wherein a single capsule included two
types offebuxostat beads. The first pulse consists of 24 mg of Composition with 30% Immediate Release
immediate releasefebuxostat beads, wherein the febuxostatis Febuxostat Beads and 70% Delayed-Controlled
released immediately after ingestion by the patient. The sec Release Febuxostat Beads
ond pulse consists of 56 mg of delayed release 6.8 febuxostat
beads, wherein the febuxostat is released when the beads are The following composition was developed as a one pulse
exposed to a pH level of at least 6.8. Tables 8 and 9 below list 65 and delayed controlled release drug delivery system, wherein
the components of the immediate release and delayed release a single capsule includes two types offebuXostat beads. The
6.8 beads. one pulse consists of 24 mg of immediate release febuXostat
US 9.248,119 B2
57 58
beads, wherein the febuxostat is released immediately after cally, six male purebred beagle dogs were used in the study,
ingestion by the patient. The remainder of the capsule com wherein each of the eight test formulations was administered
prises 56 mg of delayed controlled release beads, whereby the to the same set of six dogs. The dogs were individually housed
outermost delayed release layer becomes soluble at pH levels and were not commingled for at least 24 hours after dose
of 6.8 or greater and after the outermost layer has dissolved, administration to allow monitoring of any test article-related
the controlled release layer releases the febuxostat over a effects. The test formulations were administered as an oral
prolonged period of four to six hours. The composition of the dosage form comprising either a capsule or tablet. The dogs
immediate release febuxostat beads is listed in Table 8 above. were fasted overnight through approximately 4 hours post
The composition of the delayed-controlled release beads is dose. Individual doses were calculated based on body weights
listed in Table 11 below. 10 taken on each day of dosing. A 6 ug/kg (0.048 mL/kg) intra
muscular dose of pentagastrin was administered to each dog
TABLE 11 approximately 1 hour before test formulation administration.
The mean plasma febuXostat concentration was measured by
Composition of Delayed-Controlled Release Febuxostat Beads blood samples (approximately 2 mL), which were collected
Ingredient % Content 15 from a jugular vein via Syringe and needle into tubes contain
ing K. EDTA anticoagulant predose, at the time intervals
Febuxostat
Sugar sphere
21.4
35.5
listed above. Subsequently, the plasma samples were shipped
Hypromellose (in IR bead) 11.1 off-site for analysis. The dog model was chosen because of
Surelease E-7-19010 (solid content) 8.4 prior experience with dosing delayed release beads for other
Hypromellose (in CR coat) 3.6 drugs in dogs and the relationship of dog data with human
Methacrylic Acid Copolymer Type A 4.6 data. In previous studies, a delay in T for delayed release
Methacrylic Acid Copolymer Type B 13.6
Triethylcitrate 1.8 beads, with observed T of about 2 hours in dogs had
resulted in a delayed T. up to 8 hours in humans.
All eight febuxostat formulations comprised a total dose of
Example 9
25 80 mg febuxostat, with different compositions of immediate
release, delayed release, controlled release, and delayed-con
trolled release beads. The eight febuxostat formulations
Composition with 20% Immediate Release included:
FebuxoStat Beads and 80% Controlled Release 10-12
Hour Beads
(1) 80 mg of immediate release tablet (Reference formu
30 lation) (termed “Phase 1);
(2) 80 mg of delayed release 5.5 beads, having solubility at
The following composition was developed as a one pulse a pH level of at least 5.5 (termed "Phase 2');
and controlled release drug delivery system, wherein a single (3)24 mg of immediate release beads and 56 mg of delayed
capsule includes two types offebuxostat beads. The one pulse release pH 6.0 beads (termed “Phase 3’):
consists of 16 mg of immediate release febuXostat beads, 35 (4) 24 mg of immediate release beads and 56 mg of delayed
wherein the febuxostat is released immediately after inges release pH 6.8 beads (termed “Phase 4”);
tion by the patient. The remainder of the capsule comprises 64 (5) 24 mg of immediate release beads, 24 mg of delayed
mg of controlled release febuxostat beads, whereby the release pH 6.0 beads, and 32 mg of delayed release pH
febuxostatis released over a prolonged period often to twelve 6.8 beads (termed “Phase 5”);
hours, beginning immediately after ingestion by the patient. 40 (6) 24 mg of immediate release beads and 56 mg of con
The composition of the immediate release febuxostat beads is trolled release 4-6 hour beads (termed “Phase 6):
listed in Table 8 above. The composition of the controlled (7) 24 mg of immediate release beads and 56 mg of
release 10-12 hour beads is listed in Table 12 below. delayed-controlled release beads, wherein the delayed
release layer is soluble at pH levels of at least 6.0, and the
TABLE 12 45 controlled release layer releases febuxostat over a period
