Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ORAL EXTENDED RELEASE PHARMACEUTICAL COMPOSITIONS FOR PREVENTING OR TREATING HEARING LOSS
Document Type and Number:
WIPO Patent Application WO/2019/154893
Kind Code:
A1
Abstract:
The present invention relates to oral extended release (ER) pharmaceutical compositions comprising a PPAR agonist for preventing or treating hearing loss and/or for preventing or inhibiting hair cell degeneration or hair cell death in a subject.

Inventors:
BAUSCH ALEXANDER (CH)
ZELLER MAX (CH)
Application Number:
PCT/EP2019/052968
Publication Date:
August 15, 2019
Filing Date:
February 07, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
STREKIN AG (CH)
International Classes:
A61K9/00; A61K9/22; A61K9/24; A61K9/28; A61K9/36; A61K9/48; A61K9/50; A61K31/192; A61K31/4439; A61P27/16
Domestic Patent References:
WO2016128438A12016-08-18
Foreign References:
KR100697582B12007-03-22
EP2987489A12016-02-24
US4687777A1987-08-18
US5421818A1995-06-06
US5474529A1995-12-12
US5476446A1995-12-19
US87420897A1997-06-13
US20070167918A12007-07-19
US5135757A1992-08-04
Other References:
MARTIN P H ET AL: "Lipid metabolism disorders and deafness of the inner ear. Therapeutic approaches: clinical testing of clofibrate (Lipavlon: I.C.I. Pharma laboratories)", vol. 26, no. 9, 1 November 1977 (1977-11-01), pages 725 - 728, XP009512382, ISSN: 0398-9771, Retrieved from the Internet [retrieved on 20190403]
HYUNG JIN JUN ET AL: "Ciglitazone inhibits cigarette smoke solution-induced inflammatory responses in human middle ear epithelial cells", INTERNATIONAL JOURNAL OF PEDIATRIC OTORHINOLARYNGOLOGY, vol. 76, no. 8, 1 August 2012 (2012-08-01), pages 1136 - 1139, XP055184479, ISSN: 0165-5876, DOI: 10.1016/j.ijporl.2012.04.017
MELAS SYNDROME, ANNALS OF NEUROLOGY, vol. 43, no. 3, March 1998 (1998-03-01), pages 350 - 359
BARRENAS ML; JONSSON B; TUVEMO T; HELLSTROM PA; LUNDGREN M, J CLIN ENDOCRINOL METAB., vol. 90, no. 8, 31 May 2005 (2005-05-31), pages 4452 - 6
DORMANDY JA; CHARBONNEL B; ECKLAND DJ; ERDMANN E; MASSI-BENEDETTI M; MOULES IK; SKENE AM; TAN MH; LEFEBVRE PJ; MURRAY GD: "PROactive investigators", LANCET, vol. 366, no. 9493, 8 October 2005 (2005-10-08), pages 1279 - 89
FLOREZ JC; JABLONSKI KA; SUN MW; BAYLEY N; KAHN SE; SHAMOON H; HAMMAN RF; KNOWLER WC; NATHAN DM; ALTSHULER D: "Diabetes Prevention Program Research Group", J CLIN ENDOCRINOL METAB., vol. 92, no. 4, April 2007 (2007-04-01), pages 1502 - 9
NISSEN SE; WOLSKI K, N ENGL J MED., vol. 356, no. 24, 14 June 2007 (2007-06-14), pages 2457 - 71
N ENGL J MED., vol. 357, no. 1, 5 July 2007 (2007-07-05), pages 100
BONDS DE; CRAVEN TE; BUSE J; CROUSE JR; CUDDIHY R; ELAM M; GINSBERG HN; KIRCHNER K; MARCOVINA S; MYCHALECKYJ JC, DIABETOLOGIA, vol. 55, no. 6, June 2012 (2012-06-01), pages 1641 - 50
RABKIN SW; HAYDEN M; FROHLICH J, ATHEROSCLEROSIS, vol. 73, no. 2-3, October 1988 (1988-10-01), pages 233 - 40
SCHIMA SM; MACIEJEWSKI SR; HILLEMAN DE; WILLIAMS MA; MOHIUDDIN SM, EXPERT OPIN PHARMACOTHER, vol. 11, no. 5, April 2010 (2010-04-01), pages 731 - 8
ADABAG AS; MITHANI S; A1 ALOUL B; COLLINS D; BERTOG S; BLOOMFIELD HE: "Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group", AM HEART J., vol. 157, no. 5, May 2009 (2009-05-01), pages 913 - 8
LINCOFF AM; TARDIF JC; SCHWARTZ GG; NICHOLLS SJ; RYDEN L; NEAL B; MALMBERG K; WEDEL H; BUSE JB; HENRY RR: "AleCardio Investigators", JAMA, vol. 311, no. 15, 16 April 2014 (2014-04-16), pages 1515 - 25
FERNANDEZ M; GASTALDELLI A; TRIPLITT C; HARDIES J; CASOLARO A; PETZ R; TANTIWONG P; MUSI N; CERSOSIMO E; FERRANNINI E, DIABETES OBES METAB., vol. 13, no. 10, October 2011 (2011-10-01), pages 893 - 902
BAYS H; MCELHATTAN J; BRYZINSKI BS: "GALLANT 6 Study Group", DIAB VASC DIS RES., vol. 4, no. 3, September 2007 (2007-09-01), pages 181 - 93
SAAD MF; GRECO S; OSEI K; LEWIN AJ; EDWARDS C; NUNEZ M; REINHARDT RR: "Ragaglitazar Dose-Ranging Study Group", DIABETES CARE, vol. 27, no. 6, June 2004 (2004-06-01), pages 1324 - 9
AGRAWAL R., CURR DRUG TARGETS, vol. 15, no. 2, February 2014 (2014-02-01), pages 151 - 5
AHLAWAT P; SRINIVAS NR, EUR J DRUG METAB PHARMACOKINET, vol. 33, no. 3, July 2008 (2008-07-01), pages 187 - 90
WANG X; SNG MK; FOO S; CHONG HC; LEE WL; TANG MB; NG KW; LUO B; CHOONG C; WONG MT, J CONTROL RELEASE, vol. 197, 10 January 2015 (2015-01-10), pages 138 - 47
CARIOU B; STAELS B, EXPERT OPIN INVESTIG DRUGS, vol. 23, no. 10, October 2014 (2014-10-01), pages 1441 - 8
TAYGERLY JP; MCGEE LR; RUBENSTEIN SM; HOUZE JB; CUSHING TD; LI Y; MOTANI A; CHEN JL; FRANKMOELLE W; YE G, BIOORG MED CHEM., vol. 21, no. 4, 15 February 2013 (2013-02-15), pages 979 - 92
KNOP; KLEINBUDDE, INT. J. PHARMAC., vol. 457, no. 2, 2013, pages 527 - 536
VIVEK K. PAWAR; SHASWAT KANSAL; GARIMA GARG; RAJENDRA AWASTHI; DEEPAK SINGODIA; GIRIRAJ T. KULKARNI, DRUG DELIVERY, vol. 18, no. 2, 2011, pages 97 - 110
SOBKOWICZ HM; LOFTUS JM; SLAPNICK SM, ACTA OTOLARYNGOL, vol. 502, 1993, pages 3 - 36
Attorney, Agent or Firm:
SCHÖLLHORN, Andreas (CH)
Download PDF:
Claims:
Claims

1. An oral extended release (ER) pharmaceutical composition comprising a peroxisome proliferator activated receptor (PPAR) agonist for use in a method of preventing or treating hearing loss in a subject.

2. An oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject.

3. The oral ER pharmaceutical composition for use in a method of claim 1 or 2, wherein the PPAR agonist is pioglitazone or a pharmaceutically acceptable salt thereof.

4. The oral ER pharmaceutical composition for use in a method of any one of the preceding claims, wherein said composition is a tablet, a film coated tablet or a capsule.

5. The oral ER pharmaceutical composition for use in a method of any one of the preceding claims, comprising an extended release matrix, wherein the extended release matrix comprises a hydrophilic polymer.

6. The oral ER pharmaceutical composition for use in a method of any one of the preceding claims, further comprising at least one water-soluble and/or water-insoluble filler.

7. An oral ER pharmaceutical composition comprising:

i. an ER matrix comprising a PPAR agonist and a hydrophilic polymer;

ii. a water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D- fructose, dextran, polyoxyethylene hydrogenated castor oil,

polyoxyethylenepolyoxypropyleneglycol, sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and

iii. a water-insoluble filler selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, sodium carboxymethyl starch, microcrystalline cellulose, crystalline cellulose, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate.

8. The oral ER pharmaceutical composition for use in a method of claim 5 or the oral ER pharmaceutical composition of claim 7, wherein said hydrophilic polymer is a cellulosic polymer.

9. The oral ER pharmaceutical composition for use in a method or the oral ER pharmaceutical composition of claim 8, wherein said cellulosic polymer is

hydroxypropyl methylcellulose (hypromellose).

10. The oral ER pharmaceutical composition for use in a method of claim 6, or the oral ER pharmaceutical composition of any one of claims 7 to 9, wherein said water- insoluble filler is selected from the group consisting of dicalcium phosphate, microcrystalline cellulose and mixtures thereof.

11. The oral ER pharmaceutical composition for use in a method of claim 6 or the oral ER pharmaceutical composition of any one of claims 7 to 10, wherein said water- soluble filler is lactose.

12. The oral ER pharmaceutical composition for use in a method of any one of claims 1 to 6 and 8 to 11 or the oral ER pharmaceutical composition of any one of claims 7 to 11, further comprising at least one glidant selected from the group consisting of fumed silica and/or talc.

13. The oral ER pharmaceutical composition for use in a method of any one of claims 1 to 6 and 8 to 12 or the oral ER pharmaceutical composition of any one of claims 7 to 12, further comprising at least one lubricant selected from the group consisting of stearic acid, magnesium stearate, calcium stearate, sucrose esters of fatty acids, sodium stearyl fumarate, waxes, DL-leucine, sodium lauryl sulfate and magnesium lauryl sulfate or mixtures thereof.

14. The oral ER pharmaceutical composition for use in a method of any one of claims 1 to 6 and 8 to 13 or the oral ER pharmaceutical composition of any one of claims 7 to 13, further comprising at least one binder which is povidone (polyvinylpyrrolidone) .

15. The oral ER pharmaceutical composition for use in a method of any one of claims 1 to 6 and 8 to 14 or the oral ER pharmaceutical composition of any one of claims 7 to 14, comprising:

i. pioglitazone hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate;

v. povidone;

vi. microcrystalline cellulose;

vii. fumed silica;

viii. talc;

ix. magnesium stearate; and optionally

x. a non- functional film-coating.

16. A PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR

agonist for use in a method of preventing or treating hearing loss in a subject and/or for use in a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject;

wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

Description:
Oral extended release pharmaceutical compositions for preventing or treating hearing loss

The field of the invention

The present invention relates to oral extended release (ER) pharmaceutical compositions comprising a PPAR agonist for preventing or treating hearing loss and/or for preventing or inhibiting hair cell degeneration or hair cell death in a subject.

Background of the invention

Hearing loss is related to damage of auditory cells e.g. apoptosis of the hair cells as a consequence of e.g. a continuous stress situation or a traumatic event e.g. leading to the activation of inflammatory pathways. Hearing loss can be caused e.g. by a noise trauma, by a medical intervention, by ischemic injury, by a non specific stress leading to sudden hearing loss or by age or can be chemically induced, wherein the chemical induction is caused e.g. by an antibiotic or a chemotherapeutic agent. Child hearing loss might be caused by pre or post natal deficiencies in the energy homeostasis of auditory cells. Hearing loss can also be caused by mitochondrial dysfunction. (C. M. Sue PhD, FRACP1, Cochlear origin of hearing loss in MELAS syndrome, Annals ofNeurology. Volume 43, Issue 3, pages 350-359, March 1998). In addition a link between metabolic syndrome and hearing loss could be shown (Barrenas ML, Jonsson B, Tuvemo T, Hellstrom PA, Lundgren M, J Clin Endocrinol Metab. 2005 Aug;90(8):4452-6. Epub 2005 May 31). Hearing loss can be of sensorineural origin caused by a damage leading to malnutrition of the cells in early brain development.

Hair cells are fully differentiated and are not replaced after cell death (only a few thousand cells from birth). It is well described in the literature that after stress and damage of the hair cells, the cells can enter in a resting state with no functionality related to the hearing process but remain viable. Approaches to stimulate development or regeneration of new hair cells e.g. by administering growth factors or by stem cell-based therapies in order to achieve disease modification bear the risk of pro-tumorigenic side-effects.

Hearing impairment is a major global health issue with profound societal and economic impact affecting over 275 million people world- wide. The occurrence of hearing loss is rapidly rising, due to e.g. increasing noise exposure and aging populations. With no approved pharmaceutical therapies available today, the unmet medical need is very high. In particular there is a need for providing effective methods for prevention and subsequent treatment of hearing loss which allow for immediate as well as long term maintenance of preventive and/or therapeutic effects.

Summary of the invention

The present invention relates generally to oral extended release (hereinafter named "ER") pharmaceutical compositions comprising a PPAR agonist for use in methods of preventing or treating hearing loss and methods of preventing or inhibiting hair cell degeneration or hair cell death. The present invention provides oral ER pharmaceutical compositions which allow for protection of hair cells from stress e.g. from noise induced stress, from surgery induced stress or from chemically induced stress such as stress induced by an antibiotic or by a

chemotherapeutic agent or from unspecific stress which may cause hearing loss. By using the oral ER pharmaceutical compositions described herein, immediate and subsequent long term maintenance of preventive and/or therapeutic effect can be achieved. In a standard model established in hearing loss research, it could be shown that treatment with a PPAR agonist protects hair cells, which upon exposure to an antibiotic are normally destroyed within 48 hours. The addition of the PPAR agonist prior to antibiotic challenge was able to prevent hair cells from apoptosis and cell death in a dose-dependent manner.

In a first aspect, the present invention relates to an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating hearing loss in a subject.

In a further aspect, the present invention relates to an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject.

In a further aspect, the present invention relates to an oral ER pharmaceutical composition comprising:

i. an ER matrix comprising a PPAR agonist and a hydrophilic polymer;

ii. a water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D- fructose, dextran, polyoxyethylene hydrogenated castor oil, polyoxyethylenepolyoxypropyleneglycol, sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and

iii. a water-insoluble filler selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, sodium carboxymethyl starch, microcrystalline cellulose, crystalline cellulose, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate.

In a further aspect, the present invention relates to a kit for preventing or treating hearing loss and/or preventing or inhibiting hair cell degeneration or hair cell death in a subject comprising an oral ER pharmaceutical composition as described herein and instructions for using the kit.

In a further aspect, the present invention relates to a package comprising: at least one single unit dosage form comprising a PPAR agonist and/or at least one multiple unit dosage form comprising a PPAR agonist, wherein said single unit dosage form and said multiple unit dosage form each independently is an oral ER pharmaceutical composition as described herein; and wherein said at least one single unit dosage form and/or at least one multiple unit dosage form is/are packed in a package.

In a further aspect, the present invention relates to a process for producing an oral ER pharmaceutical composition described herein, comprising the steps of:

i. screening and blending the active substance, e.g. the PPAR agonist and all excipients except for the optional glidants and lubricants, optionally using pre- blending steps;

ii. granulating the blend using water or appropriate solvents;

iii. wet screening the mass obtained in the preceeding step through an appropriate sieve or mill;

iv. drying the wet granulate in an oven or fluid bed

v. dry screening (“calibrating”) the dry granulate through an appropriate sieve or mill

vi. optionally pre-mixing the glidants and lubricants if any with part of the dry granulate, and end-mixing the pre-mix with the remaining part of the granulate vii. compressing this final mass to tablets of appropriate size and shape

viii. optionally film coating the tablets by e.g. an immediate release standard film coating solution/suspension. In a further aspect, the present invention provides a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating hearing loss in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and

administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject;

wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

In a further aspect, the present invention provides a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or inhibting hair cell degeneration or hair cell death in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and

administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject;

wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

Brief description of the figures

Figure 1 A-C show quantitation of the average number of hair cells remaining in the apical, basal and middle turns of the organ of Corti (OC). While gentamicin (200mM) treatment resulted in a consistent reduction of hair cell number of approximately 50 - 70% in each segment, Pioglitazone at both concentrations (2mM and IOmM) was able to significantly prevent gentamicin-dependent hair cell loss in all turns. The values for each turn were averaged for the 10 OCs used for each condition. Significant differences between treatment groups in OHC and IHC (OHC = outer hair cell; IHC = inner hair cell) were determined using analysis of variance (A OVA) followed by the least significant difference (LSD) post-hoc test (Stat View 5.0). Differences associated with P-values of less than 0.05 were considered to be statistically significant. All data are presented as mean ± SD.

Figure 2 shows the change in the average hearing thresholds in guinea pigs determined by auditory brainstem response (ABR) one week or two weeks after noise challenge vs. pre treatment values. Threshold shifts at individual frequencies were calculated for each animal by subtracting post-noise from pre-noise values. Group averages at each frequency were determined. An overall threshold shift was calculated for each treament group and timepoint by averaging the individual frequency shifts over 8 - 20 KHz. Data are mean ± S.D. * p< 0.05.