Composition of Controlled Release 10-12 Hour Febuxostat Beads of 4-6 hours (termed “Phase 7'); and
(8) 24 mg of immediate release beads and 56 mg of con
Ingredient % Content trolled release 10-12 hour beads (termed “Phase 8').
Febuxostat 29.6 The plasma concentrations for the various dosage forms are
Sugar sphere 49.1 50 shown in FIG. 7. Specifically, FIG. 7 illustrates a line graph
Hypromellose (in IR bead) 15.3 for each formulation, disclosing the average febuXostat
Ethylcellulose 3.6 plasma concentration for each formulation over a six hour
Hypromellose (in CR coat) 2.4 period after ingestion of the dosage forms. Although plasma
samples were collected up to 24 hours, very low plasma
55 concentrations were measured beyond 6-8 hours for most
Example 10 animals. This is consistent with literature reports, which Sug
gest that the length of the gastro-intestinal tract in dogs is
Febuxostat Modified Release Pharmacokinetic Data short compared to humans. Therefore, Solid dosage forms
in Dogs transition quickerthrough the dog gastro-intestinal tract com
60 pared to humans. As a result, delayed release formulations
A study was performed in which eight distinct febuxostat designed to release febuXostat rapidly at specific pH triggers
formulations were administered to 6 dogs in a cross-over were absorbed much better compared to controlled release
manner, and the plasma concentration (ng/ml) for the dog was formulations which release drugs over a period of time.
measured at 0.25, 0.5, 1, 2, 3, 4, 5, 6, 8, 12, 18, and 24 hours In addition, a comparison for all eight formulations was
post dosing. The test was performed to determine how the 65 performed, examining the mean plasma concentration of
various formulations are absorbed in the dog model, and to febuxostat for each Phase of the study at four hours, five
determine plasma concentration profiles over time. Specifi hours, and six hours post dosing. The results were generally
US 9.248,119 B2
59 60
expected as the formulations comprising controlled release In addition, the mean plasma concentrations at 6 hours post
beads did not achieve febuXostat plasma concentrations sig dosing showed similar comparative data to the data at 4 hours
nificantly higher than the formulation comprising only imme and 5 hours post dosing. The Phase 1 Formulation had a mean
diate release beads. To reiterate, this result is expected as the plasma concentration of 53.8 ng/ml. Similar to the data at 4
gastrointestinal tract of dogs is much shorter than that of a and 5 hours, the delayed release formulations of Phase 2,
human. This physiological consequence related to pharma Phase 3, Phase 4, and Phase 5 showed higher mean plasma
cokinetic testing in dog models is discussed in more detail in concentrations of 79.1 ng/ml, 105.3 ng/ml, 69.3 ng/ml, and
the following articles: Stephen C. Sutton, Companion animal 104.9 ng/ml, respectively. The controlled release formula
physiology and dosage form performance, ADVANCED DRUG 10
tions of Phase 6, Phase 7, and Phase 8 showed mean plasma
DELIVERY REVIEWS, 2004, vol. 56, pp. 1383-1398; and Jennifer concentrations of 64.2 ng/ml. 44.3 ng/ml, and 25.6 ng/ml.
B. Dressman, Comparison of Canine and Human Gas respectively.
trointestinal Physiology, PHARMACEUTICAL RESEARCH, 1986, Thus, even though the collection of plasma concentrations
vol. 3, no. 3, pp. 123-131. Accordingly, controlled release in the dog model is inherently limited by the shorter length of
formulations that do not completely release the active com 15 the dog gastrointestinal tract compared to the human gas
ponent until 4-6 hours or 10-12 hours after ingestion may trointestinal tract, the results of the comparison Support
have passed through the majority, if not the entirety, of the improved febuXostat plasma concentrations for modified
dog's gastrointestinal tract by the time the active begins release formulations compared to immediate release formu
release, and high plasma concentrations cannot be achieved. lations. The delayed release formulations of Phase 2, Phase 3,
A summary of these results is included in Table 13 below. Phase 4, and Phase 5 displayed higher mean plasma concen
TABLE 13
trations at 4, 5, and 6 hours post dosing for all formulations
compared to the reference immediate release formulation.
Mean Plasma Concentrations for Febuxostat Formulations in Dogs The controlled release formulations of Phase 6 and Phase 7
25
(both incorporating 4-6 hour controlled release beads)
Mean Concentration (ng/ml showed improved mean plasma concentrations at 4, 5 and 6
Formulation Description 4hrs 5hrs 6 hrs hours (with the exception of the mean concentration for Phase
7 at 6 hours), although not to the extent seen with the delayed
Reference Tablet (Phase 1) 289.8 73.2 53.8 release formulations. These results are likely due to the fact
DR5.5 (phase 2) 632.6 2O7.8 79.1
IR + DR 6.0 (Phase 3) 785.2 283.7 105.3 30 that the controlled release component for Phase 6 and 7 were
IR + DR 6.8 (Phase 4) 448.3 151.3 69.3 designed to release the active over 4-6 hours, meaning that the
IR-DR 6.O- DR 6.8 827.9 249.1 104.9 formulations may have released only a portion of the febux
(Phase 5) ostat before the formulation passed through the entire length
IR + CR Short (Phase 6) 319.0 1242 642
IR + DCR 6.0 (Phase 7) 329.8 95.5 44.3 of the gastrointestinal tract. The controlled release formula
IR + CR Long (Phase 8) 86.2 SO.2 25.6 35 tion of Phase 8 (incorporating 10-12 hour controlled release
beads) displayed the lowest mean plasma concentration,
lower than the reference immediate release formulation. As
At four hours after dosing, the Phase 1 formulation, com discussed previously, this result is not unexpected, as it is
prising 80 mg of immediate release beads, had a mean plasma likely that the controlled release 10-12 hours beads (compris
concentration of 289.8 ng/ml. This value was significantly ing 70% of the formulation) released only a small portion of
40
lower than the mean plasma concentrations for the dosage the febuXostat prior to passing through the dog's gastrointes
forms incorporating delayed release beads, as the Phase 2. tinal tract.
Phase 3, Phase 4, and Phase 5 formulations showed mean
concentrations of 632.6 ng/ml, 785.2 ng/ml. 448.3 ng/ml, and Example 11
827.9 ng/ml, respectively. The dog gastrointestinal pH levels 45
are similar to those found in a human, so the delayed release
beads were not affected by the length of the dog gastrointes Results of a Phase 1. Single-Dose Study in Humans
tinal tract, as seen with the controlled release beads. The of Four Febuxostat Extended Release Formulations
dosage forms incorporating a combination of immediate and One Immediate Release Febuxostat Formulation
release beads and controlled release beads generally experi 50
enced mean plasma concentrations similar to Phase 1, as This examples describes the results of a phase 1, single
expected. Phase 6. Phase 7, and Phase 8 formulations saw center, open-label, randomized, 5-way crossover study.
mean plasma concentrations of 319.0 ng/ml, 329.8 ng/ml, and Thirty-five adult male and female subjects aged 18 to 55
86.2 ng/ml, respectively. In these cases it is likely that only years, inclusive, in good health, were selected to participate in
small amounts of the febuxostat in the controlled release 55 this study. Subjects were randomly assigned in equal numbers
beads was released, as the plasma concentrations were depen to 1 of 5 formulation sequence groups as shown in Table 14.
dent, at least in part, on the immediate release beads found in
the Phase 6, Phase 7, and Phase 8 formulations. TABLE 1.4
Looking at the mean concentrations seen at 5 hours post
dosing, the Phase 1 formulation had a mean plasma concen 60 Formulation Sequence Groups
tration of 73.2 ng/ml. In comparison, the delayed release Number of Formulations
formulations of Phase 2, Phase 3, Phase 4, and Phase 5
showed higher mean plasma concentrations of 207.8 ng/ml. Sequence Subjects Period 1 Period 2 Period 3 Period 4 Period 5
283.7 ng/ml, 151.3 ng/ml, and 249.1 ng/ml, respectively. The 1 7 A. B E C D
controlled release formulations of Phase 6, Phase 7, and 65 2 7 B C A. D E
Phase 8 showed mean plasma concentrations of 124.2 ng/ml. 3 7 C D B E A.
95.5 ng/ml, and 50.2 ng/ml, respectively.
US 9.248,119 B2
61 62
TABLE 14-continued points for 48 hours following administration offebuxostat 80
Formulation Sequence Groups
mg in Periods 1 through 5. FebuXostat plasma concentrations
were quantified using a validated liquid chromatography/
Number of Formulations
tandem mass spectrometry (LCVMS\MS) assay.
Sequence Subjects Period 1 Period 2 Period 3 Period 4 Period 5
4 7 D E C A. B Pharamcokinetic Results
5 7 E A. D B C
Summary of the mean pharmacokinetic parameters esti
Formulation A (reference): Febuxostat (Uloric (R) IR 80 mg tablet,
Formulation B (test): Two-pulsatile prototype (80 mg) febuxostat capsule (TMX-67 XR 10 mated for febuxostat following administration of five differ
Formulation B). ent 80 mg febuxostat formulations are summarized in Table
Formulation C (test): Three-pulsatile prototype (80 mg) febuxostat capsule (TMX-67 XR
Formulation C). 16. Mean plasma concentration-time profiles (linear and log
Formulation D (test); Combination of pulsatile and continuous release (80 mg) febuxostat
capsule (TMX-67 Formulation D).
Formulation E (test); Continuous release (80 mg) prototype febuxostat capsule (TMX-67 linear formats) for febuxostat following oral administration
XRFormulation E).
15 of a single 80 mg dose offebuxostat immediate release tablet
All subjects in the study received 5 febuxostatformulations and four extended release 80 mg capsule formulations are
(IR, 2-pulsatile, 3-pulsatile, combination of pulsatile and presented in FIGS. 8A and 8B. In all subjects, febuxostat was
continuous release, and continuous release) in a crossover detected in plasma immediately with no absorption lag-time
fashion, according to the randomization schedule. A sche
matic of the study design is shown in Table 15. after oral dosing. The plasma febuXostat concentrations fell
TABLE 1.5
Schematic of Study Design
Screening Check-in Treatment Period Washout