Figure 3 A-C show quantitation of the average number of hair cells remaining in defined segments in the medio-basal turns of the organ of Corti (OC). While gentamicin (50mM) treatment resulted in a consistent reduction of hair cell number of approximately 50 %, tesaglitazar, muraglitazar and fenofibric acid were all able to significantly prevent gentamicin-dependent hair cell loss. The values were averaged for the 5-7 OCs used for each condition. Significant differences between treatment groups in hair cell numbers were determined using analysis of variance (ANOVA) followed by the least significant difference (LSD) post-hoc test (Stat View 5.0). Differences associated with P-values of less than 0.05 were considered to be statistically significant. All data are presented as mean ± SD. **** = p< 0.001.

Figure 4 shows the dissolution curve of an uncoated pioglitazone-HCl ER tablet based on hypromellose (according to Example 10), compressed at 200N tablet hardness (2 batches using different quantities of granulation liquid).

Figure 5 shows the dissolution curve of an uncoated pioglitazone-HCl ER tablet based on hypromellose (according to Example 10), compressed at 150N tablet hardness, compared to those compressed at 200N tablet hardness.

Figure 6 shows the dissolution curve of a film-coated pioglitazone-HCl ER tablet based on hypromellose (according to Example 10), compressed at 200N tablet hardness, compared to uncoated tablets.

Figure 7 shows the dissolution curve of a film-coated pioglitazone-HCl ER tablet based on hypromellose with different concentrations and types of hypromellose (according to Example 11).

Detailed description of the invention

The present invention provides oral ER pharmaceutical compositions comprising a PPAR agonist for use in a method of preventing or treating hearing loss and/or for use in a method of preventing or inhibiting hair cell degeneration or hair cell death.

For the purposes of interpreting this specification, the following definitions will apply and whenever appropriate, terms used in the singular will also include the plural and vice versa. It is to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting. The terms "comprising", "having", and "including" are to be construed as open-ended terms (i.e., meaning "including, but not limited to") unless otherwise noted.

As indicated above, the terms "extended release" and "ER" are used herein

interchangeably.

The term "extended release" as used herein in relation to the composition according to the invention or used herein in relation to coating or coating material or used in any other context means release of the active pharmaceutical ingredient (API) e.g. a PPAR agonist which is not immediate release, but release over a pre-defmed, longer time period of up to 24 hours, such as 4-24 hours, preferably 4-10 hours, more preferably 5-8 hours. Within this time window, 70-100%, preferably 75-95%, more preferably 80-90% of the content of the API is released.

An ER composition may increase T max or reduce C max , or both increase T max and reduce C max , as compared to an immediate release composition. An ER composition as compared to an immediate release composition comprises one or more agents which act to prolong release of the API; for example, the API may be embedded in a matrix and/or surrounded by a membrane which, in either case, controls (reduces) the rate of diffusion of the API into the GI tract. One version of membrane-control is osmotic pump systems in which a core comprises e.g. solid API or a highly concentrated API in combination with an osmotic agent which acts to imbibe water from the GI tract through a semi-permeable membrane, the API being driven out of an orifice in the device by the osmotic pressure generated in the device, such that release of the API is controlled by water influx across the semipermeable membrane.

“C max ” means the the peak concentration of the drug in the plasma.

"T max ” means the time from administration to reachC max .

The term "PPAR agonist " as used herein refers to a drug that is activating peroxisome proliferator activated receptor (PPAR) such as PPAR gamma receptor, PPAR alpha receptor, PPAR delta receptor or combinations thereof and includes PPAR gamma agonists such as e.g. pioglitazone, troglitazone or rosiglitazone, PPAR alpha agonists such as e.g. fibrates such as fenofibrate (fenofibric acid), clofibrate or gemfibrozil, PPAR dual agonists (PPAR

alpha/gamma or PPAR alpha/delta agonists) such as e.g. aleglitazar, muraglitazar,

tesaglitazar, ragaglitazar, saroglitazar, GFT505 or naveglitazar, PPAR delta agonists such as e.g. GW501516, PPAR pan agonists (PPAR alpha/delta/gamma agonist) or selective PPAR modulators such as e.g. INT131 and the pharmaceutically acceptable salts of these compounds. Usually PPAR gamma agonists, PPAR modulators, PPAR alpha agonists and/or PPAR alpha/gamma dual agonists are used in the oral ER pharmaceutical compositions of the present invention, in particular PPAR gamma agonists, PPAR alpha agonists and/or PPAR alpha/gamma dual agonists are used in the oral ER pharmaceutical compositions of the present invention, more particularly PPAR gamma agonists selected from the group consisting of pioglitazone, rosiglitazone, troglitazone and pharmaceutically acceptable salts thereof, preferably pioglitazone or pharmaceutically acceptable salts thereof, PPAR alpha agonists selected from the group consisting of fenofibrate (fenofibric acid), clofibrate, gemfibrozil and pharmaceutically acceptable salts thereof, preferably fenofibrate (fenofibric acid) or pharmaceutically acceptable salts thereof and/or PPAR alpha/gamma dual agonists selected from the group consisting of aleglitazar, muraglitazar, tesaglitazar, ragaglitazar, saroglitazar, GFT505, naveglitazar or pharmaceutically acceptable salts thereof, preferably muraglitazar, tesaglitazar or pharmaceutically acceptable salts thereof are used in the oral ER

pharmaceutical compositions of the present invention. Preferably PPAR gamma agonists are used in the oral ER pharmaceutical compositions of the present invention, more preferably PPAR gamma agonists or modulators selected from the group consisting of pioglitazone, rosiglitazone, troglitazone, INT131 and pharmaceutically acceptable salts thereof, even more preferably PPAR gamma agonists selected from the group consisting of pioglitazone, rosiglitazone, troglitazone and pharmaceutically acceptable salts thereof are used. In particular, pioglitazone or pharmaceutically acceptable salts thereof, e.g. pioglitazone hydrochloride is used in the oral ER pharmaceutical compositions of the present invention. Most preferably, pioglitazone hydrochloride is used.

In one embodiment, a micronized PPAR agonist is used. Preferably micronized pioglitazone or pharmaceutically acceptable salts, most preferably micronized pioglitazone hydrochloride is used.

Particle size is generally reported on a cumulative distribution by volume basis. The term "micronized" as used herein means particles with a median particle size, expressed by using the cumulative distribution by volume, of between about 1 pm and about 75 pm.

Preferably 50 % of the particles (Dv50) of the micronized PPAR agonist as referred herein are smaller than or equal to about about 50 pm, more preferably smaller than or equal to about 10 pm, most preferably smaller than or equal to about 5 pm, in particular about 1 to about 10 pm, more particular about about 2 to about 8 pm, even more particular about 3 to about 5 pm, most particular about 4 pm. Micronization is a process of reducing the average diameter of particles of a solid material, whereby the particles are mostly passed through a jet mill. Other mill types may be used as well, e.g. pin mills. The required particle size specification may as well be achieved without a specific milling step by appropriate process conditions in the final precipitation step of the drug substance chemical production. Size reduction is used to increase the surface area of a drug substance and thereby modulate formulation dissolution properties. Micronization is also used to maintain a narrow and consistent particle size distribution for any formulation described herein.

A further purpose of micronization is to allow an easy application of the formulations of the invention by a parenteral syringe. In some embodiments, the needle is wider than a 18 gauge needle. In another embodiment, the needle gauge is from 18 gauge to 30 gauge. In a further embodiment, the needle is a 21 gauge needle. Depending upon the thickness or viscosity of a composition disclosed herein, the gauge level of the syringe or hypodermic needle are varied accordingly. Thus, the formulations of the invention comprising micronized PPAR agonists are ejected e.g. from a 1 mL syringe adapted with a 50 mm length 21 G needle (nominal inner diameter 0.495 mm) without any plugging or clogging.

In one embodiment, a thiazolidinedione PPAR agonist is used in the oral ER

pharmaceutical compositions of the invention. Suitable thiazolidinedione PPAR agonists are for example pioglitazone, troglitazone, rosiglitazone or pharmaceutically acceptable salts thereof.

Pioglitazone is described e.g. in US Patent No. 4,687,777 or in Dormandy JA,

Charbonnel B, Eckland DJ, Erdmann E, Massi-Benedetti M, Moules IK, Skene AM, Tan MH, Lefebvre PJ, Murray GD, Standl E, Wilcox RG, Wilhelmsen L, Betteridge J, Birkeland K, Golay A, Heine RJ, Koranyi L, Laakso M, Mokan M, Norkus A, Pirags V, Podar T, Scheen A, Scherbaum W, Schemthaner G, Schmitz O, Skrha J, Smith U, Taton J; PROactive investigators. Lancet. 2005 Oct 8; 366(9493): 1279-89, and is represented by the structural formula indicated below:

Troglitazone is described e.g. in Florez JC, Jablonski KA, Sun MW, Bayley N, Kahn SE, Shamoon H, Hamman RF, Knowler WC, Nathan DM, Altshuler D; Diabetes Prevention Program Research Group. J Clin Endocrinol Metab. 2007 Apr;92(4): 1502-9 and is represented by the structural formula indicated below:

Rosiglitazone is described e.g. in Nissen SE, Wolski K. N Engl J Med. 2007 Jun 14;356(24):2457-71. Erratum in: N Engl J Med. 2007 Jul 5;357(l): 100. Fenofibrate is described e.g. in Bonds DE, Craven TE, Buse J, Crouse JR, Cuddihy R, Elam M, Ginsberg HN, Kirchner K, Marcovina S, Mychaleckyj JC, O'Connor PJ, Sperl-Hillen JA. Diabetologia. 2012 Jun;55(6): 1641-50 and is represented by the structural formula indicated below:

Clofibrate is described e.g. in Rabkin SW, Hayden M, Frohlich J. Atherosclerosis. 1988 Oct;73(2-3):233-40 and is represented by the structural formula indicated below:

Fenofibrate (fenofibric acid) is described e.g. in Schima SM, Maciejewski SR, Hilleman DE, Williams MA, Mohiuddin SM. Expert Opin Pharmacother. 2010 Apr;l 1 (5):731 -8 and is represented by the structural formula indicated below:

Gemfibrozil is described e.g. in Adabag AS, Mithani S, Al Aloul B, Collins D, Bertog S, Bloomfield HE; Veterans Affairs High-Density Lipoprotein Cholesterol Intervention Trial Study Group. Am Heart J. 2009 May;l57(5):9l3-8 and is represented by the structural formula indicated below:

Aleglitazar is described e.g. in Lincoff AM, Tardif JC, Schwartz GG, Nicholls SJ, Ryden L, Neal B, Malmberg K, Wedel H, Buse JB, Henry RR, Weichert A, Cannata R, Svensson A, Volz D, Grobbee DE; AleCardio Investigators. JAMA. 2014 Apr

l6;311(15): 1515-25 and is represented by the structural formula indicated below:

Muraglitazar is described e.g. in Fernandez M, Gastaldelli A, Triplitt C, Hardies J, Casolaro A, Petz R, Tantiwong P, Musi N, Cersosimo E, Ferrannini E, DeFronzo RA.

Diabetes Obes Metab. 2011 Oct;l3(l0):893-902 and is represented by the structural formula indicated below:

Tesaglitazar is described e.g. in Bays H, McElhattan J, Bryzinski BS; GALLANT 6 Study Group. Diab Vase Dis Res. 2007 Sep;4(3): l8l-93 and is represented by the structural formula indicated below:

Ragaglitazar is described e.g. in Saad ML, Greco S, Osei K, Lewin AJ, Edwards C, Nunez M, Reinhardt RR; Ragaglitazar Dose-Ranging Study Group. Diabetes Care. 2004 Jun;27(6): 1324-9 and is represented by the structural formula indicated below:

Saroglitazar is described e.g. in Agrawal R. Curr Drug Targets. 2014 Leb;l5(2): l5 l-5. and is represented by the structural formula indicated below:

Naveglitazar is described e.g. in Ahlawat P, Srinivas NR. Eur J Drug Metab

Pharmacokinet. 2008 Jul-Sep;33(3): 187-90. GW501516 is described e.g. in Wang X, Sng MK, Poo S, Chong HC, Lee WL, Tang MB, Ng KW, Luo B, Choong C, Wong MT, Tong BM, Chiba S, Loo SC, Zhu P, Tan NS. J Control Release. 2015 Jan 10;197: 138-47 and is represented by the structural formula indicated below:

GFT505 is described e.g. in Cariou B, Staels B. Expert Opin Investig Drugs. 2014 Oct;23(lO): 1441-8 and is represented by the structural formula indicated below:

INT131 is described e.g. in. Taygerly JP, McGee LR, Rubenstein SM, Houze JB, Cushing TD, Li Y, Motani A, Chen JL, Frankmoelle W, Ye G, Learned MR, Jaen J, Miao S, Timmermans PB, Thoolen M, Kearney P, Flygare J, Beckmann H, Weiszmann J, Lindstrom M, Walker N, Liu J, Biermann D, Wang Z, Hagiwara A, Iida T, Aramaki H, Kitao Y, Shinkai H, Furukawa N, Nishiu J, Nakamura M. Bioorg Med Chem. 2013 Feb 15;21(4):979-92 and is represented by the structural formula indicated below:

PPAR activation by the PPAR agonist is usually strong in the low nano molar range to low micromolar range, e.g in a range of 0.1 nM to 100 mM. In some embodiments the PPAR activation is weak or partial, i.e. a PPAR agonist is used in the oral ER pharmaceutical compositions of the present invention which yields maximal activation of PPAR-receptor in a reporter assay system of 10% to 100% compared to a reference PPAR agonist which is known to cause a maximum PPAR activation. The preferred target for interaction of the PPAR agonist is the hair cell, which is most preferred; neural cells; and endothelial cells; and further includes adipocytes, hepatocytes, immune cells such as e.g. macrophages or dendritic cells, or skeletal muscle cells.

The term "hearing loss" which is used herein interchangeably with the term“hearing impairment” refers to a diminished sensitivity to the sounds normally heard by a subject. The severity of a hearing loss is categorized according to the increase in volume above the usual level necessary before the listener can detect it. The term "hearing loss" as used herein includes sudden hearing loss (SHL) which is indicated in the literature also as“sudden sensorineural hearing loss (SSHL)”. SHL refers to illness which is characterized by a sudden, rapid sensorineural hearing loss mostly in one ear without obvious causes, normally accompanied with dizziness, and without vestibular symptomatology. SHL is defined as greater than 30 dB hearing reduction, over at least three contiguous frequencies, occurring over a period of 72 hours or less. SHL can be caused e.g. by unspecific stress.

Hearing loss as referred herein is defined as a diminished ability to hear sounds like other people do. This can be caused either by conductive hearing loss, sensorineural hearing loss or a combination of both.

Conductive hearing loss means that the vibrations are not passing through from the outer ear to the inner ear, specifically the cochlea. It can be due to an excessive build-up of earwax, glue ear, an ear infection with inflammation and fluid buildup, a perforated or defective eardrum, or a malfunction of the ossicles (bones in the middle ear).

Sensorineural hearing loss is caused by dysfunction of the inner ear, the cochlea, auditory nerve, or brain damage. Usually, this kind of hearing loss is due to damage of the hair cells in the cochlea.

Hearing loss as referred herein is usually sensorineural hearing loss or a combination of conductive hearing loss and sensorineural hearing loss. Sensorineural hearing loss can be related to age, to an acute or constant exposure to noise or chemicals, to a brain trauma or non specific stress which may lead to sudden hearing loss.

The term "hair cell degeneration" as used herein refers to a gradual loss of hair cell function and integrity and/or leading ultimately to hair cell death.

The term "hair cell death" as used herein refers to apoptosis of the hair cells in the inner ear.

The terms "identification of hair cell damage" or "detection of hair cell damage" which are used interchangeably herein refer to a method by which the degree of hair cell damage in the inner ear can be determined. Such methods are known in the art and comprise for example fluorescent imaging of the hair cells, as described in the examples. An audiogram that demonstrates loss of hearing sensitivity at moderate to high frequencies is also indicative of hair cell damage. A decrease of hearing potential with no subsequent recovery is also diagnostic of hair cell damage. The term“chemically induced hearing loss” or“hearing loss induced by a chemical” as referred herein refers to hearing loss which is induced and/or caused by chemical agents such as solvents, gases, paints, heavy metals, and/or medicaments which are ototoxic.

The term sound pressure level (SPL) or acoustic pressure level as referred herein is a logarithmic measure of the effective sound pressure of a sound relative to a reference value. Sound pressure level, denoted L p and measured in dB, above a standard reference level, is given by:

Lp = 10 logio (Prms 2 /p 0 2 ) = 20 logio (Prms/po) dB(SPL) where p rms is the root mean square sound pressure, measured in Pa and po is the reference sound pressure, measured in Pa. The commonly used reference sound pressure in air is po = 20 pPa (Root Mean Squared) or 0.0002 dynes/cm 2 , which is usually considered the threshold of human hearing.

The term "individual," "subject" or "patient" are used herein interchangeably. In certain embodiments, the subject is a mammal. Mammals include, but are not limited to primates (including human and non- human primates). In a preferred embodiment, the subject is a human.

The term "about" as used herein refers to +/- 10% of a given measurement.