Days -28 Day -1 of Febuxostat 80 mg QD on Day 1 of each period At least 7 days between
to -2 each period 1 of 5 different formulations (A-E) containing the dose received in one
febuxostat 80 mg dose given in each period period and the dose in
Days 1-3 of each period the Subsequent period
s--------- Confinement to Unit -------- ->

Pharmacokinetic Sample Collection below the target concentration of 100 ng/mL at approxi
Blood samples (4 mL) for the determination of plasma mately 16 hours postdose following administration of
febuXostat concentration were collected at designated time extended release formulations (Formulations B-E).
TABLE 16
SUMMARY OF PLASMAPHARMACOKINETIC PARAMETERS FOR
FEBUXOSTAT FOLLOWING ADMINISTRATION OF SINGLE ORAL
80 MGDOSE OF FEBUXOSTAT IMMEDIATE RELEASE TABLET
AND FOUR 80 MG EXTENDED RELEASE FORMULATIONS.

Tmax(a) Cmax AUC(O-tlqc) AUC(0-inf) T/3 CL/F VZ.F


(hr) (ng/mL) (ngi hr/mL) (ngi hr/mL) (h) (L/hr) (L)
IRFormulation A

35 35 35 33 33 33 33
Mean 1.O 3136.5 9495.8 9679.8 6.5 9.18 85.3
SD O.S.-3.O 1139.73 2769.23 2836.58 1.88 3.693 34.91
96 CV 36 29 29 29 40 41
Extended-Release Formulation B

34 34 34 27 27 27 27
Mean 3.0 1186.6 6749.3 7133.2 9.7 12.52 171.1
SD O.5-6.O 4O7.31 2198.84 2497.88 3.6S 4.333 78.2
96 CV 34 33 35 38 35 46
Extended-Release Formulation C

34 34 34 32 32 32 32
Mean 3.0 1292.2 74O7.7 7726.7 9.0 11.49 146.6
SD O.5-6.O 477.16 2S56.45 2668.54 3.90 3.683 77.26
96 CV 37 35 35 43 32 53
Extended-Release Formulation D

35 35 35 31 31 31 31
Mean 1.O 1132.9 S832.O 6175.8 9.3 14.SS 187.7
SD O.5-6.O 424.30 2254.62 2357.24 3.78 4.844 82.04
96 CV 37 39 38 41 33 44
US 9.248,119 B2
63 64
TABLE 16-continued
SUMMARY OF PLASMAPHARMACOKINETICPARAMETERS FOR
FEBUXOSTAT FOLLOWING ADMINISTRATION OF SINGLE ORAL
80 MGDOSE OF FEBUXOSTAT IMMEDIATE RELEASE TABLET
AND FOUR 80 MG EXTENDED RELEASE FORMULATIONS.

Tmax(a) Cmax AUC(O-tlqc) AUC(0-inf) T/3 CL/F


(hr) (ng/mL) (ngi hr/mL) (ngi hr/mL) (h) (L/hr)
Extended-Release Formulation E

N 34 34 34 31 31 31 31
Mean 1.5 1246.6 S182.8 5334.2 7.6 17.05 169.1
SD O.S.-3.0 382.04 1911.91 2019.25 4.48 7.423 71.78
96 CV 31 37 38 59 44 42

A = Febuxostat 80 mg DR Tablet
B = Febuxostat 80 mg extended release (Two pulsatile) capsule formulation
C = Febuxostat 80 mg extended release (Three pulsatile) capsule formulation
D = Febuxostat 80 mg extended release (Combination of pulsatile and Continuous) capsule formulation.
E = Febuxostat 80 mg extended release (Continuous Release) capsule formulation.

Example 12 Osmotic tablets can be overcoated with an immediate


release layer of the drug (febuXostat) to overcome time-lag as
Osmotic Tablets of Febuxostat shown in FIG.10. In this FIG. 10, a 60mg tablet offebuxostat
(formulation 2 above) is over-coated with 20 mg offebuxostat
An osmotic tablet formulation was prepared using the 25 which is expected to be release immediately and thus become
swellable core technology. The tablet consists of a drug layer available for absorption.
and a swellable polymer layer. This bilayer tablet is coated Osmotic multiparticulates were prepared by layering
with a semipermeable membrane comprising cellulose febuXostat on microcrystalline cellulose spheres using rou
acetate and polyethylene glycol. A hole was drilled on the top tine techniques in the art. The drug layered beads are coated
surface of the tablet by laser. The semipermeable membrane 30 with a layer of disintegrant (Such as croScarmellose Sodium,
allows water to be absorbed into the tablet, however does not crospovidone, etc.) and then overcoated with an aqueous
allow diffusion of any other material across the membrane. dispersion of ethylcellulose. As seen in the FIG. 11, it is
The Swellable polymer layer swells as it absorbs water and possible to obtain a multiparticulates of the desired release
pushes the drug out of the laser drilled orifice. Composition of characteristics by varying the amount of ethylcellulose coat
the swellable polymer layer and thickness of the semiperme 35 ing. Bursting multiparticulates can be combined uncoated
able membrane affect release of the drug. A tablet composi beads to provide a 2-pulse formulation systems similar to
tion various tablet formulations is shown in Table 17 below.
Formulation 1 is designed to give a longer duration of release other 2-pulse systems described herein.
since it contains lower amount of osmogen NaCl. Higher One skilled in the art would readily appreciate that the
amount of osmogen in formulation 2 is expected to produce present disclosure is well adapted to carry out the objects and
faster swelling of the polymer. 40 obtain the ends and advantages mentioned, as well as those
inherent therein. It will be readily apparent to one skilled in
TABLE 17 the art that varying Substitutions and modifications may be
made to the disclosure disclosed herein without departing
Formulation 1 Formulation 2 Formulation 3 from the scope and spirit of the disclosure. All patents and
45 publications mentioned in the specification are indicative of
Wt 9% Weight Wt 9% Weight Wt 9% Weight the levels of those skilled in the art to which the disclosure
of per tab of per tab of per tab
Layer (mg) Layer (mg) Layer (mg) pertains. All patents and publications are herein incorporated
Drug Layer 300 300 225
by reference to the same extent as if each individual publica
Febuxostat 26.7 26.7 35.6
tion was specifically and individually indicated to be incor
PEOWSRN 72.8 72.8 63.9
50 porated by reference.
10 The disclosure illustratively described herein suitably may
Magnesium O.S O.S O.S be practiced in the absence of any element or elements, limi
Stearate
Push layer 150 150 150
tation or limitations, which is not specifically disclosed
PEO Coag 64.5 49.7 64.5 herein. Thus, for example, in each instance herein any of the
NaCl 34.8 49.6 34.8 55 terms "comprising "consisting essentially of and “consist
Blue Lake #2 O.2 O.2 O.2 ing of may be replaced with either of the other two terms.
Magnesium O.S O.S O.S The terms and expressions which have been employed are
Stearate
used as terms of description and not of limitation, and there is
no intention that in the use of Such terms and expressions of
Formulation 3 was coated with a semipermeable coat con 60 excluding any equivalents of the features shown and
sisting of cellulose acetate (CA) and PEG 3350. The ratio of described orportions thereof, but it is recognized that various
CA:PEG can be varied. For example, the ratio of CA:PEG can modifications are possible within the scope of the disclosure
be in the range of 5:5 to 9:1. A CA:PEG ratio 6:4 results in claimed. Thus, it should be understood that although the
faster release from the tablet compared to 7:3 ratio as seen in present disclosure has been specifically disclosed by pre
the FIG.9. The amount of coating can be varied using routine 65 ferred embodiments and optional features, modification and
techniques known in the art to adjust the dissolution profile as variation of the concepts herein disclosed may be resorted to
desired. by those skilled in the art, and that such modifications and
US 9.248,119 B2
65 66
variations are considered to be within the scope of this dis wherein the beads are incorporated into an oral dosage form
closure as defined by the appended claims. selected from the group consisting of a pill, a tablet, and a
In addition, where features or aspects of the disclosure are capsule, wherein febuXostat is the only active agent in the
described in terms of Markush groups, those skilled in the art modified release pharmaceutical composition,
will recognize that the disclosure is also thereby described in 5 wherein the immediate release beads comprise