The term "solubility" as used herein refers to simplified descriptive solubilities in water in accordance with the US Pharmacopoeia, Chapter“General Notices”, § 5.30 "Description and Solubility" (see table below).

Parts of Solvent Required

Descriptive Term for 1 Part of Solute

Very soluble Less than 1

Freely soluble From 1 to 10

Soluble From 10 to 30

Sparingly soluble From 30 to 100

Slightly soluble From 100 to 1,000

Very slightly soluble From 1 ,000 to 10,000 Practically insoluble, or Greater than or equal to

Insoluble 10,000

Preferably, the term "water-soluble" as used herein usually refers to a solubility of a given compound in water of less than 1 part of solvent required for 1 part of solute to 30 parts of solvent required for 1 part of solute. Preferably, the term "water-insoluble" as used herein usually refers to a solubility of a given compound in water of more than 30 parts of solvent required for 1 part of solute to greater than or equal to 10,000 parts of solvent required for 1 part of solute.

The term "hydrophilic polymer" as used herein refers to a water-soluble or water- swellable polymer or copolymer comprising one or more hydrophilic chemical groups or moieties. Examples of hydrophilic chemical groups include, but are not limited to, chemical groups capable of forming hydrogen bonds, for example, carboxylate groups, amide groups, hydroxyl groups, amine groups, guanidino groups, sulfate groups and phosphate groups. Hydrophilic polymers comprise linear, branched, forked, branched- forked, dendrimeric, multi-armed, cross-linked or star-shaped polymers including, but not limited to, cellulosic polymers, poly(hydroxyalkanol methacrylates) (MW 5 kD - 5 MD), acrylic acid derivatives, alginates, anionic and cationic hydrogels, polyvinyl alcohols having a low acetate residual, pectins (MW 30 kD - 300 kD), polyethylene oxides, polysaccharides, Carbomer (polyacrylate polymers), polyacrylamides, natural gums and diesters of polyglucan. Examples of polysaccharides include agar, acacia, algins and scleroglucan. Examples of polyethylene oxides include Polyox® (Dow Chemical Company). Examples of alginates include ammonium alginate, sodium, calcium and/or potassium alginate, propylene glycol alginate. Examples of natural gums include gum Arabic (e.g. gum Arabic powder), gum karaya, locust bean gum, gum tragacanth, carrageens, guar gum and xanthan gum. Furthermore, hydrophilic polymers include, but are not limited to, polyvinyl alcohol, crosslinked polyvinyl alcohol, crosslinked poly N-vinyl-2-pyrrolidone, crospovidone, methyl cellulose, carboxymethyl cellulose (carmellose), cross-linked carboxymethyl cellulose (croscarmellose), sodium or calcium carboxymethyl cellulose, sodium or calcium croscarmellose, hydroxypropyl methyl cellulose (hypromellose), hydroxypropyl cellulose, hydroxyethyl cellulose, ethyl

hydroxyethylcellulose, nitro cellulose, methyl cellulose, ethyl cellulose, methyl ethyl cellulose, cellulose acetate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, cellulose butyrate, cellulose propionate, gelatin, collagen, maltodextrin, pullulan, polyvinyl acetate, polyvinyl acetate phthalate, polyacrylic acid and a swellable mixture of agar and carboxymethyl cellulose. Preferably, the molecular weight of the hydrophilic polymer ranges from about 10,000 Da to about 8,000,000 Da.

The term "excipient" as used herein refers to an agent comprised by the oral ER pharmaceutical composition of the invention and may include binders, lubricants, glidants, fillers and the like. The term "multiparticulate" as used herein refers to a state of matter which is characterized by the presence of a plurality of discrete or aggregated, particles, pellets, beads, granules, small tablets or mixtures thereof irrespective of their shape or morphology. The term “multiparticulate” includes every subunit of a size smaller than 5 mm, e.g. pellets, granules, sugar seeds (non-pareil), mini-tablets, powders, and crystals, with drugs being entrapped in or layered around cores. Accordingly, a "multiparticulate composition" is, for example a pellet composition, a bead composition, a granule composition, a powder composition or a small tablet composition ("mini-tablet" composition).

The term "capsule" as used herein refers to either a rigid, hard shell or a soft, pliable container that serves as a vehicle for solid pharmaceutical compositions, such as

multiparticulate compositions. Most capsules are made form gelatin derived from hydrolyzed animal collagen but as used herein, non-animal sources are also included. Capsules are often formed from two separate halves sealed together, but alternatively may be a one-piece form that is filled by injection after suspending in an appropriate liquid, e.g. medium-chain triglycerides and subsequently sealed. As used herein, the term "capsule" includes commonly used terms such as Gelcaps™, Softgel™ and Soft- gel™.

The term "hard capsule" as used herein refers to any type of hard capsule made from gelatin or a different material, e.g. hydroxypropyl methylcellulose (hypromellose) and gellan gum (Vcaps™) or pullulan and carrageenan (NPcaps™).

The term "enteric coating" as used herein refers to a barrier applied on oral medication that prevents its dissolution or disintegration in the gastric environment.

The term "colloidal silicon dioxide"“or colloidal silica” which are used

interchangeably, refers to Si0 2 particles in the micro-meter to nano-meter size range.

Colloidal silica is described in the United States Pharmacopeia - National Formulary

(USP/NF) under the designation Colloidal Silicon Dioxide. Colloidal silicon dioxide comprise anhydrous silicic acid containing hydrated silicon dioxide (Si0 2 nH 2 0, wherein n is an integer) as a main component, and examples thereof include Sylysia 320 (trade name, Fuji Silysia Chemical Ltd.), AEROSIL 200 (Evonik Industries), Cab-O-Sil (Cabot Corporation), and the like. Colloidal silicon dioxide comprises submicroscopic silica prepared by the vapor- phase hydrolysis of a silicon compound. Colloidal silicon dioxide also includes light anhydrous silicic acid and/or fumed silica. Light anhydrous silicic acid as used herein is preferably light anhydrous silicic acid without hydrophobizing treatment (example with hydrophobizing treatment: Silica, hydrophobic colloidal NF; AEROSIL® R972 (Evonik Industries)) or amorphous silica fineparticle with a particle size greater than 0.1 micron. The term“fumed silica”, also known as pyrogenic silica because it is produced in a flame, refers to microscopic droplets of amorphous silica fused into branched, chainlike, three-dimensional secondary particles which then agglomerate into tertiary particles. The resulting powder has an extremely low bulk density and high surface area. Its three-dimensional structure results in viscosity- increasing, thixotropic behavior when used as a thickener or reinforcing filler.

Fumed silica has a very strong thickening effect. Primary particle size is 5-50 nm. The particles are non-porous and have a surface area of 50-600 m2/g. The density is 160-190 kg/m 3 . Examples thereof include AEROSIL 200 (Evonik Industries).

The term "cellulosic polymer" as used herein refers to a class of polymers derived from cellulose such as cellulose ethers, e.g. hydroxyalkyl celluloses and alkyl celluloses and cellulose esters. Examples of hydroxyalkyl celluloses include hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (hypromellose, HPMC), hydroxyethyl methyl cellulose (HEMC), ethyl hydroxyethyl cellulose (EHEC) and hydroxyethyl cellulose (HEC). Examples of alkyl celluloses include methyl cellulose and ethyl cellulose. The group of cellulose ethers further comprises carboxymethyl cellulose (carmellose), carboxymethyl hydroxyethyl cellulose (CMHEC) and sodium or calcium carmellose. Examples of cellulose esters include nitro cellulose, cellulose acetate, cellulose butyrate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate and cellulose acetate trimellitate. A cellulosic polymer may also be both a cellulosic ether and a cellulosic ester, e.g. hydroxypropyl methylcellulose phthalate. The term "cellulosic polymer" as used herein further comprises cross-linked cellulosic polymers such as cross-linked carboxymethyl cellulose (croscarmellose) and sodium or calcium croscarmellose.

When the cellulosic polymer is HPC, said HPC is preferably selected from the group consisting of HPC-SSL, HPC-SL and HPC-L.

When the cellulosic polymer is hypromellose (hydroxypropyl methylcellulose

(hypromellose, HPMC), said hypromellose is preferably selected from the group consisting of hypromellose type 2910 (Methocel™ E-types, Premium quality, Dow Chemical Company) and hypromellose type 2208 (Methocel™ K-types, Premium quality, Dow Chemical

Company), especially Methocel™Kl5M or Methocel™K4M, in particular Methocel™Kl5M with a viscosity of 11,250-21,000 mPa.s (2% in water at 20° C); Dow Chemical Company).

The term "monocalcium phosphate" is synonymous to "calcium dihydrogen phosphate" and refers to the inorganic compound with the formula Ca(H 2 P0 4 ) 2 . The term "dicalcium phosphate" is synonymous to "calcium hydrogen phosphate" and "dibasic calcium phosphate" and refers to the inorganic compound with the formula CaHP0 4 .

The term "tricalcium phosphate" is synonymous to "calcium phosphate" and refers to the inorganic compound with the formula Ca3(P0 4 ) 2 .

The term "non- functional film-coating" as used herein refers to a film-coating comprising polyvinyl alcohol (PVA) and/or hypromellose, titanium dioxide, polyethylene glycol (PEG) and talc, such as Opadry® II (Colorcon). Usually non- functional film-coatings of the present invention do not comprise an API i.e. usually non- functional film-coatings of the present invention do not comprise a PPAR agonist. Typically, non- functional film- coatings are applied to improve the patient compliance by better appearance and

distinguishability, easier intake and swallowing of the oral ER pharmaceutical compositions of the invention and to protect the dosage form against environmental influences.

The term "mini-tablets coating" as used herein refers to a coating which can be used to mask the bad taste (e.g. a mixture of Surelease® (i.e. a coating comprising an aqueous ethylcellulose dispersion) and Opadry® II, Colorcon) or smell of a product, to protect the API (active pharmaceutical ingredient) against the acid environment of the stomach or the gastric mucosa against an aggressive API. A large part of functional coatings lead to prolonged release of the API [Knop & Kleinbudde, Int. J. Pharmac. 457 (2), 527-536 (2013)].

The term "polyvinylpyrrolidone" also mentioned“povidone” herein as used herein refers to a polymer consisting essentially of linear l-vinyl-2-pyrrolidinone groups, the degree of polymerization of which results in polymers of various molecular weights. The different types of povidone are characterized by their viscosity in aqueous solution, relative to that of water, expressed as a K-value. Examples include e.g. Plasdone K-29/32 or Povidone K90.

The term "crospovidone" as used herein refers to a water-insoluble synthetic cross- linked homopolymer of N-vinyl-2-pyrrolidinone.

The term "glidant" as used herein refers to an excipient promoting the flow of tablet granulation or powder materials by reducing friction between the particles of said tablet granulation or powder materials. Examples of glidants include fumed silica, light anhydrous silicic acid such as fine particle anhydrous silicic acids (light anhydrous silicic acids without hydrophobizing treatment or amorphous silica fine particles with particle size of not more than 0.1 micron) and talc. Preferred glidants are fumed silica and light anhydrous silicic acids, such as e.g. Sylysia 320 (trade name, Fuji Silysia Chemical Ltd.), Aerosil® 200 (trade name, Evonik Industries) and the like and talc. Particularly preferred glidants are Aerosil® 200 and/or talc.

The term "lubricant" as used herein refers to an excipient reducing friction encountered during ejection of tablets between the walls of the tablets and the die cavity. Such excipients include, by way of example and without limitation, stearic acid and pharmaceutically acceptable salts thereof, sucrose esters of fatty acids and waxes. Examples of

pharmaceutically acceptable salts of stearic acid include magnesium stearate, calcium stearate and sodium stearyl fumarate. Further examples of lubricants include DL-leucine, sodium lauryl sulfate and magnesium lauryl sulfate.

The term "binder" as used herein refers to pharmaceutically acceptable excipients used to cause adhesion of powder particles in solid dosage formulations. Such excipients include, by way of example and without limitation, polyvinylpyrrolidones such as povidone and other materials known to one of ordinary skill in the art. Further examples include starch (in form of starch paste); starch (in form of Prejel® pre-gelatinized starch); gelatine; Methyl Cellulose (in form of Methocel® MC, Tylose®) and Ethyl Cellulose (in form of Ethocel®).

The term "filler" as used herein refers to pharmaceutically acceptable excipients which are added to increase the bulk of a solid pharmaceutical composition and may make a pharmaceutical dosage form containing the composition easier for the patient and/or a care giver to handle. The term "filler" as used herein is intended to comprise water-soluble and water-insoluble fillers.

Examples of water-insoluble fillers include starch, cereal flour containing starch (e.g. com starch), cellulose and calcium phosphates. Examples of starch include com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch and sodium carboxymethyl starch. Examples of cellulose include crystalline cellulose and microcrystalline cellulose like microcrystalline cellulose Vivapur® 101 (JRS Pharma).

Examples of calcium phosphates include monocalcium phosphate, dicalcium phosphate, such as dibasic calcium phosphate dihydrate (USP) (EMCOMPRESS®), anhydrous dibasic calcium phosphate (USP) (EMCOMPRESS® Anhydrous) (both by JRS Pharma) and Di- Cafos (dibasic calcium phosphate by Budenheim) like Di-Cafo D9 (Buddenheim) and tricalcium phosphate.

Examples of water-soluble fillers include sugars and sugar alcohols, water-soluble polymers, surfactants, inorganic salts, organic acids, amino acids and amino sugars. Examples of sugars and sugar alcohols include lactose like lactose monohydrate (e.g. lactose monohydrate by GanuLac®, Meggle Pharma), glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D-fructose, dextran, and the like. Examples of water- soluble polymers include polyethylene glycols (e.g., macrogol 400, macrogol 1500, macrogol 4000, macrogol 6000, macrogol 20000). Examples of surfactants include polyoxyethylene hydrogenated castor oil (e.g., Cremophor RH40, HCO-40, HCO-60),

polyoxyethylenepolyoxypropyleneglycol (e.g., pluronic F68) or sorbitan polyoxyethylene higher fatty acid ester (e.g., Tween80) and the like. Examples of inorganic salts include sodium chloride, magnesium chloride and the like. Examples of organic acids include citric acid, tartaric acid and the like. Examples of amino acids include glycine, b-alanine, lysine hydrochloride and the like. Examples of amino sugars include meglumine and the like.

In one aspect, the present invention provides an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating hearing loss in a subject. In a further aspect, the present invention provides a method of preventing or treating hearing loss in a subject, which method comprises administering to the subject an oral ER pharmaceutical composition comprising a PPAR agonist. In some embodiments the oral ER pharmaceutical composition comprising a PPAR agonist is administered to the subject in an amount that is sufficient to prevent or treat hearing loss in the subject. In a further aspect the present invention provides the use of an oral ER pharmaceutical composition comprising a PPAR agonist for the manufacture of a medicament for preventing or treating hearing loss in a subject.

In a further aspect, the present invention provides the use of an oral ER pharmaceutical composition comprising a PPAR agonist for preventing or treating hearing loss in a subject.

In a further aspect, the present invention provides a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating hearing loss in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and

administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject;

wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR

agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In a further aspect, the present invention provides a method of preventing or treating hearing loss in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and

administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject; wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

In a further aspect the present invention provides the use of a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for the manufacture of a medicament for preventing or treating hearing loss in a subject, wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In a further aspect, the present invention provides the use of a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for preventing or treating hearing loss in a subject, wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

In some preferred embodiments, hearing loss to be prevented or treated by the oral ER pharmaceutical compositions of the present invention is caused by a noise trauma, by a medical intervention, by ischemic injury, by age or is chemically induced. The hearing loss can be thus a consequence of a medical intervention such as e.g. cochlear implantation. The chemical induction is usually caused by a chemical agent e.g. by an antibiotic or a

chemotherapeutic agent. In some preferred embodiments, hearing loss is sudden hearing loss. Hearing loss caused by age comprises e.g. presbycusis. Preferably hearing loss caused by a noise trauma, cochlear implantation, or which is chemically induced, preferably by an antibiotic, is prevented or treated by the oral ER pharmaceutical compositions of the present invention. More preferably, hearing loss caused by a noise trauma or which is chemically induced, preferably by an antibiotic, is prevented or treated by the methods of the present invention. In some embodiments, hearing loss is of sensorineural origin caused by a damage leading to malnutrition of the cells in early brain development. In this case, early treatment with a PPAR agonist can be disease modifying, preventing further damage.

In some embodiments, the oral ER pharmaceutical composition comprising a PPAR agonist is administered before the subject has developed or before it is at risk to develop hearing loss, hair cell degeneration, hair cell death and/or a condition characterized by hair cell damage. In some embodiments, the oral ER pharmaceutical composition comprising a PPAR agonist is administered after the subject has acquired hearing loss, hair cell

degeneration, hair cell death and/or a condition characterized by hair cell damage.

In some embodiments, the oral ER pharmaceutical composition of the invention is administered orally to a subject before the subject has developed or before it is at risk to develop hearing loss, hair cell degeneration, hair cell death and/or a condition characterized by hair cell damage and a PPAR agonist is administered intratympanically after the subject has acquired hearing loss, hair cell degeneration, hair cell death and/or a condition

characterized by hair cell damage.

In some embodiments, the oral administration of the ER pharmaceutical composition of the invention and the intratympanical administration of a PPAR agonist are carried out simultaneously to a subject after the subject has acquired hearing loss, hair cell degeneration, hair cell death and/or a condition characterized by hair cell damage.

Further diseases, disorders or conditions which are related to, caused or characterized by hair cell degeneration and/or hair cell death and which can be prevented or treated by the oral ER pharmaceutical compositions of the present invention are e.g. Meniere's disease, acute peripheral vestibulopthy and tinnitus.

Thus, in some embodiments the present invention provides an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject, wherein hair cell degeneration or hair cell death is related to and/or caused by Meniere's disease, acute peripheral vestibulopthy and/or tinnitus.

In some embodiments, the present invention provides an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating Meniere's disease in a subject. In some embodiments, the present invention provides a method of preventing or treating Meniere's disease in a subject which method comprises administering to the subject an oral ER pharmaceutical composition comprising a PPAR agonist.

In some embodiments, the present invention provides the use of an oral ER

pharmaceutical composition comprising a PPAR agonist for the manufacture of a medicament for preventing or treating Meniere's disease in a subject.

In some embodiments, the present invention provides the use of an oral ER

pharmaceutical composition comprising a PPAR agonist for preventing or treating Meniere's disease in a subject.

In some embodiments, the present invention provides an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating acute peripheral vestibulopthy in a subject.

In some embodiments, the present invention provides a method of preventing or treating acute peripheral vestibulopthy in a subject which method comprises administering to the subject an oral ER pharmaceutical composition comprising a PPAR agonist.

In some embodiments, the present invention provides the use of an oral ER

pharmaceutical composition comprising a PPAR agonist for the manufacture of a medicament for preventing or treating acute peripheral vestibulopthy in a subject.

In some embodiments, the present invention provides the use of an oral ER

pharmaceutical composition comprising a PPAR agonist for preventing or treating acute peripheral vestibulopthy in a subject.

In some embodiments, the present invention provides an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating tinnitus in a subject.

In some embodiments, the present invention provides a method of preventing or treating tinnitus in a subject which method comprises administering to the subject an oral ER pharmaceutical composition comprising a PPAR agonist.

In some embodiments, the present invention provides the use of an oral ER

pharmaceutical composition comprising a PPAR agonist for the manufacture of a medicament for preventing or treating tinnitus in a subject.

In some embodiments, the present invention provides the use of an oral ER

pharmaceutical composition comprising a PPAR agonist for preventing or treating tinnitus in a subject. Hearing loss, hair cell degeneration or hair cell death caused by a noise trauma or by medical intervention

Exposure to loud noise causes noise-induced hearing loss (NIHL) by damaging the organs of Corti. Damage by NIHL depends upon both the level of the noise and the duration of the exposure. Hearing loss may be temporary (temporary threshold shift, TTS) if a repair mechanism is able to restore the organ of the Corti. However, it becomes permanent (permanent threshold shift, PTS) when hair cells or neurons die. Structural modifications correlated to noise trauma are of two types: (1) mild damage of synapses and or hair cell stereocilia which can be repaired by cellular repair mechanisms and accounts for TTS and recovery and (2) severe damage inducing hair cell and neuronal apoptosis which can not be repaired by cellular repair mechanisms and accounts for PTS.

A noise trauma as referred herein is a noise trauma which is sufficient to cause damage to the organs of corti, in particular a noise trauma causing temporary or permanent hearing loss. A noise trauma can be caused by exposure to a sound pressure level of e.g., at least 70 dB (SPL), at least 90 dB (SPL), at least 100 dB (SPL), at least 120 dB (SPL) or at least 130 dB (SPL).

Hearing loss can also be caused by a medical intervention usually by a medical intervention in the ear e.g. by cochlea surgery such as cochlear implantation.

In some embodiments the oral ER pharmaceutical composition of the invention is administered before the subject is exposed to a noise trauma or medical intervention. In some embodiments, the oral ER pharmaceutical composition of the invention is administered after the subject is exposed to a noise trauma or medical intervention. In a particular embodiment the oral ER pharmaceutical composition of the invention is administered prior to cochlear surgery i.e. before the subject undergoes cochlear surgery.

In some embodiments a PPAR agonist is injected intratympanically to a subject before the subject is exposed to a noise trauma or medical intervention and the oral ER

pharmaceutical composition of the invention is administered to said subject while or after the subject is exposed to a noise trauma or medical intervention. In a particular embodiment, a PPAR agonist is injected intratympanically to a subject prior to cochlear surgery i.e. before the subject undergoes cochlear surgery and the oral ER pharmaceutical composition of the invention is administered to said subject after the subject has undergone cochlear surgery. Hearing loss, hair cell degeneration or hair cell death caused by age

Hearing loss caused by age also referred in the literature as“age-related hearing loss” is the cumulative effect of aging on hearing. It is normally a progressive bilateral symmetrical age-related sensorineural hearing loss. The hearing loss is most marked at higher frequencies.

There are four pathological types of hearing loss caused by age:

1) sensory: characterised by degeneration of organs of corti. 2) neural: characterised by degeneration of cells of spiral ganglion. 3) strial/metabolic: characterised by atrophy of stria vascularis in all turns of cochlea. 4) cochlear conductive: due to stiffening of the basilar membrane thus affecting its movement.

Hearing loss caused by age to be prevented or treated by the methods of the present invention is usally related to the first pathological type i.e. hearing loss characterised by degeneration of the organ of Corti. Thus, in some embodiments the oral ER pharmaceutical composition according to the invention is administered to the subject prior to degeneration of the organ of Corti, e.g. prior to damage or apoptosis of hair cells and/or prior to hair cell degeneration or hair cell death.

Chemically induced hearing loss, hair cell degeneration or hair cell death

Hearing loss, hair cell degeneration or hair cell death can be induced chemically i.e. by a chemical agent e.g. by an antibiotic, a drug, a chemotherapeutic agent, heavy metals or organic agents. Antibiotics which may cause hearing loss include for example cephalosporins such as cephalexin (Keflex), cefaclor (Ceclor), and cefixime (Suprax); aminoglycosides such as gentamicin, tobramycin and streptomycin; macrolides, such as erythromycin, azithromycin (Zithromax) and clarithromycin; sulfonamides such as trimethoprim-sulfamethoxazole or tetracylines such as tetracycline, or doxycycline. In particular hearing loss, hair cell degeneration or hair cell death is effectively prevented or treated by the methods of the present invention in a subject exposed to gentamicin.

Chemotherapeutic agents, e.g. anti-cancer agents which may cause hearing loss, hair cell degeneration or hair cell death include for example platinum-containing agents e.g.

cisplatin, and carboplatin, preferably cisplatin. Drugs which may cause hearing loss, hair cell degeneration or hair cell death include for example furosemide, quinine, aspirin and other salicylates. Heavy metals which may cause hearing loss include for example mercury, lead. Organic agents which may cause hearing loss, hair cell degeneration or hair cell death include for example toluene, xylene, or styrene. In some embodiments, the oral ER pharmaceutical composition of the invention is administered to the subject before the subject is exposed to a chemical agent, thereby preventing the subject from chemically induced hearing loss, hair cell degeneration or hair cell death. In some embodiments, the oral ER pharmaceutical

composition of the invention is administered to the subject after the subject is exposed to a chemical agent thereby treating the subject having chemically induced hearing loss, hair cell degeneration or hair cell death.

In a preferred embodiment, when hearing loss is caused by a noise trauma or is chemically induced, the oral ER pharmaceutical composition of the invention is administered to the subject prior to exposure of the subject to the noise trauma or to the chemical wherein at least 50%, preferably at least 60%, more preferably at least 70%, in particular at least 80%, more particular at least 90% of the cell damage of the hair cells caused by the noise trauma or the chemical agent is prevented.

In one aspect of the invention, the present invention provides an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject. In a further aspect of the invention, the present invention provides a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject, which method comprises administering to the subject an oral ER

pharmaceutical composition comprising a PPAR agonist. In some embodiments, the oral ER pharmaceutical composition of the invention is administered to the subject in an amount that is sufficient to prevent or inhibit hair cell degeneration or hair cell death in the subject. In a further aspect, the present invention provides the use of an oral ER pharmaceutical

composition comprising a PPAR agonist for the manufacture of a medicament for preventing or inhibiting hair cell degeneration or hair cell death in a subject. In a further aspect, the present invention provides the use of an oral ER pharmaceutical composition comprising a PPAR agonist for preventing or inhibiting hair cell degeneration or hair cell death in a subject. In a further aspect, the present invention provides a PPAR agonist and an oral ER

pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or inhibting hair cell degeneration or hair cell death in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and

administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject;

wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

In a further aspect, the present invention provides a method of preventing or inhibting hair cell degeneration or hair cell death in a subject, comprising:

administering a PPAR agonist by intratympanic injection to said subject; and

administering orally an ER pharmaceutical composition comprising a PPAR agonist to said subject;

wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

In a further aspect the present invention provides the use of a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for the manufacture of a medicament for preventing or inhibting hair cell degeneration or hair cell death in a subject, wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In a further aspect, the present invention provides the use of a PPAR agonist and an oral ER pharmaceutical composition comprising a PPAR agonist for preventing or inhibting hair cell degeneration or hair cell death in a subject, wherein wherein the PPAR agonist is injected intratympanically before, simultaneously or after the oral administration of the ER pharmaceutical composition comprising a PPAR agonist. In some preferred embodiments, the PPAR agonist is injected intratympanically before or simultaneously to the oral administration of the ER pharmaceutical composition comprising a PPAR agonist.

In some embodiments, hair cell degeneration or hair cell death in a subject is caused by a noise trauma, by age, a medical intervention, sudden hearing loss, or ischemic events such as ischemic injury, or is chemically induced wherein the chemical induction is caused by e.g. an antibiotic or a chemotherapeutic agent. Noise trauma, age, a medical intervention, sudden hearing loss, or ischemic events, or chemical induction can cause hair cell degeneration or hair cell death in a subject as described above for methods or preventing or treating hearing loss.

In some embodiments, hearing loss, hair cell degeneration or hair cell death is caused by hair cell damage. In some embodiments, the oral ER pharmaceutical composition of the invention is administered to the subject prior to identification of said hair cell damage i.e. prior to occurrence of hair cell damage. In a preferred embodiment, when hair cell damage is caused by a noise trauma or is chemically induced, the oral ER pharmaceutical composition of the invention is administered to the subject prior to exposure of the subject to the noise trauma or to the chemical agent wherein at least 50%, preferably at least 60%, more preferably at least 70%, in particular at least 80%, more particular at least 90% of the cell damage of the hair cells caused by the noise trauma or the chemical agent is prevented.

In some embodiments, a PPAR agonist is injected intratympanically to a subject prior to identification of hair cell damage, i.e. prior to occurrence of hair cell damage and the oral ER pharmaceutical composition of the invention is administered to said subject after hair cell damage has been identified, i.e. after hair cell damage has occurred.

In a preferred embodiment, when hair cell damage is caused by a noise trauma or is chemically induced, a PPAR agonist is injected intratympanically to a subject prior to exposure of the subject to the noise trauma or to the chemical agent and the oral ER pharmaceutical composition of the invention is administered to said subject after exposure of the subject to the noise trauma or to the chemical agent.

Identification/occurrence of hair cell damage is usually determined by evaluation of the state of the hair cells which can be easily accomplished as described above or as disclosed in the examples.

Oral Extended Release Pharmaceutical compositions

In one embodiment, the oral ER pharmaceutical composition of the invention is a single unit dosage form or a multiple unit dosage form.

The term "single unit" denotes that the oral ER pharmaceutical composition contains the PPAR agonist homogenously distributed over the composition and the term "multiple unit" denotes that the PPAR agonist is present in several discrete units within the composition.

As used herein, the term "unit dose" refers to the dose of PPAR agonist to be

administered to a subject at a time, each unit containing a predetermined quantity of PPAR agonist calculated to produce the desired therapeutic effect. Preferably, the subject is administered one unit dose of PPAR agonist per day for preventing or treating hearing loss and/or for preventing or inhibiting hair cell degeneration or hair cell death.

Accordingly, in a preferred embodiment, there is provided an oral ER pharmaceutical composition comprising a PPAR agonist for use in a method of preventing or treating hearing loss in a subject or for use in a method of preventing or inhibiting hair cell degeneration or hair cell death in a subject, wherein said oral ER pharmaceutical composition is a single unit dosage form and wherein said oral ER pharmaceutical composition is administered to the subject once-daily.

In one embodiment, the oral ER pharmaceutical composition according to the invention is a multiparticulate composition, such as a pellet composition, a bead composition, a granule composition, a powder composition or a small tablet composition ("mini-tablet" composition).

In a preferred embodiment, the oral ER pharmaceutical composition according to the invention is a powder composition. Preferably, a powder composition according to the invention is suitable for the manufacture of tablets, such as film-coated tablets.

In one embodiment, the oral ER pharmaceutical composition according to the invention is a multiparticulate composition, wherein the particles comprised by said multiparticulate composition are film-coated particles.

As indicated above, the oral ER pharmaceutical composition according to the invention may be a fim-coated tablet or a film-coated capsule. Furthermore, when the oral ER pharmaceutical composition according to the invention is a multiparticulate composition, the particles comprised by said multiparticulate composition may be film-coated particles. Film- coatings for multiparticulate composition are preferably functional film-coatings, such as e.g. Surelease® Ethylcellulose Dispersion Type B NF. In one embodiment, said film-coating is an ER film-coating, such as an enteric coating. When the film-coating is an enteric coating, it preferably comprises a gastric resistant polymer, such as cellulose acetate phthalate (CAP), hydroxypropyl methyl cellulose phthalate (HPMCP); methacrylic acid copolymer, Type A (NF) (EUDRAGIT® L 100; anionic copolymers based on methacrylic acic and methyl methacrylate); methacrylic acid copolymer, Type B (NF) (EUDRAGIT® S 100, anionic copolymers based on methacrylic acic and methyl methacrylate) and methacrylic acid copolymer dispersion (NF) (EUDRAGIT® L 30 D-55, aqueous dispersion of anionic polymers with methacrylic acid as a functional group).

In one embodiment, the oral ER pharmaceutical composition according to the invention is a reservoir-type ER composition comprising an ER film-coating. ER film-coatings for reservoir-type oral ER pharmaceutical composition according to the invention comprise, for example one or more excipients selected from the group consisting of cellulose acetate phthalate, cellulose acetate trimellitate, hydroxy propyl methylcellulose phthalate, polyvinyl acetate phthalate, ammonio methacrylate copolymers such as those sold under the trademark Eudragit® RS and RL, polyacrylic acid and polyacrylate and methacrylate copolymers such as those sold under the trademark Eudragit® S and L, polyvinyl acetaldiethylamino acetate, hydroxypropyl methylcellulose acetate succinate, shellac.

In one embodiment, the oral ER pharmaceutical composition according to the invention is a solid composition.

In one embodiment, the oral ER pharmaceutical composition according to the invention is a tablet, a film-coated tablet or a capsule.

In a preferred embodiment, the oral ER pharmaceutical composition according to the invention is a film-coated tablet, more preferably a film-coated tablet with a non- functional film-coating.

In a further preferred embodiment, the oral ER pharmaceutical composition according to the invention is a hard capsule, such as a gelatin capsule.

In a preferred embodiment, the oral ER pharmaceutical composition according to the invention is a film-coated capsule more preferably a film-coated capsule with a non- functional film-coating.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises an extended release matrix.

In a preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises an extended release matrix, wherein the extended release matrix is coated and wherein the coating does not comprise an API i.e. wherein the coating does not comprise a PPAR agonist.

In a further preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises an extended release matrix as core, wherein the core is coated and wherein the coating does not comprise an API i.e. wherein the coating does not comprise a PPAR agonist.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises an extended release matrix, wherein the extended release matrix is selected from the group consisting of disintegrating, non-disintegrating and eroding matrices, preferably eroding matrices. In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble and/or water-insoluble filler.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer, a water-soluble filler and a water-insoluble filler.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-insoluble filler selected from the group consisting of starch, cereal flour containing starch (e.g. com starch), cellulose and calcium phosphates.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-insoluble filler being starch, wherein said starch is selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch and sodium carboxymethyl starch.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-insoluble filler being cellulose, wherein said cellulose is selected from the group consisting of crystalline cellulose and microcrystalline cellulose.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-insoluble filler being a calcium phosphate, wherein said calcium phosphate is selected from the group consisting of monocalcium phosphate, dicalcium phosphate and tricalcium phosphate, preferably dicalcium phosphate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-insoluble filler selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, sodium carboxymethyl starch, microcrystalline cellulose, crystalline cellulose, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate.