terms of any individual member or subgroup of members of (a) an inert core in an amount ranging from about 50% to
the Markush group. For example, if X is described as selected about 55% (w/w) of the weight of the immediate release
from the group consisting of bromine, chlorine, and iodine, bead, and
claims for X being bromine and claims for X being bromine (b) an immediate release layer that encapsulates the inert
and chlorine are fully described. 10
core comprising a mixture of febuXostat and hydrox
What is claimed is:
1. A modified release pharmaceutical composition com ypropyl methylcellulose in an amount ranging from
prising a mixture of immediate release febuXostat beads in an about 45% to about 50% (w/w) of the weight of the
amount ranging from approximately 20% to approximately immediate release bead, the ratio of febuxostat to
40% (w/w) of the total composition weight and delayed 15 hydroxypropyl methylcellulose ranging from about 1.5
release febuxostat beads having a solubility at pH levels to about 3; and
greater than or equal to 6.8 in an amount ranging from wherein the delayed-controlled release beads comprise
approximately 60% to approximately 80% (w/w) of the total (a) an inert core in an amount ranging from about 34% to
composition weight, wherein the beads are incorporated into about 37% (w/w) of the weight of the delayed-controlled
an oral dosage form selected from the group consisting of a release bead,
pill, a tablet, and a capsule, wherein febuXostat is the only (b) an immediate release layer that encapsulates the inert
active agent in the modified release pharmaceutical compo core comprising a mixture of febuXostat and hydrox
sition, ypropyl methylcellulose in an amount ranging from
wherein the immediate release beads comprise about 31% to about 34% (w/w) of the weight of the
(a) an inert core in an amount ranging from about 50% to 25 delayed-controlled release bead, the ratio offebuxostat
about 55% (w/w) of the weight of the immediate release to hydroxypropyl methylcellulose ranging from about
bead, and 1.5 to about 2.5,
(b) an immediate release layer that encapsulates the inert (c) a controlled release layer that encapsulates the imme
core comprising a mixture of febuXostat and hydrox diate release layer comprising a controlled release poly
ypropyl methylcellulose in an amount ranging from 30 merin an amount ranging from about 10% to about 14%
about 45% to about 50% (w/w) of the weight of the (w/w) of the weight of the delayed-controlled release
immediate release bead, the ratio of febuxostat to bead, the controlled release polymer comprising a mix
hydroxypropyl methylcellulose ranging from about 1.5 ture of ethylcellulose aqueous dispersion and hydrox
to about 3; and ypropyl methylcellulose, the ratio of ethylcellulose
wherein the delayed release beads comprise 35 aqueous dispersion to hydroxypropyl methylcellulose
(a) an inert core in an amount ranging from about 40.5% to ranging from about 1.5 to about 3,
about 43% (w/w) of the weight of the delayed release (d) a delayed release pH 6.8 layer that encapsulates the
bead, controlled release layer comprising a delayed release pH
(b) an immediate release layer encapsulating the inert core 6.8 polymer in an amount ranging from about 1% to
comprising a mixture offebuXostat and hydroxypropyl 40 about 20% (w/w) of the weight of the delayed-controlled
methylcellulose in an amount ranging from about 35% release bead, the delayed release pH 6.8 polymer com
to about 40% (w/w) of the weight of the delayed release prising a mixture of methacrylic acid copolymer type A
bead, the ratio offebuxostat to hydroxypropyl methyl and methacrylic acid copolymer type B, the ratio of
cellulose ranging from about 1.5 to about 3, copolymer type A to copolymer type B ranging from
(c) a delayed release enteric polymer layer encapsulating 45 about 0.1 to about 0.5, and
the immediate release layer comprising a delayed (e) a plasticizer in an amount ranging from about 1% to
release enteric polymer in an amount ranging from about about 3% (w/w) of the weight of the delayed-controlled
1% to about 20% (w/w) of the delayed release bead, the release bead, the plasticizer comprising triethylcitrate.
delayed release enteric polymer comprising a mixture of 4. The modified release pharmaceutical composition of
methacrylic acid copolymer type A and methacrylic acid 50 claim 3, wherein the total amount offebuxostat contained in
copolymer type B in a ratio ranging from approximately the composition is 80 mg.
0.1 to approximately 0.5, and 5. A modified release pharmaceutical composition com
(d) a plasticizer in an amount ranging from about 1% to prising a mixture of immediate release febuXostat beads in an
about 3% (w/w) of the weight of the delayed-controlled amount ranging from approximately 10% to approximately
release bead, the plasticizer comprising triethylcitrate. 55 30% (w/w) of the total composition weight and controlled
2. The modified release pharmaceutical composition of release febuXostat beads providing prolonged release of
claim 1, wherein the total amount offebuxostat contained in febuxostat over a period of about 10 hours to about 12 hours,
the composition is 80 mg. in an amount ranging from approximately 70% to approxi