In a preferrred embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-insoluble filler selected from the group consisting of microcrystalline cellulose, dicalcium phosphate and mixtures thereof.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler selected from the group consisting of sugars and sugar alcohols, water-soluble polymers, surfactants, inorganic salts, organic acids, amino acids and amino sugars.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being a sugar or a sugar alcohol, wherein said sugar or sugar alcohol is selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D-fructose, dextran and glucose.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being a water-soluble polymer, wherein said water- soluble polymer is selected from the group consisting of polyethylene glycols (e.g., macrogol 400, macrogol 1500, macrogol 4000, macrogol 6000, macrogol 20000).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being a surfactant, wherein said surfactant is selected from the group consisting of polyoxyethylene hydrogenated castor oil (e.g.,

Cremophor REMO, HCO-40, HCO-60), polyoxyethylenepolyoxypropyleneglycol (e.g., pluronic F68) and sorbitan polyoxyethylene higher fatty acid ester (e.g., Tween80).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being an inorganic salt, wherein said inorganic salt is selected from the group consisting of sodium chloride and magnesium chloride.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being an organic acid, wherein said organic acid is selected from the group consisting of citric acid and tartaric acid.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being an amino acid, wherein said amino acid is selected from the group consisting of glycine, b-alanine and lysine hydrochloride.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler being an amino sugar, wherein said amino sugar is meglumine.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D-fructose, dextran, polyethylene glycol (e.g., macrogol 400, macrogol 1500, macrogol 4000, macrogol 6000, macrogol 20000), polyoxyethylene hydrogenated castor oil (e.g., Cremophor RH40, HCO-40, HCO-60), polyoxyethylenepolyoxypropyleneglycol (e.g., pluronic F68), sorbitan polyoxyethylene higher fatty acid ester (e.g., Tween80), sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D-fructose, dextran, polyoxyethylene hydrogenated castor oil (e.g., Cremophor RH40, HCO-40, HCO- 60), polyoxyethylenepolyoxypropyleneglycol (e.g., pluronic F68), sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine.

In a preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises sugar as water-soluble filler. In a particularly preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises lactose as water- soluble filler, preferably lactose monohydrate.

The hydrophilic polymers for use in the oral ER pharmaceutical compositions of the invention are preferably cellulosic polymers, such as e.g. cellulose ethers and cellulose esters.

Thus, in one embodiment, the oral ER pharmaceutical composition according to the invention comprises a cellulosic polymer selected from the group consisting of cellulose ethers and cellulose esters, preferably selected from the group consisting of hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (hypromellose, HPMC), hydroxyethyl methyl cellulose (HEMC), ethyl hydroxyethyl cellulose (EHEC), hydroxyethyl cellulose (HEC), methyl cellulose, ethyl cellulose, carboxymethyl cellulose (carmellose),

carboxymethyl hydroxyethyl cellulose (CMHEC), sodium or calcium carmellose, nitro cellulose, cellulose acetate, cellulose butyrate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropyl methylcellulose phthalate, cross-linked carboxymethyl cellulose (croscarmellose) and sodium or calcium croscarmellose. A particularly preferred hydrophilic polymer for use in the oral ER

pharmaceutical compositions of to the invention is hydroxypropyl methylcellulose

(hypromellose). Thus, in a particularly preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises hydroxypropyl methylcellulose

(hypromellose) most preferably hydroxypropyl methylcellulose selected from the group consisting of hypromellose type 2910 (Methocel™ E-types, Premium quality, Dow Chemical Company) and hypromellose type 2208 (Methocel™ K-types, Premium quality, Dow

Chemical Company), preferably Methocel™Kl5M or Methocel™K4M (both premium quality, Dow Chemical Company),, more preferably Methocel™Kl5M with a viscosity of 11,250-21,000 mPa.s (2% in water at 20° C); Dow Chemical Company). In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer selected from the group consisting of linear, branched, forked, branched- forked, dendrimeric, multi-armed and star-shaped polymers.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer selected from the group consisting of cellulosic polymers, poly(hydroxyalkanol methacrylates) (MW 5 kD - 5 MD), acrylic acid derivatives, alginates, anionic and cationic hydrogels, polyvinyl alcohols having a low acetate residual, pectins (MW 30 kD - 300 kD), polyethylene oxides, polysaccharides, polyacrylate polymers,

polyacrylamides, natural gums and diesters of polyglucan.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer being a polysaccharide selected from the group consisting of agar, acacia, algins and scleroglucan.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer being a polyethylene oxide, wherein said polyethylene oxide is Polyox® (Dow Chemical Company).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer being an alginate selected from the group consisting of ammonium alginate, sodium, calcium and/or potassium alginate and propylene glycol alginate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer being a natural gum selected from the group consisting of gum Arabic (e.g. gum Arabic powder), gum karaya, locust bean gum, gum tragacanth, carrageens, guar gum, xanthan gum.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer selected from the group consisting of polyvinyl alcohol, crosslinked polyvinyl alcohol, crosslinked poly N-vinyl-2-pyrrolidone, crospovidone, methyl cellulose, carboxymethyl cellulose (carmellose), cross-linked carboxymethyl cellulose (croscarmellose), sodium or calcium carboxymethyl cellulose, sodium or calcium

croscarmellose, hydroxypropyl methyl cellulose (hypromellose), hydroxypropyl cellulose, hydroxyethyl cellulose, ethyl hydroxyethylcellulose, nitro cellulose, methyl cellulose, ethyl cellulose, methyl ethyl cellulose, cellulose acetate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, cellulose butyrate, cellulose propionate, gelatin, collagen, maltodextrin, pullulan, polyvinyl acetate, polyvinyl acetate phthalate, polyacrylic acid and a swellable mixture of agar and carboxymethyl cellulose.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer selected from the group consisting of agar, acacia, algins, polyethylene oxides (e.g. Polyox®, Dow Chemical Company), ammonium alginate, sodium, calcium and/or potassium alginate, propylene glycol alginate, gum Arabic (e.g. gum Arabic powder), gum karaya, locust bean gum, gum tragacanth, carrageens, guar gum, xanthan gum and scleroglucan.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises a hydrophilic polymer selected from the group consisting of polyvinyl alcohol, crosslinked polyvinyl alcohol, crosslinked poly N-vinyl-2-pyrrolidone, crospovidone, methyl cellulose, carboxymethyl cellulose (carmellose), cross-linked carboxymethyl cellulose (croscarmellose), sodium or calcium carboxymethyl cellulose, sodium or calcium

croscarmellose, hydroxypropyl methyl cellulose (hypromellose), hydroxypropyl cellulose, hydroxyethyl cellulose, ethyl hydroxyethylcellulose, nitro cellulose, methyl cellulose, ethyl cellulose, methyl ethyl cellulose, cellulose acetate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, cellulose butyrate, cellulose propionate, gelatin, collagen, maltodextrin, pullulan, polyvinyl acetate, polyvinyl acetate phthalate, polyacrylic acid and a swellable mixture of agar, carboxymethyl cellulose, agar, acacia, algins, polyethylene oxides (e.g. Polyox®, Dow Chemical Company), ammonium alginate, sodium, calcium and/or potassium alginate, propylene glycol alginate, gum Arabic (e.g. gum Arabic powder), gum karaya, locust bean gum, gum tragacanth, carrageens, guar gum, xanthan gum and scleroglucan.

In a preferred embodiment, said hydrophilic polymer is comprised by an ER matrix. Alternatively or additionally, said hydrophilic polymer may also be comprised by a coating, such as a tablet coating or a capsule coating. In a preferred embodiment, the oral ER pharmaceutical composition of the invention comprises a PPAR agonist and an ER matrix, wherein said ER matrix comprises a hydrophilic polymer, preferably a cellulosic polymer, more preferably a cellulosic polymer selected from the group consisting of cellulose ethers, e.g. hydroxyalkyl celluloses and alkyl celluloses and cellulose esters, even more preferably a cellulosic polymer selected from the group consisting of hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (hypromellose, HPMC), hydroxyethyl methyl cellulose (HEMC), ethyl hydroxyethyl cellulose (EHEC), hydroxyethyl cellulose (HEC), methyl cellulose, ethyl cellulose, carboxymethyl cellulose (carmellose), carboxymethyl hydroxyethyl cellulose (CMHEC), sodium or calcium carmellose, nitro cellulose, cellulose acetate, cellulose butyrate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropyl methylcellulose phthalate, cross-linked carboxymethyl cellulose (croscarmellose) and sodium or calcium croscarmellose; yet more preferably hydroxypropyl methylcellulose (hypromellose), most preferably hydroxypropyl methylcellulose selected from the group consisting of hypromellose type 2910 (Methocel™ E-types, Premium quality, Dow Chemical Company) and hypromellose type 2208

(Methocel™ K-types, Premium quality, Dow Chemical Company), preferably

Methocel™Kl5M or Methocel™K4M (both premium quality, Dow Chemical Company),, more preferably Methocel™Kl5M with a viscosity of 11,250-21,000 mPa.s (2% in water at 20° C); Dow Chemical Company).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one glidant, such as colloidal silicon dioxide, preferably fine particle anhydrous silicic acids (light anhydrous silicic acids without hydrophobizing treatment or amorphous silica fine particles with particle size of not more than 0.1 micron or fumed silica) or talc. Preferred glidants are fumed silica and light anhydrous silicic acids, such as Sylysia 320 (trade name, Fuji Silysia Chemical Ltd.), Aerosil® 200 (Evonik Industries) and the like and talc. Particularly preferred glidants are Aerosil®200 and/or talc.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one lubricant, e.g. stearic acid or a pharmaceutically acceptable salt thereof, a sucrose ester of a fatty acid a wax and/or a mixture thereof. In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one lubricant, wherein said lubricant is selected from stearic acid, magnesium stearate, calcium stearate, sucrose esters of fatty acids, sodium stearyl fumarate, waxes, DL-leucine, sodium lauryl sulfate and magnesium lauryl sulfate or mixtures thereof. A particularly preferred lubricant for use in the oral ER pharmaceutical composition of the invention is magnesium stearate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises at least one binder. Preferred binders are polyvinylpyrrolidones. Particularly preferred binders are povidones with K-values of about 29 to about 32 or about 90 (e.g.

Plasdone K-29/32, Plasdone K-90 e.g. from BASF) It is generally known in the art that a given excipient may fulfill different functions depending on the composition. Hence, a water-soluble filler, a water-insoluble filler or a hydrophilic polymer may also act as a binder and vice versa.

Multiparticulate compositions based on spheres and/or non-spherical kernels prepared by layering technique

In one embodiment, the oral ER composition according to the invention is a

multiparticulate composition, wherein the particles comprised by said multiparticulate composition are spheres and/or non-spherical kernels. In a preferred embodiment, said particles are spheres, preferably spheres prepared from microcrystalline cellulose or co processed materials, such as sucrose and com starch (sugar spheres NF). Methods for preparing spheres for use in multiparticulate oral ER formulations are commonly known in the art. In a further preferred embodiment, said particles are non-spherical kernels, preferably sugar crystals, such as Sucrose.

A multiparticulate oral ER composition according to the invention, wherein the particles comprised by said multiparticulate composition are spheres and/or non-spherical kernels, can be prepared as follows:

A PPAR agonist is suspended in an aqueous solution of a film forming polymer or film forming combination. Spheres or non-spherical Kernels of appropriate size are layered with the PPAR agonist/film former suspension, in a fluid bed equipment, and the layered material is thoroughly dried. A seal coat is applied in a fluid bed equipment, followed by an extended- release layer. A final top coat is then applied. The coated multiparticulates are cured at elevated temperatures (i.e. above room temperature), e.g. at 40 -75°C for 2 - 24 hours, to ensure that polymer particles coalesce to form a smooth membrane on extended release multiparticulates. An appropriate quantity of the final beads are filled into hard capsules of appropriate size, either with or without addition of additional glidants or lubricants.

In one embodiment, the oral ER composition according to the invention is a

multiparticulate composition comprising:

i. a PPAR agonist as defined herein;

ii. a film-forming composition;

iii. spheres and/or non-spherical kernels;

iv. a seal coat;

v. an extended-release layer; and vi. a top coat.

Film- forming compositions (ii) for use in said multiparticulate oral ER composition typically comprise film-forming polymers, pigments, and plasticizers. Suitable film-forming compositions are known in the art and commercially available, e.g. those sold under the designation Opadry® Complete Film Coating System by Colorcon.

Preferably, said spheres (iii) are prepared from sugar, microcrystalline cellulose or co processed materials, such as SUGLETS® Sugar Spheres by Colorcon. Suitable non-spherical kernels (iii) are for example sugar crystals.

A suitable seal coat (iv) is, for example, Opadry® II.

A suitable extended-release layer (v) is, for example, an ER layer comprising a film forming polymer, a plasticizer and a stabilizer. ER layers are commonly known in the art, e.g. aqueous ethylcellulose dispersions (such as Surelease®).

A suitable top coat (vi) is, for example, Opadry® II.

In a preferred embodiment, the oral ER composition according to the invention is a multiparticulate composition comprising:

i. 1% to 8% wt/wt of a PPAR agonist as defined herein;

ii. 0.6% to 24% wt/wt of a film-forming composition and

iii. 29% to 95% wt/wt of spheres or non-spherical kernels.

iv. 0.6% to 32% wt/wt of a seal coat;

v. 1% to 45% wt/wt of an extended-release layer; and

vi. 0.2% to 24% wt/wt of a top coat.

Multiparticulate compositions based on mini-tablets

In one embodiment, the oral ER composition according to the invention is a

multiparticulate composition, wherein the particles comprised by said multiparticulate composition are small tablets ("mini-tablets").

A multiparticulate oral ER composition according to the invention, wherein the particles comprised by said multiparticulate composition are small tablets ("mini-tablets"), can be prepared as follows:

A first option is to prepare the mini-tablets by an aqueous or organic wet granulation process. PPAR agonist, fillers and tablet binder(s) used for wet granulation are mixed. The mixture is wet granulated with purified water or organic solvent(s), dried and calibrated. The dry granules are mixed with glidant(s) and lubricant(s), and compressed to kernels of appropriate shape and mass.

As an alternative, the mini-tablets are prepared by a direct compression process. PPAR agonist and direct compression excipient(s) are intimately mixed, part of the mixture is shortly pre-mixed with glidant(s) and lubricant(s). The two mixtures are shortly mixed and compressed to tablet kernels as above.

To prevent any interaction of the extended-release coating system with the drug substance, the kernels may first be coated with a seal coat in a side- vented pan or a fluid bed coating equipment. The extended-release layer excipient(s) are mixed with purified water or organic solvent(s) and pore former(s). The coating solution/suspension is applied in an appropriate equipment, e.g. a side- vented pan or a fluid-bed coater. The final mini-tablets are cured for an appropriate time at an appropriate temperature to allow coalescence of the latex particles. An appropriate number of mini-tablets are filled into hard capsules to provide the final product.

In one embodiment, the oral ER composition according to the invention is a

multiparticulate composition comprising:

i. 0.8% to 4% wt/wt of a PPAR agonist as defined herein;

ii. 19.8% to 98.8% wt/wt of a filler;

iii. 0.4% to 9.9% wt/wt of a binder;

iv. 0% to 39.5% wt/wt of an external phase;

v. 0% to 0.8% wt/wt of a glidant; and

vi. 0% to 0.8% wt/wt of a lubricant;

wherein the particles comprised by said multiparticulate composition are small tablets ("mini tablets").

One or several different fillers (ii) may be used, e.g. lactose, cornstarch, dibasic calcium phosphate dihydrate (USP) (EMCOMPRESS®) or anhydrous dibasic calcium phosphate (USP) (EMCOMPRESS® Anhydrous) (both by JRS Pharma), microcrystalline cellulose or mixtures thereof.

A Suitable binder (iii) is e.g. polyvinylpyrrolidone K-30 (povidone).

As external phase (iv), lactose, cornstarch, dibasic calcium phosphate dihydrate (USP) (EMCOMPRESS®) or anhydrous dibasic calcium phosphate (USP) (EMCOMPRESS® Anhydrous) (both by JRS Pharma), microcrystalline cellulose or a mixture thereof may be used. Suitable glidants are for example talcum, colloidal silicon dioxide (Aerosil®), STA- Rx® 1500 (i.e. meachanically treated com starch with a higher content of cold-watersoluble parts than ordinary com starch; originally manufactured by A. E. Staley Mfg. Co., Decatur, Ill.) or mixtures thereof

Suitable lubricants are for example magnesium stearate, sodium stearyl fumarate, stearic acid, PRECIROL® ATO-5 (Glyceryl distearate NF; e.g. as manufactured by Gattefosse) and/or mixtures thereof

In one embodiment, the oral ER composition according to the invention is a

multiparticulate composition comprising:

i. 0.5% to 2.5% wt/wt of a PPAR agonist as defined herein;

ii. 24.8% to 99.3% wt/wt of a direct compression excipient;

iii. 0.1% to 3.7% wt/wt of a glidant; and

iv. 0.1 % to 3.7% wt/wt of a lubricant.

wherein the particles comprised by said multiparticulate composition are small tablets ("mini tablets").

A suitable direct compression excipient (ii) is for example modified, spray dried lactose ("Fast Flo® Lactose").

A suitable glidant is for example colloidal silicon dioxide (Aerosil).

Suitable lubricants (iv) are for example magnesium stearate, sodium stearyl fumarate, stearic acid, PRECIROL® and/or mixtures thereof.

In a preferred embodiment, the oral ER composition according to the invention is a multiparticulate composition; wherein the particles comprised by said multiparticulate composition are small tablets ("mini-tablets") and wherein said small tablets are film-coated tablets, the film-coating comprising:

i. 2.4% to 24.2% wt/wt of a seal coat;

ii. 6.1% to 97.0% wt/wt of an extended-release layer; and

iii. 0.6% to 24.2% wt/wt of a pore former.