3. A modified release pharmaceutical composition com mately 90% (w/w) of the total composition weight, wherein
prising a mixture of immediate release febuXostat beads in an 60 the beads are incorporated into an oral dosage form selected
amount ranging from approximately 20% to approximately from the group consisting of a pill, a tablet, and a capsule,
40% (w/w) of the total composition weight and delayed wherein febuxostat is the only active agent in the modified
controlled release febuxostat beads having a solubility at pH release pharmaceutical composition,
levels greater than or equal to 6.8 and providing prolonged wherein the immediate release beads comprise
release offebuxostat over a period of about 4 hours to about 65 (a) an inert core in an amount ranging from about 50% to
6 hours, in an amount ranging from approximately 60% to about 55% (w/w) of the weight of the immediate release
approximately 80% (w/w) of the total composition weight, bead, and
US 9.248,119 B2
67 68
(b) an immediate release layer that encapsulates the inert 13. The modified release pharmaceutical composition of
core comprising a mixture of febuXostat and hydrox claim 1, wherein the oral dosage form, after a single admin
ypropyl methylcellulose in an amount ranging from istration to an adult human Subject in need of treatment
about 45% to about 50% (w/w) of the weight of the thereof produces in the Subject a mean time to peak concen
immediate release bead, the ratio of febuxostat to tration (T) of 2-3-cyano-4-(2-methylpropoxy)phenyl-4-
hydroxypropyl methylcellulose ranging from about 1.5 methylthiazole-5-carboxylic acid of about 3.0 hours.
to about 3; and 14. The modified release pharmaceutical composition of
wherein the controlled release beads comprise claim 1, wherein the oral dosage form, after a single admin
(a) an inert core in an amount ranging from about 47% to istration to an adult human Subject in need of treatment
about 51% (w/w) of the weight of the controlled release 10
thereof produces in the Subject a mean area under the curve
bead,
(b) an immediate release layer that encapsulates the inert (0-inf) (AUC) between about 53342ng hr/mL to about
core comprising a mixture of febuXostat and hydrox 7726.7 ng hr/mL.
ypropyl methylcellulose in an amount ranging from 15. The modified release pharmaceutical composition of
about 42% to about 48% (w/w) of the weight of the 15 claim 1, wherein the oral dosage form, after a single admin
controlled release bead, the ratio of febuxostat to istration to an adult human Subject in need of treatment
hydroxypropyl methylcellulose ranging from about 1.5 thereof produces in the Subject a mean area under the curve
to about 2.5, and (0-inf) (AUC) between about 53342ng hr/mL to about
(c) a controlled release layer that encapsulates the imme 7133.2 ng hr/mL.
diate release layer comprising a controlled release poly 16. The modified release pharmaceutical composition of
mer, the controlled release polymer comprising a mix claim 2, wherein the oral dosage form exhibits a mean Cof
ture of ethylcellulose and hydroxypropyl 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
methylcellulose in an amount ranging from about 4% to 5-carboxylic acid of about 1.19 Lig/mL and a mean time to
about 8% (w/w) of the weight of the controlled release peak concentration (T) of 2-3-cyano-4-(2-methylpro
bead, the ratio of ethylcellulose to hydroxypropyl meth 25 poxy)phenyl]-4-methylthiazole-5-carboxylic acid of about
ylcellulose ranging from about 1 to about 2. 3.0 hours.
6. The modified release pharmaceutical composition of 17. The modified release pharmaceutical composition of
claim 5, wherein the total amount offebuxostat contained in claim 2, wherein the oral dosage form exhibits a mean
the composition is 80 mg. AUCo. of 2-3-cyano-4-(2-methylpropoxy)phenyl]-4-me
7. The modified release pharmaceutical composition of 30 thylthiazole-5-carboxylic acid in the amount of about 7133.2
claim 1, wherein the oral dosage form, after a single admin ng.hr/mL.
istration to an adult human subject in need of treatment 18. The modified release pharmaceutical composition of
thereof produces in the Subject a mean maximum plasma claim 2, wherein the oral dosage form exhibits a mean Cof
concentration (C) of 2-3-cyano-4-(2-methylpropoxy) 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
phenyl]-4-methylthiazole-5-carboxylic acid between about 35 5-carboxylic acid of about 1.19 g/mL, a mean T of 2-3-
0.8 ug/mL to about 1.7 ug/mL. cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole-5-car
8. The modified release pharmaceutical composition of boxylic acid of about 3.0 hours and a mean AUCo. of 2-3-
claim 1, wherein the oral dosage form, after a single admin cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole-5-
istration to an adult human Subject in need of treatment carboxylic acid in the amount of about 7133.2 ng hr/mL.
thereof produces in the subject a mean C of 2-3-cyano 40 19. The modified release pharmaceutical composition of
4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic claim 5, wherein the oral dosage form, after a single admin
acid of between about 0.8 ug/mL to about 1.6 lug/mL. istration to an adult human Subject in need of treatment
9. The modified release pharmaceutical composition of thereof produces in the Subject a mean maximum plasma
claim 1, wherein the oral dosage form, after a single admin concentration (C) of 2-3-cyano-4-(2-methylpropoxy)
istration to an adult human Subject in need of treatment 45 phenyl]-4-methylthiazole-5-carboxylic acid between about
thereof produces in the Subject a mean C of 2-3-cyano 0.8 ug/mL to about 1.7 ug/mL.
4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic 20. The modified release pharmaceutical composition of
acid of between about 0.8 ug/mL to about 1.5 lug/mL. claim 5, wherein the oral dosage form, after a single admin
10. The modified release pharmaceutical composition of istration to an adult human Subject in need of treatment
claim 1, wherein the oral dosage form, after a single admin 50 thereof produces in the Subject a mean C of 2-3-cyano
istration to an adult human Subject in need of treatment 4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic
thereof produces in the Subject a mean C of 2-3-cyano acid of between about 0.8 ug/mL to about 1.6 lug/mL.
4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic 21. The modified release pharmaceutical composition of
acid of between 1.13 ug/mL to about 1.29 ug/mL. claim 5, wherein the oral dosage form, after a single admin
11. The modified release pharmaceutical composition of 55 istration to an adult human Subject in need of treatment
claim 1, wherein the oral dosage form, after a single admin thereof produces in the subject a mean C of 2-3-cyano
istration to an adult human Subject in need of treatment 4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic
thereof produces in the Subject a mean C of 2-3-cyano acid of between about 0.8 ug/mL to about 1.5 lug/mL.
4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic 22. The modified release pharmaceutical composition of
acid of about 1.19 g/mL. 60 claim 5, wherein the oral dosage form, after a single admin
12. The modified release pharmaceutical composition of istration to an adult human Subject in need of treatment
claim 1, wherein the oral dosage form, after a single admin thereof produces in the Subject a mean C of 2-3-cyano
istration to an adult human Subject in need of treatment 4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic
thereof produces in the Subject a mean time to peak concen acid of between 1.13 ug/mL to about 1.29 ug/mL.
tration (T) of 2-3-cyano-4-(2-methylpropoxy)phenyl-4- 65 23. The modified release pharmaceutical composition of
methylthiazole-5-carboxylic acid of between about 1.5 hours claim 5, wherein the oral dosage form, after a single admin
to about 3.0 hours. istration to an adult human Subject in need of treatment
US 9.248,119 B2
69 70
thereof produces in the Subject a mean C of 2-3-cyano thereof produces in the Subject a mean area under the curve
4-(2-methylpropoxy)phenyl]-4-methylthiazole-5-carboxylic (0-inf) (AUC) between about 53342ng hr/mL to about
acid of about 1.19 g/mL. 7133.2 ng hr/mL.
24. The modified release pharmaceutical composition of 28. The modified release pharmaceutical composition of
claim 5, wherein the oral dosage form, after a single admin claim 6, wherein the oral dosage form exhibits a mean Cof
istration to an adult human Subject in need of treatment 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
thereof produces in the Subject a mean time to peak concen 5-carboxylic acid of about 1.19 Lig/mL and a mean time to
tration (T) of 2-3-cyano-4-(2-methylpropoxy)phenyl-4- peak concentration (T) of 2-3-cyano-4-(2-methylpro
methylthiazole-5-carboxylic acid of between about 1.5 hours poxy)phenyl]-4-methylthiazole-5-carboxylic acid of about
to about 3.0 hours. 10
3.0 hours.
25. The modified release pharmaceutical composition of 29. The modified release pharmaceutical composition of
claim 5, wherein the oral dosage form, after a single admin claim 6, wherein the oral dosage form exhibits a mean
istration to an adult human Subject in need of treatment AUCo. of 2-3-cyano-4-(2-methylpropoxy)phenyl]-4-me
thereof produces in the Subject a mean time to peak concen thylthiazole-5-carboxylic acid in the amount of about 7133.2
tration (T) of 2-3-cyano-4-(2-methylpropoxy)phenyl-4- 15
ng.hr/mL.
methylthiazole-5-carboxylic acid of about 3.0 hours. 30. The modified release pharmaceutical composition of
26. The modified release pharmaceutical composition of
claim 5, wherein the oral dosage form, after a single admin claim 6, wherein the oral dosage form exhibits a mean C of
istration to an adult human Subject in need of treatment 2-3-cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole
thereof produces in the Subject a mean area under the curve 5-carboxylic acid of about 1.19 Jug/mL, a mean T of 2-3-
cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole-5-car
(0-inf) (AUC) between about 5334.2 ng hr/mL to about boxylic acid of about 3.0 hours and a mean AUCo. of 2-3-
7726.7 ng hr/mL. cyano-4-(2-methylpropoxy)phenyl-4-methylthiazole-5-
27. The modified release pharmaceutical composition of
claim 5, wherein the oral dosage form, after a single admin carboxylic acid in the amount of about 7133.2 nghr?mL.
istration to an adult human Subject in need of treatment k k k k k

Potrebbero piacerti anche