A suitable seal coat (i) is for example Opadry® II Clear.

A suitable ER layer (ii) is for example a composition comprising film-forming polymers, plasticizers and stabilizers, e.g. an aqueous ethylcellulose dispersion (such as Sure lease®).

A suitable pore former (iii) is for example Opadry® II Clear. In one embodiment, the oral ER composition according to the invention is an osmotically controlled composition.

Osmotically controlled oral delivery system ("Push-Pull")

In one embodiment, the oral ER composition according to the invention is an osmotically controlled composition ("push-pull") comprising a push layer (osmotic core) comprising a PPAR agonist as defined herein, a drug layer and a semipermeable membrane.

Osmotic drug delivery systems are commonly known in the art and some are commercially available (e.g. OROS®, which is a trade name by ALZA Corp. and an abbreviation for“osmotic controlled-release oral delivery system”).

An osmotically controlled oral ER composition ("push-pull") according to the invention can be prepared as follows:

Drug layer

PPAR agonist, polyethylene oxide and sodium chloride are mixed and granulated by an aqueous solution of binder(s) in a high-shear mixer or fluid-bed equipment. The granulated, dried mass is passed through a screen. Part of the mass is thoroughly pre-mixed with the antioxydant(s), lubricant(s) and glidant(s), and thereafter the two parts were mixed.

Push layer

Sodium chloride and dye(s) are mixed and passed through a screen. This mix and polyethylene oxide are granulated with an aqueous solution of binder(s) in a fluid-bed equipment. After drying, the granules are calibrated through an appropriate screen. Part of the granules, antioxydant(s) and lubricant(s) / glidant(s) are pre-mixed for a short time, and thereafter blended with the residual mass.

Layer tablet

On a layer tableting press, the drug layer is compressed at low compression force to a first layer. After feeding the push layer, the tablets are compressed at standard compression force to cylindric, biconvex kernels with a double radius of curvature.

Semipermeable membrane

In a side- vented pan with appropriate explosion-proof precautions, a solution of cellulose acetate and plasticizer / pore former in acetone/water is sprayed on the kernels. The coated kernels are thoroughly dried in the equipment.

Laser drilling The coated kernels are fed into a laser-drilling equipment and oriented e.g. by optical methods to allow the drug layer to be positioned constantly in the same direction. By a laser pulse of appropriate strength, an exit passageway is drilled through the semipermeable membrane coat to connect the drug layer with the exterior of the system. A final drying process follows to remove any residual solvent from the coating process.

In one embodiment, the oral ER composition according to the invention is an osmotically controlled composition ("push-pull") comprising:

i. 0.7% to 2.9% wt/wt of a PPAR agonist as defined herein;

ii. 21.8% to 92.8% wt/wt of a polyethylene oxide NF;

iii. 2.9% to 23.2% wt/wt of sodium chloride;

iv. 0.6% to 11.6% wt/wt of a binder;

v. 0.01% to 0.6% wt/wt of an antioxidant;

vi. 0.1% to 6.4% wt/wt of a lubricant;

vii. 2.9% to 29% wt/wt of cellulose acetate; and

viii. 0% to 2.9% wt/wt of a plasticizer/pore former; "

ix. 0% to 5.8% wt/wt of a dye

A suitable polyethylene oxide (ii) is for example Polyox WSR N-80 (200 kDa) (Dow Chemical Company) and/or Polyox WSR-303 (7,000 kDa) (Dow Chemical Company).

A suitable binder (iv) is for example povidone K-30.

A suitable antioxidant (v) is for example butylated hydroxytoluene NF.

A suitable lubricant (vi) is for example magnesium stearate.

A suitable plasticizer/pore former (viii) is for example Macrogol 3350 (PEG 3350 Da).

A suitable dye (ix) is for example iron oxide

Inert matrix-type tablet formulations

In one embodiment, the oral ER composition according to the invention is an inert matrix-type tablet composition. In a preferred embodiment, the inert matrix of said inert matrix-type tablet composition is based on Eudragit RL.

An inert matrix-type tablet oral ER composition according to the invention can be prepared as follows:

PPAR agonist, an inert matrix type polymer, and filler(s) are thoroughly mixed and sieved. A release-controlling polymer and acetone are added to obtain a wet mass, which is dried in an explosion-proof environment at not more than 40°C to get a solid mass. This mass is crushed and sieved adequately lubricant(s) / glidant(s) are mixed for 3 minutes with the dry granulate, and the resulting mass is compressed to oblongue, biconvex tablets of appropriate weight.

In one embodiment, the oral ER composition according to the invention is an inert matrix-type tablet composition comprising:

i. 0.3% to 2.8% wt/wt of a PPAR agonist as defined herein;

ii. 9.5% to 94.9% wt/wt of an inert matrix type polymer;

iii. 1.9% to 38% wt/wt of a filler;

iv. 2.8% to 38% wt/wt of a release-controlling polymer; and

v. 0.1% to 9.5% wt/wt of a lubricant and/or a glidant.

A suitable inert matrix type polymer (ii) is for example Eudragit RL.

A suitable filler (iii) is for example Dibasic Calcium Phosphate Dihydrate (USP) (EMCOMPRESS®).

A suitable release-controlling polymer (iv) is for example Eudragit S.

A suitable lubricant is for example magnesium stearate.

A suitable glidant (v) is for example talc.

Single-Unit Matrix Tablet Based On Hydrophilic Polymers

In one embodiment, the oral ER composition according to the invention is a matrix tablet composition e.g. an ER matrix tablet composition, wherein the matrix of said matrix tablet composition comprises a hydrophilic polymer.

An oral ER matrix tablet composition according to the invention, wherein the matrix comprises a hydrophilic polymer, can be prepared as follows:

Hydrophilic polymer(s) and soluble or insoluble Filler(s) are intimately blended within a high shear mixer. Purified water is added, and the mass is kneaded until a sharp rise in power consumption occurs. Thereafter, the mass is calibrated, dried in a fluid bed dryer and passed through a 20 mesh screen. The dried granules are intimately mixed in a V-cone blender with a PPAR agonist. A small part of the mixture is shortly blended with lubricant(s) and glidant(s). Thereafter, this pre-blend and the residual mass are shortly blended to provide the ready-to- compress mass. The mass is compressed to oblonge, biconvex tablets with breaking score, which may be film-coated by any appropriate taste-masking film coat.

In a preferred embodiment, said hydrophilic polymer is xanthan gum, locust bean gum or a mixture thereof.

In one embodiment, there is provided a matrix tablet oral ER composition comprising: i. 1.6% to 6.2% wt/wt of a PPAR agonist as defined herein;

ii. 4.6% to 78.1% wt/wt of a hydrophilic polymer;

iii. 12.5% to 93.7% wt/wt of a filler (water-soluble filler, water-insoluble filler or a mixture thereof);

iv. 0.1% to 3.1% wt/wt of a lubricant and or a glidant; and optionally

v. 4.5 % to 7.5 % wt/wt of a taste-masking coat (calculated on basis of 100% tablet weight)

A suitable hydrophilic polymer (ii) is for example xanthan gum, locust bean gum or a mixture thereof.

A suitable filler (iii) is for example lactose, a calcium phosphate (such as calcium hydrogen phosphate dihydrate or anhydrous calcium hydrogen phosphate,

EMCOMPRESS®), or a mixture thereof.

A suitable lubricant/glidant (iv) is for example magnesium stearate.

A suitable glidant (iv) is for example talc.

A suitable taste-masking coat (v) is for example Opadry® taste mask film coating system (Colorcon).

In one embodiment, the oral ER composition according to the invention is a matrix tablet composition e.g. an ER matrix tablet composition, wherein the matrix of said matrix tablet composition comprises a cellulosic polymer.

An oral ER composition according to the invention comprising an ER matrix comprising a cellulosic polymer can be prepared as follows:

PPAR agonist and soluble or insoluble filler(s) (lst part) are pre-mixed and sieved. The pre-mix is thoroughly mixed with sieved soluble or insoluble filler(s) (2nd part), and hydrophilic, cellulosic polymer(s). A minor part of this mixture is shortly mixed with sieved lubricant(s) and glidant(s), to form a 2nd premix. A final mixture is compressed to biconvex tablets with an appropriate shape and diameter.

In one embodiment, said cellulosic polymer is selected from the group consisting of methylcellulose (Methocel A4M Premium, Colorcon); Carmellose (Blanose 7H4FD,

Aqualon) and mixtures thereof.

In one embodiment, there is provided a matrix tablet oral ER composition comprising: i. 1.4% to 5.8% wt/wt of a PPAR agonist as defined herein;

ii. 1.4% to 86.9% wt/wt of a cellulosic polymer;

iii. 5.8% to 44.9% wt/wt of a water-soluble filler iv. 5.8% to 44.9% wt/wt of a water-insoluble filler;

v. 0.03% to 8.7% wt/wt of a lubricant and or a glidant; and optionally

vi. 3.0 % to 7.5 % wt/wt of a taste-masking coat (calculated on basis of 100% tablet weight).

A suitable hydrophilic polymer (ii) is for example Methylcellulose (Methocel A4M Premium, Colorcon); Carmellose (Blanose 7H4FD, Aqualon) or a mixture thereof.

A suitable filler (iii) is for example lactose, a calcium phosphate (such as calcium hydrogen phosphate dihydrate or anhydrous calcium hydrogen phosphate,

EMCOMPRESS®), or a mixture thereof.

A suitable lubricant (iv) is for example magnesium stearate.

A suitable glidant (iv) is for example talc.

A suitable taste-masking coat (v) is for example Opadry® taste mask film coating system (Colorcon).

In one embodiment, the oral ER composition according to the invention is an ER matrix tablet composition, wherein the matrix of said ER matrix tablet composition comprises a hydrophilic polymer, preferably hypromellose as hydrophilic polymer.

Thus in one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. a PPAR agonist;

ii. a hydrophilic polymer;

iii. a water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D- fructose, dextran, polyoxyethylene hydrogenated castor oil, polyoxyethylenepolyoxypropyleneglycol, sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and

iv. a water-insoluble filler selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, sodium carboxymethyl starch, microcrystalline cellulose, crystalline cellulose, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate.

wherein said oral ER pharmaceutical composition does not comprise metformin. In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. not more than one API wherein the API is a PPAR agonist;

ii. a hydrophilic polymer;

iii. a water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D- fructose, dextran, glucose, polyoxyethylene hydrogenated castor oil, polyoxyethylenepolyoxypropyleneglycol, sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and

iv. a water-insoluble filler selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, sodium carboxymethyl starch, microcrystalline cellulose, crystalline cellulose, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate.

In one embodiment the oral ER composition according to the invention is an ER matrix tablet composition, wherein the matrix of said ER matrix tablet composition comprises a hydrophilic polymer, preferably hypromellose as hydrophilic polymer, and a PPAR agonist.

Thus in one preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. an ER matrix comprising a PPAR agonist and a hydrophilic polymer;

ii. a water-soluble filler selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D- fructose, dextran, polyoxyethylene hydrogenated castor oil, polyoxyethylenepolyoxypropyleneglycol, sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and

iii. a water-insoluble filler selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, sodium carboxymethyl starch, microcrystalline cellulose, crystalline cellulose, monocalcium phosphate, dicalcium

phosphate and tricalcium phosphate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. a PPAR agonist or an ER matrix comprising a PPAR agonist;

ii. a hydrophilic polymer;

iii. a water-soluble filler;

iv. a water-insoluble filler;

v. a binder;

vi. a glidant;

vii. a lubricant; and optionally

viii. film-coatings, preferably a non- functional film-coating.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. a PPAR agonist or an ER matrix comprising a PPAR agonist;

ii. a hydrophilic polymer;

iii. a water-soluble filler;

iv. a water-insoluble filler;

v. a binder;

vi. a glidant;

vii. a lubricant; and optionally

viii. film-coatings, preferably a non- functional film-coating

wherein said PPAR agonist is a PPAR gamma agonist, preferably a PPAR gamma agonist selected from the group consisting of pioglitazone, rosiglitazone, troglitazone and pharmaceutically acceptable salts thereof; and

wherein said hydrophilic polymer is a cellulosic polymer selected from the group consisting of hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (hypromellose, HPMC), hydroxyethyl methyl cellulose (HEMC), ethyl hydroxyethyl cellulose (EHEC), hydroxyethyl cellulose (HEC), methyl cellulose, ethyl cellulose, carboxymethyl cellulose (carmellose), carboxymethyl hydroxyethyl cellulose (CMHEC), sodium or calcium

carmellose, nitro cellulose, cellulose acetate, cellulose butyrate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropyl methylcellulose phthalate, cross-linked carboxymethyl cellulose (croscarmellose) and sodium or calcium croscarmellose; and

wherein said water-soluble filler is selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D-fructose, dextran, polyethylene glycol (e.g., macrogol 400, macrogol 1500, macrogol 4000, macrogol 6000, macrogol 20000); polyoxyethylene hydrogenated castor oil (e.g., Cremophor RH40, HCO-40, HCO-60), polyoxyethylenepolyoxypropyleneglycol (e.g., pluronic F68), sorbitan polyoxyethylene higher fatty acid ester (e.g., Tween80); sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and wherein said water-insoluble filler is selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, crystalline cellulose, sodium carboxymethyl starch, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate; and

wherein said binder is selected from polyvinylpyrrolidones; and

wherein said glidant is selected from light anhydrous silicic acids; and

wherein said lubricant is selected from the group consisting of stearic acid, magnesium stearate, calcium stearate, sucrose esters of fatty acids, sodium stearyl fumarate, waxes, DL- leucine, sodium lauryl sulfate, magnesium lauryl sulfate and mixtures thereof; and

wherein said film coatings are non- functional film-coatings, such as Opadry® II.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.3% to 5.6% wt/wt of a PPAR agonist;

ii. 1.4% to 55.5% wt/wt of a hydrophilic polymer, preferably 20% to 30%

wt/wt of a hydrophilic polymer;

iii. 8.1% to 98.3% wt/wt of a water-soluble filler and a water-insoluble filler, preferably 4.05% to 49.15% wt/wt of a water-soluble filler and 4.05% to 49.15% wt/wt of a water-insoluble filler;

iv. 0.03% to 8.3% wt/wt of a binder;

v. 0.01% to 4.2% wt/wt of a glidant;

vi. 0.01% to 4.2% wt/wt of a lubricant;

vii. 3.0 % to 7.5 % wt/wt of a non- functional film-coating (calculated on basis of 100% tablet weight). In a preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.3% to 5.6% wt/wt of a PPAR agonist;

ii. 1.4% to 55.5% wt/wt of a hydrophilic polymer, preferably 20% to 30%

wt/wt of a hydrophilic polymer;

iii. 8.1% to 98.3% wt/wt of a water-soluble filler and a water-insoluble filler, preferably 4.05% to 49.15% wt/wt of a water-soluble filler and 4.05% to 49.15% wt/wt of a water-insoluble filler;

iv. 0.03% to 8.3% wt/wt of a binder;

v. 0.01% to 4.2% wt/wt of a glidant;

vi. 0.01% to 4.2% wt/wt of a lubricant;

vii. 3.0 % to 7.5 % wt/wt of a non- functional film-coating (calculated on basis of 100% tablet weight); wherein

said PPAR agonist is a PPAR gamma agonist, preferably a PPAR gamma agonist selected from the group consisting of pioglitazone, rosiglitazone, troglitazone and pharmaceutically acceptable salts thereof; and

wherein said hydrophilic polymer is a cellulosic polymer selected from the group consisting of hydroxypropyl cellulose (HPC), hydroxypropyl methylcellulose (hypromellose, HPMC), hydroxyethyl methyl cellulose (HEMC), ethyl hydroxyethyl cellulose (EHEC), hydroxyethyl cellulose (HEC), methyl cellulose, ethyl cellulose, carboxymethyl cellulose (carmellose), carboxymethyl hydroxyethyl cellulose (CMHEC), sodium or calcium carmellose, nitro cellulose, cellulose acetate, cellulose butyrate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate trimellitate, hydroxypropyl methylcellulose phthalate, cross-linked carboxymethyl cellulose (croscarmellose) and sodium or calcium croscarmellose; and

wherein said water-soluble filler is selected from the group consisting of lactose, glucose, mannitol, trehalose, D-sorbitol, xylitol, sucrose, maltose, lactulose, D-fructose, dextran, polyethylene glycol (e.g., macrogol 400, macrogol 1500, macrogol 4000, macrogol 6000, macrogol 20000); polyoxyethylene hydrogenated castor oil (e.g., Cremophor REMO, HCO-40, HCO-60), polyoxyethylenepolyoxypropyleneglycol (e.g., pluronic F68), sorbitan polyoxyethylene higher fatty acid ester (e.g., Tween80); sodium chloride, magnesium chloride, citric acid, tartaric acid, glycine, b-alanine, lysine hydrochloride and meglumine; and wherein said water- insoluble filler is selected from the group consisting of com starch, potato starch, wheat starch, rice starch, partially pregelatinized starch, hydroxypropylstarch, crystalline cellulose, sodium carboxymethyl starch, monocalcium phosphate, dicalcium phosphate and tricalcium phosphate; and

wherein said binder is a polyvinylpyrrolidone; and

wherein said one or more glidants are colloidal silicon dioxides, preferably colloidal silicon dioxides selected from light anhydrous silicic acids and fumed silica; and

wherein said one or more lubricants are selected from the group consisting of stearic acid, magnesium stearate, calcium stearate, sucrose esters of fatty acids, sodium stearyl fumarate, waxes, DL-leucine, sodium lauryl sulfate, magnesium lauryl sulfate and mixtures thereof; and

wherein said film coatings are non- functional film-coatings, such as Opadry® II.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone hydrochloride;

ii. hypromellose;

iii. lactose monohydrate; and

iv. dicalcium phosphate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate; and at least one excipient selected from the group

consisting of: povidone, microcrystalline cellulose, fumed silica, talc and magnesium stearate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone hydrochloride; ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate; and

v. povidone.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone

hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate;

v. povidone; and

vi. microcrystalline cellulose.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone

hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate;

v. povidone;

vi. microcrystalline cellulose; and

vii. fumed silica ( e.g. Aerosil®).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone

hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate;

v. povidone;

vi. microcrystalline cellulose;

vii. fumed silica (Aerosil®); and viii. talc.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone

hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate;

v. povidone;

vi. microcrystalline cellulose;

vii. fumed silica ( e.g. Aerosil®);

viii. talc; and

ix. magnesium stearate.

In a particularly preferred embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. pioglitazone hydrochloride or an ER matrix comprising pioglitazone

hydrochloride;

ii. hypromellose;

iii. lactose monohydrate;

iv. dicalcium phosphate;

v. povidone;

vi. microcrystalline cellulose;

vii. fumed silica ( e.g. Aerosil®);

viii. talc;

ix. magnesium stearate; and optionally

x. a film-coating, preferably a non- functional film-coating such as e.g.

Opadry® II.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.4% wt/wt pioglitazone hydrochloride;

ii. 6.5% to 43.5% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose;

iii. 10.9% to 77% wt/wt lactose monohydrate; and iv. 16.3% to 32.6% wt/wt dicalcium phosphate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.3% wt/wt pioglitazone hydrochloride;

ii. 6.4% to 42.8% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose;

iii. 10.7% to 75.7% wt/wt lactose monohydrate;

iv. 16.0% to 32.1 % wt/wt dicalcium phosphate; and

v. 1.6% to 3.2% wt/wt povidone.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.2% wt/wt pioglitazone hydrochloride;

ii. 6.3% to 41.9% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose;

iii. 10.5% to 74.1 % wt/wt lactose monohydrate;

iv. 15.7% to 31.4% wt/wt dicalcium phosphate;

v. 1.6% to 12.6% wt/wt povidone; and

vi. 2.1 % to 41 9%wt/wt microcrystalline cellulose.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.2% wt/wt pioglitazone hydrochloride;

ii. 6.3% to 41.8% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose;

iii. 10.5% to 74.0% wt/wt lactose monohydrate;

iv. 15.7% to 31.4% wt/wt dicalcium phosphate;

v. 1.6% to 12.5% wt/wt povidone;

vi. 2.1% to 41 8%wt/wt microcrystalline cellulose; and

vii. 0.17% to 9.4% wt/wt fumed silica ( e.g. Aerosil®).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.2% wt/wt pioglitazone hydrochloride;

ii. 6.3% to 41.8% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose; iii. 10.4% to 74.0% wt/wt lactose monohydrate;

iv. 15.7% to 31.3% % wt/wt dicalcium phosphate;

v. 1.6% to 12.5% wt/wt povidone;

vi. 2.1% to 41 8%wt/wt microcrystalline cellulose;

vii. 0.17% to 9.4%wt/wt fumed silica ( e.g. Aerosil®); and

viii. 0.04% to 4.2% wt/wt talc.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.2% wt/wt pioglitazone hydrochloride;

ii. 6.3% to 41.7% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose;

iii. 10.4% to 73.8% wt/wt lactose monohydrate;

iv. 15.6% to 31.3% wt/wt dicalcium phosphate;

v. 1.6% to 12.5% % wt/wt povidone;

vi. 2.1 % to 41.7% wt/wt microcrystalline cellulose;

vii. 0.17% to 9.4% wt/wt fumed silica ( e.g. Aerosil®);

viii. 0.04% to 4.2 wt/wt talc; and

ix. 0.19% to 10.4% wt/wt magnesium stearate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 0.2% to 5.2% wt/wt pioglitazone hydrochloride;

ii. 6.3% to 41.7% wt/wt hypromellose, preferably 20% to 30% wt/wt hypromellose;

iii. 10.4% to 73.8% wt/wt lactose monohydrate;

iv. 15.6% to 31.3% wt/wt dicalcium phosphate;

v. 1.6% to 12.5% wt/wt povidone;

vi. 2.1 % to 41.7% wt/wt microcrystalline cellulose;

vii. 0.17% to 9.4% wt/wt fumed silica ( e.g. Aerosil®);

viii. 0.04% to 4.2% wt/wt talc;

ix. 0.19% to 10.4% wt/wt magnesium stearate; and optionally

x. 2.5 % to 8.0 % wt/wt of Opadry® II (calculated on Opadry dry matter, and on basis of 100% tablet weight). In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride;

ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate; and

iv. 75.0 mg dicalcium phosphate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride;

ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate;

iv. 75.0 mg dicalcium phosphate; and

v. 14.47 mg povidone.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride;

ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate;

iv. 75.0 mg dicalcium phosphate;

v. 14.47 mg povidone; and

vi. 75.0 mg microcrystalline cellulose.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride;

ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate;

iv. 75.0 mg dicalcium phosphate;

v. 14.47 mg povidone;

vi. 75.0 mg microcrystalline cellulose; and

vii. 2.25 mg fumed silica ( e.g. Aerosil®).

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride; ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate;

iv. 75.0 mg dicalcium phosphate;

v. 14.47 mg povidone;

vi. 75.0 mg microcrystalline cellulose;

vii. 2.25 mg fumed silica ( e.g. Aerosil®); and

viii. 1.40 mg talc.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride;

ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate;

iv. 75.0 mg dicalcium phosphate;

v. 14.47 mg povidone;

vi. 75.0 mg microcrystalline cellulose;

vii. 2.25 mg fumed silica ( e.g. Aerosil®);

viii. 1.40 mg talc; and

ix. 2.10 mg magnesium stearate.

In one embodiment, the oral ER pharmaceutical composition according to the invention comprises:

i. 5.51 mg pioglitazone hydrochloride;

ii. 78.75 mg hypromellose;

iii. 95.25 mg lactose monohydrate;

iv. 75.0 mg dicalcium phosphate;

v. 14.47 mg povidone;

vi. 75.0 mg microcrystalline cellulose;

vii. 2.25 mg fumed silica ( e.g. Aerosil®);

viii. 1.40 mg talc;

ix. 2.10 mg magnesium stearate; and optionally

x. 10 mg Opadry® II.

In one embodiment, the oral ER pharmaceutical composition of the invention is a composition that provides for prolonged gastric retention ("prolonged gastric retention system"). Accordingly, in one embodiment, the oral ER pharmaceutical composition of the invention unfolds rapidly within the stomach to a size that resists gastric emptying; or it comprises a hydrophilic erodible polymer system that swells over a short period of time to a size that encourages prolonged gastric retention. Prolonged gastric retention systems are commonly known in the art and commercially available, e.g. Minextab Floating (Galenix Innovations, France), Acuform® (Depomed, Inc.) (c.f. Vivek K. Pawar, Shaswat Kansal, Garima Garg, Rajendra Awasthi, Deepak Singodia, and Giriraj T. Kulkami; Drug Delivery, 2011; 18(2): 97-110; Gastroretentive dosage forms: A review with special emphasis on floating drug delivery systems).

In other embodiments, the oral ER pharmaceutical compositions according to the invention also contain other therapeutic substances. Optionally, otoprotective agents, such as antioxidants, alpha lipoic acid, calcium, fosfomycin or iron chelators, to counteract potential ototoxic effects that may arise from the use of specific therapeutic agents or excipients, diluents or carriers are included in the pharmaceutical compositions.

In some embodiments, the oral ER pharmaceutical compositions according to the invention include a dye to help enhance the visualization of the pharmaceutical composition when applied. In other embodiments, the pharmaceutical compositions also include one or more pH adjusting agents or buffering agents. Suitable pH adjusting agents or buffers include, but are not limited to acetate, bicarbonate, ammonium chloride, citrate, phosphate, pharmaceutically acceptable salts thereof or combinations or mixtures thereof. Such pH adjusting agents and buffers are included in an amount required to maintain pH of the composition between a pH of about 5 and about 9, in a preferred embodiment a pH between about 6.5 to about 7.5.

In one embodiment, the oral ER pharmaceutical composition comprises an ion exchange resin, such as sodium polystyrene sulfonate USP (e.g. AMBERLITE™ IRP69 pharmaceutical grade cation exchange resin, Rohm and Haas).

Using the oral ER pharmaceutical composition of the invention

The oral ER pharmaceutical composition of the invention is administered for preventive and/or therapeutic treatments. Preventive treatments comprise prophylactic treatments. In preventive applications, the oral ER pharmaceutical composition of the invention is administered to a subject suspected of having, or at risk for developing a disease, disorder or condition as described herein. In therapeutic applications, the oral ER pharmaceutical composition of the invention is administered to a subject such as a patient already suffering from a disorder disclosed herein, in an amount sufficient to cure or at least partially arrest the symptoms of the disease, disorder or condition as described herein. Amounts effective for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the subject's health status and response to the drugs, and the judgment of the treating physician. In a preferred embodiment, the oral ER pharmaceutical composition as described herein is administered prior to surgical procedures, in particular prior to cochlea surgery.

In the case wherein the subject’s condition does not improve, the administration of the oral ER pharmaceutical composition of the invention may be administered chronically, which is, for an extended period of time, including throughout the duration of the subject's life in order to ameliorate or otherwise control or limit the symptoms of the subject’s disease or condition.

In the case wherein the subject’s status does improve, the administration of the oral ER pharmaceutical composition of the invention may be given continuously; alternatively, the dose of drug being administered may be temporarily reduced or temporarily suspended for a certain length of time (i.e., a“drug holiday”).

Once improvement of the patient’s otic conditions has occurred, a maintenance dose of the oral ER pharmaceutical composition of the invention is administered if necessary.

Subsequently, the dosage or the frequency of administration, or both, is optionally reduced, as a function of the symptoms, to a level at which the improved disease, disorder or condition is retained.

For some routes of administration, e.g. for injection into the inner ear and/or into the middle ear such as intratympanic injection a sustained release sytem can be used. In some routes of administration the penetration of the active ingredient is facilitated by transport enhancers as e.g. hyaluronic acid, DMSO. In some routes of administration, in particular when the PPAR agonist is administered by injection into the inner ear and/or into the middle ear a tixotropic or thermogeling formulation is used to enable a painless administration and forming a gel or a high viscous composition ensuring prolonged and continuous release of the active ingredient into the inner ear and/or into the middle ear.

If administered intratympanically the PPAR agonist will be administered on or near the round window membrane via intratympanic injection. The PPAR agonist can be applied via syringe and needle, wherein the needle is inserted through the tympanic membrane and guided to the area of the round window or crista fenestrae cochleae. The PPAR agonist is then deposited on or near the round window or crista fenestrae cochleae for localized treatment. Intratympanic injection of therapeutic agents is the technique of injecting an agent behind the tympanic membrane into the middle and/or inner ear, preferably into the middle ear.

In one embodiment, the PPAR agonist described herein is administered directly onto the round window membrane via transtympanic injection. In another embodiment, the PPAR agonist described herein is administered onto the round window membrane via a non- transtympanic approach to the inner ear. In additional embodiments, the composition described herein is administered onto the round window membrane via a surgical approach to the round window membrane comprising modification of the crista fenestrae cochleae. In one embodiment the delivery system is a syringe and needle apparatus that is capable of piercing the tympanic membrane and directly accessing the round window membrane or crista fenestrae cochleae of the auris interna. In some embodiments, the delivery device is an apparatus designed for administration of therapeutic agents to the middle and/or inner ear. By way of example only: GYRUS Medical Gmbh offers micro-otoscopes for visualization of and drug delivery to the round window niche; Arenberg has described a medical treatment device to deliver fluids to inner ear structures in U.S. Pat. Nos. 5,421,818; 5,474,529; and 5,476,446. U.S. patent application Ser. No. 08/874,208 describes a surgical method for implanting a fluid transfer conduit to deliver therapeutic agents to the inner ear. U.S. Patent Application

Publication 2007/0167918 further describes a combined otic aspirator and medication dispenser for intratympanic fluid sampling and medicament application.

Administration of the oral ER pharmaceutical composition of the invention can be provided chronically, which is, for an extended period of time, including throughout the duration of the subject's life. In some embodiments after long term treatment, hearing capacity is increased based on a reactivation of hair cells from a resting state. In some embodiments after long term treatment, hearing capacity is increased based on an increase of the number of hair cells or hair cell function subsequent to PPAR activation.

The amount of the oral ER pharmaceutical composition of the invention to be administered will vary depending upon factors such as disease condition and its severity, according to the particular circumstances surrounding the case, including, e.g., the specific PPAR agonist being administered, the condition being treated, the target area being treated, and the subject or host being treated.

In some embodiments, the amount of oral ER pharmaceutical composition of the invention administered to the subject comprises a dose of PPAR agonist that is below the dose needed for the treatment of diabetes using a PPAR agonist. In some embodiments, the amount of oral ER pharmaceutical composition of the invention administered to the subject comprises a dose of PPAR agonist that is a factor of 8-20 fold lower than the top dose evaluated and approved for the treatment of diabetes, in particular a factor of 8-20 fold lower than the top dose evaluated and approved for the treatment of diabetes in human. The top dose evaluated and tested for the treatment of diabetes in human e.g for a PPAR gamma agonist such as pioglitazone is usually in the range from about 30-45 mg/day. In some embodiments at the PPAR dose used the side effects seen in treatment of diabetes are not present.

In some embodiments, the amount of the oral ER pharmaceutical composition of the invention administered to the subject comprises a dose of PPAR agonist that is below the active dose for antidiabetic or anti-dyslipidemic effect of the PPAR agonist, in particular a dose that is below the active dose for antidiabetic or anti-dyslipidemic effect of the the PPAR agonist in human.

In some embodiments, the oral ER pharmaceutical composition of the invention is administered to a human at a dose of a PPAR agonist, usually PPAR gamma agonists, PPAR alpha agonists and/or PPAR alpha/gamma dual agonists, preferably a PPAR gamma agonist, more preferably pioglitazone, most preferably pioglitazone hydrochloride of about 0.05-60 mg/day, preferably of about 0.1-20 mg/day, more preferably of about 0.1-10 mg/day, more preferably of about 0.5-7.5 mg/day, more preferably of about 0.5-5 mg/day, more preferably of about 4-6 mg/day, more preferably of about 5-5.9 mg/day, most preferably of about 5.5 mg/day.

Methods of identification of patients who are suspected of having, or being at risk for developing hearing loss, hair cell degeneration or hair cell death are also comprised by the present invention. In some embodiments, patients who are suspected of having, or being at risk for developing hearing loss, hair cell degeneration or hair cell death are identified by measurement of serum and/or plasma adiponectin levels, in particular the measurement of high molecular weight adiponectin levels. In some embodiments the monitoring of the treatment success and/or the identification of the subject e.g. the identification of the subject who is suspected of having, or being at risk for developing hearing loss, hair cell degeneration or hair cell death, is achieved by measurement of serum and/or plasma adiponectin levels.

Kits/ Articles of Manufacture The disclosure also provides kits for preventing or treating hearing loss and/or preventing or inhibiting hair cell degeneration or hair cell death in a subject, preferably in human. Such kits generally will comprise the oral ER pharmaceutical composition disclosed herein, and instructions for using the kit. The disclosure also contemplates the use of the oral ER pharmaceutical composition disclosed herein, in the manufacture of medicaments for treating, abating, reducing, or ameliorating the symptoms of a disease, dysfunction, or disorder in a mammal, such as a human that has, is suspected of having, or being at risk for developing hearing loss, hair cell degeneration or hair cell death.

In some embodiments, kits include a carrier, package, or container that is

compartmentalized to receive one or more containers such as vials, tubes, and the like, each of the container(s) including one of the separate elements to be used in a method described herein. Suitable containers include, for example, bottles, vials, and test tubes. In other embodiments, the containers are formed from a variety of materials such as glass or plastic.

The articles of manufacture provided herein generally will comprise the oral ER pharmaceutical composition disclosed herein and packaging materials. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, bags, vials, containers, and any packaging material suitable for a selected composition and treatment.

In a further aspect, the present invention relates to a package comprising: at least one single unit dosage form comprising a PPAR agonist and/or at least one multiple unit dosage form comprising a PPAR agonist, wherein said single unit dosage form and said multiple unit dosage form each independently is an oral ER pharmaceutical composition as described herein; and wherein said at least one single unit dosage form and/or at least one multiple unit dosage form is/are packed in a package.

Examples

Example 1: Protection against antibiotic-induced hair cell damage

Organs of Corti were obtained from post-natal day 5 Sprague-Dawley rats and placed in organ culture. Gentamicin treatment resulted in 50 - 70% loss of hair cells after 48h in culture. Pioglitazone co-treatment was protective, almost completely preventing gentamicin- dependent hair cell loss, and largely preserving organ morphology.

Methods

Animal procedures

All animal procedures were carried out according to protocols approved by the

Kantonales Veterinaramt, Basel, Switzerland. Postnatal day 5 (p5) Sprague-Dawley rats were used for the studies. Studies were performed in vitro, using organ of Corti (OC) explants from p5 animals. Animals were sacrificed and the cochleae carefully dissected to separate the organ of Corti from the spiral ganglion, stria vascularis and Reissner's membrane [Sobkowicz HM, Loftus JM, Slapnick SM. Acta Otolaryngol Suppl. 1993;502:3-36]

Tissue culture

OCs were harvested then placed in culture medium [Dulbecco’s Modified Eagle Medium supplemented with 10% FCS, 25 mM HEPES and 30 U/ml penicillin (Invitrogen, Carlsbad, CA, USA)] and incubated for 24 hours at 37°C in an atmosphere of 95% 02/5% C02. After that period, the culture medium was replaced with fresh medium containing no compound or 200mM gentamicin alone or 200 mM gentamicin with either 2 or 10 mM pioglitazone, and incubated for a further 48 hours at 37°C. Ten OC explants were used for each treatment condition.

Hair cell counting

After incubation with compounds, the OCs were fixed in 4% paraformaldehyde, washed and then stained with a fluorescein (FITC)-conjugated phalloidin to detect inner and outer hair cells. After staining, the OCs were visualized and photographed using a fluorescence microscope (Olympus FSX100). Outer and inner hair cells were separately quantitated for the apical, basal, and middle turn of each organ of Corti. The values for each turn were averaged for the 10 OCs used for each condition. Significant differences between treatment groups in numbers OHC and IHC were determined using analysis of variance (ANOVA) followed by the least significant difference (LSD) post-hoc test (Stat View 5.0). Differences associated with P-values of less than 0.05 were considered to be statistically significant. All data are presented as mean ± SD. Results

Untreated organs of Corti were well preserved after 48 hours in culture presenting with intact ordered rows of outer hair cells (OHC) and inner hair cells (IHC). Pioglitazone treatment alone, at either 2 or 10 mM had no effect on hair cell number or morphology, indicating no direct adverse effect of pioglitazone (Fig 1 A-C). In contrast, 200mM gentamicin treatment resulted in almost complete destruction and loss of hair cells (Fig 1 A-C).

Pioglitazone at both 2 and 10 mM was able to antagonize the effects of gentamicin and to preserve hair cell number and morphology (Fig 1 A-C). Quantative image analysis was performed to count IHC and OHC separately in the apical, basal, and middle turns of each organ of Corti. While gentamicin treatment resulted in a consistent reduction of hair cell number of approximately 50 - 70% in each segment, pioglitazone at both concentrations was able to completely prevent gentamicin-dependent hair cell loss in all turns.

Example 2: Protection against noise-induced hearing loss

A formulation of pioglitazone or vehicle alone was applied into the middle ears of guinea pigs. The animals were then exposed to a noise trauma (broadband noise 4 - 20 kHz ,

115 dB (SPL) and recording of hearing sensitivity over the standard frequency range was performed 7-14 days later. Results obtained in the hearing test were compared to baseline values before injury. Pioglitazone protected hearing, resulting in a reduction of >50% in the threshold shifts in pioglitazone-treated animals vs. vehicle controls.

Methods

Animal Procedures

The guinea pig model is the preferred animal species in hearing research. The agent application as well as the noise trauma was applied under general anesthesia. Upon arrival, animals underwent an acclimatization period of at least one week prior to experiments.

Animals were housed in pairs with ad libitum access to food and water in a temperature and humidity controlled environment on a l2h/l2h light/dark cycle. The protocol was approved by the governmental animal use committee of Berlin, Germany.

Guinea pigs were first anaesthetized and hearing evaluated by a standard ABR method and then divided into treatment groups. Each animal received a single round window application of test substance to both ears. The following day, animals were exposed to 115 dB broad band noise for 2 hrs under anaesthesia. At one and two weeks following noise exposure, the animals underwent a second hearing evaluation.

Animals were dosed with a single 40 mΐ application of pioglitazone formulation or matching vehicle, onto the cochlear round window in both ears the day prior to noise exposure. For this approach, a hole was drilled in the rostral part of the skull to directly access the Bulla which allows drug application to the round window under visual control.

Animals were noise-exposed in a soundproof chamber (0.8x0.8x0.8 m, minimal attenuation 60 dB) for 2 h to broadband white noise (5-20 kHz) at 115 dB sound pressure level (SPL) under anaesthesia (60 mg/kg ketamine and 6 mg/kg xylazine). Noise was delivered binaurally by loudspeakers (HTC 11.19; Visaton, Haan, Germany) placed above the animal’s head. The speakers were connected to an audio amplifier (Tangent AMP-50; Aulum, Denmark) and a DVD player.

Hearing assessment At baseline before the noise exposure and on day 7 and 14 after noise, frequency- specific (2; 4; 8; 12; 16; 20; 24; 28; 32; 36; 40 kHz) auditory brainstem responses (ABR) were recorded in all treated animals and in controls. Auditory stimuli were delivered binaurally at different SPLs with a sinusoid generator (Model SSU2; Werk Femmeldewesen, Berlin, Germany). Frequency output was controlled and adjusted with a digital counter (1941A Digital Counter; Fluke, Scarborough, Ontario, Canada). Sub-dermal needle electrodes were placed at the vertex (active), mastoid (reference), and in one foot (ground). ABR recordings were carried out with a Viking IV- measurement system (Viasys Healthcare, Conshohocken, Pennsylvania). The brainstem responses were amplified (IOO,OOOc), filtered (bandpass 0.15-3 kHz), and averaged (300x) by the Viking IV-system. The amplitudes of the ABR waves were measured at different sound intensities by changing the attenuation of signal amplification. The amplitude-growth function was calculated for each tested frequency, and a linear regression was fitted to the linear portion of the data. The hearing threshold could be calculated for each frequency by extrapolating the linear amplitude-grow function of the regression line to zero. From these data, threshold differences (mean threshold shifts) were calculated between the control and the noise-exposed animals using the average values.

Results are represented as mean relative hearing loss (±SD) in decibels (dB) of the experimental groups compared to controls.

Results

Vehicle treated animals showed a significant average hearing loss of 31.9 ± 2.2 dB

(mean ± SD) over the frequency range of the noise challenge (5 - 20 kHz) at one week.

Pioglitazone afforded significant protection of approximately 60 % from noise-induced hearing loss, with only modest threshold shifts of 12.7 ± 1.3 dB (mean ± SD) (Fig 2).

At two weeks, slight recovery in both groups was noted. Vehicle treated animals showed a significant average hearing loss of 27.3 ± 12.6 dB (mean ± SD) at two weeks. Pioglitazone treated animals showed only modest threshold shifts of 6.3 ± 3.9 dB (mean ± SD) at two weeks. (Fig 2). These data demonstrate efficacy of pioglitazone to protect hearing. Example 3: Protection against antibiotic-induced hair cell damage by dual PPARa/g agonists and a PPARa-selective agonist

The experiment was carried out similarly to the experiments in example 1. Gentamicin treatment resulted is 50% loss of hair cells after 24h exposure to mouse OC’s in culture. Treatment with the dual PPARa/g agonists muraglitazar and tesaglitazar and with the PPARa- selective agonist fenofibric acid protected from gentamicin-dependent hair cell loss.

Methods

Methods were similar to those in Example 1. The main differences were that mouse OC’s were used rather than rat OC’s. Moreover, treatment was performed for 24 hrs with 50mM gentamicin. The number of OC’s used for each experimental condition was 3-5. The concentrations of test substances were 2mM and 10mM for tesaglitazar and muraglitazar, and 25mM and 150mM for fenofibric acid.

Results

Untreated organs of Corti were well preserved after 24 hours in culture presenting with intact ordered rows of outer hair cells (OHC) and inner hair cells (IHC). None of the test substances alone and any concentration had an effect on hair cell number or morphology, indicating no direct adverse effects (Fig 3 A-C). In contrast, 50mM gentamicin treatment resulted in approximately 50% loss of hair cells (Fig 3 A-C). Tesaglitazar at both 2 and 10 mM was able to antagonize the effects of gentamicin and to preserve hair cell number and morphology (Fig 3 A). Muraglitazar was not effective at 2 mM but was partially protective at 10 mM (Fig 3 B). Fenofibric acid was not effective at 25 mM but was completely protective at 150 mM (Fig 3 C).

Example 4: Multiparticulates based on sugar spheres prepared by layering technique

Process

Pioglitazone Hydrochloride is suspended in an aqueous solution of polyvinylpyrrolidone K-30 (approx. 10% solids). Sugar spheres of appropriate size are layered with the

Pioglitazone - polyvinylpyrrolidone suspension, in a fluid bed equipment, and the layered spheres are thoroughly dried. A seal coat of Opadry® is applied in a fluid bed equipment, followed by an extended-release layer of ethylcellulose (in form of a Sure lease® dispersion). A final top coat of Opadry® is then applied. The coated multiparticulates are cured at 60°C for 12 hours, to ensure that polymer particles coalesce to form a smooth membrane on extended release multiparticulates. A quantity of approx. 200 mg of the final beads are filled into hard capsules of size“2”.

Formulation

Table 1: Pioglitazone HC1 Prolonged Release Multiparticulate Hard Capsules (5.0 mg Dose)

*) includes film-forming polymer, plasticizer and stabilizer

Example 5: Multiparticulates based on Mini-Tablets (5.0 and 2.0 mg Dose)

Process

A first option is to prepare the tablets by a wet granulation process. Pioglitazone Hydrochloride, lactose hydrous, cornstarch and polyvinylpyrrolidone K-30 are mixed. The mixture is wet granulated with purified water, dried and calibrated. The dry granules are mixed with talcum and magnesium stearate and compressed to double-radius, biconvex kernels of 10 mg, with a diameter of 2.0 mm.

As an alternative, the tablets are prepared by a direct compression process. Pioglitazone Hydrochloride and lactose are intimately mixed, part of the mixture is shortly pre-mixed with AEROSIL, stearic acid and magnesium stearate. The two mixtures are shortly mixed and compressed to tablet kernels as above.

To prevent any interaction of the extended-release coating system with the drug substance, the kernels may first be coated with an Opadry seal coat in a side- vented pan. The extended-release coating of Surelease is mixed with purified water and Opadry II Clear as pore former. The coating solution/suspension is applied in a side- vented pan. The final tablets are cured for 24 hours at 40°C to allow coalescence of the latex particles. Twelve coated mini tablets are filled into a hard capsule to provide the final product.

Formulation

Table 2: Multiparticulates based on Mini-Tablets (5.0 and 2.0 mg Dose)

Example 6: Osmotically controlled oral delivery system comprising Pioglitazone Hydrochloride ("Push-Pull")

Drug layer

Pioglitazone Hydrochloride, polyethylene oxide and sodium chloride are mixed and granulated by an aqueous solution of povidone in a fluid-bed equipment. The granulated, dried mass is passed through a screen. Part of the mass is shortly pre-mixed with butylated hydroxytoluene and magnesium stearate, and thereafter the two parts are mixed.

Push layer Sodium chloride and iron oxide are mixed and passed through a screen. This mix and polyethylene oxide are granulated with an aqueous solution of hypromellose in a fluid-bed equipment. After drying, the granules are calibrated through an appropriate screen. Part of the granules, butylated hydroxytoluene and magnesium stearate are pre-mixed for a short time, and thereafter blended with the residual mass.

Layer tablet

On a layer tableting press, the drug layer is compressed at low compression force to a first layer. After feeding the push layer, the tablets are compressed at standard compression force to cylindric, biconvex kernels with a double radius of curvature.

Semipermeable membrane

In a side- vented pan with appropriate explosion-proof precautions, a solution of cellulose acetate and macrogol in acetone/water is sprayed on the kernels. The coated kernels are thoroughly dried in the equipment.

Laser drilling

The coated kernels are fed into a laser-drilling equipment and oriented to allow the drug layer to be positioned constantly in the same direction. By a laser pulse of appropriate strength, an exit passageway is drilled through the semipermeable membrane coat to connect the drug layer with the exterior of the system. A final drying process follows to remove any residual solvent from the coating process.

Formulation

Table 3: Osmotically controlled oral delivery system comprising Pioglitazone Hydrochloride (5.0 mg Dose)

Example 7: Inert Matrix-Type Tablet Formulation (5.0 mg Dose)

Process

An inert matrix-type tablet formulation is prepared as follows. Pioglitazone

Hydrochloride, Eudragit RL, and Emcompress® are thoroughly mixed and sieved. Eudragit S and Acetone are added to obtain a wet mass, which is dried in an explosion-proof

environment at not more than 40°C to get a solid mass. This mass is crushed and sieved adequately. Talc and Magnesium Stearate are mixed for 3 minutes with the dry granulate, and the resulting mass is compressed to ob longue, biconvex tablets of 300 mg weight.

Formulation

Table 4: Inert Matrix-Type Tablet Formulation (5.0 mg Dose)

Example 8: Single-Unit Matrix Tablet Based On Hydrophilic Polymers (10.0 mg Dose)

Process

Xanthan gum, locust bean gum and lactose are intimately blended within a high shear mixer. Purified water is added, and the mass is kneaded until a sharp rise in power consumption occurs. Thereafter the mass is calibrated, dried in a fluid bed dryer and passed through a 20 mesh screen. The dried granules are intimately mixed in a V-cone blender with Pioglitazone Hydrochloride. A small part of the mixture is shortly blended with talc and magnesium stearate. Thereafter this pre-blend and the residual mass are shortly blended to provide the ready-to-compress mass. The mass is compressed to oblonge, biconvex tablets with breaking score, which may be film-coated by any appropriate taste-masking film coat. Formulation

The following example uses a technology in analogy to US 5,135,757 (TimeRx®, Penwest).

Table 5: Single-Unit Matrix Tablet Based On Hydrophilic Polymers (10.0 mg Dose)

Example 9: Single-Unit Matrix Tablet Based On Cellulosic Polymers (7.5 mg Dose)

Process

Pioglitazone hydrochloride and lactose, anhydrous (I st part) are pre-mixed and sieved. The pre-mix is thoroughly mixed with sieved lactose, anhydrous (2 nd part), methylcellulose and carmellose. A minor part of this mixture is shortly mixed with sieved talc and magnesium stearate, to form a 2 nd premix. A final mixture is compressed to biconvex tablets with a diameter of 8 mm.

Formulation

Table 6: Tablet based on Methylcellulose and Carmellose (7.5 mg Dose)

Example 10: Pioglitazone HC1 Extended Release Film-Coated Tablet based on

Hypromellose

Process

CR tablets were developed based on Hypromellose (in form of Methocel™ K15M

Premium, DOW), a water-soluble hydroxypropyl methylcellulose polymer described in USP and EP. Except for the lubricants and glidants, all further excipients were included in the granulate phase prepared by aqueous wet granulation. Kernels of appropriate shape and weight were compressed to a tablet hardness of 150 N or 200 N, respectively, as measured by a diametral-compression test. The kernels were thereafter coated by an immediate release standard coat of Opadry™ II (Colorcon). The batch size was 5.6 kg.

Formulation

Table 7: Pioglitazone HC1 Prolonged Release Film-Coated Tablet based on Hypromellose

Example 11: Pioglitazone HC1 Extended Release Film-Coated Tablet based on

Hypromellose

Process

CR tablets were developed based on Hypromellose as described in Example 10.

Formulation

Table 8: Pioglitazone HC1 Prolonged Release Film-Coated Tablet based on Hypromellose

* % of tablet core mass

** 5.51 mg of Pioglitazone HC1 correspond to 5.00 mg of the pioglitazone base. Example 12: Dissolution Test with Pioglitazone HC1 Extended Release Tablet based on Hypromellose

Dissolution Test Method

A USP Apparatus 2 (Paddle Apparatus) with sinkers was used to determine the dissolution rate of the ER film-coated tablets, as well as uncoated kernels of Example 10 and 11, using a phosphate buffer pH 6.8 with 3% SDS as dissolution medium. The percentage of dissolved pioglitazone versus time was determined using a HPLC method with UV detection at 269 nm. Details of the method are described in 9. Table 9: Dissolution Method

Dissolution Results

Dissolution data are shown in Figures 4 to 7.

Uncoated tablets (“kernels”) produced using different quantities of water as granulation liquid showed practically identical dissolution curves (Fig. 4). No major difference can be found between kernels prepared using different compression force, resulting in kernels of different hardness (Fig. 5). The process can therefore be considered as robust regarding the two parameters“quantity of granulation liquid” and“compression force”. The coated tablets show a slightly slower dissolution rate than the kernels, a difference not considered to be relevant in-vivo (Fig. 6).

Overall, the formulations showed a near zero-order type release and a rate of 80% release within 5-7 hours.

The dissolution rate of Hypromellose based extended release tablets may be changed in wide limits using either different viscosity qualities of the same Hypromellose (e.g. of type 2208), or using different quantities of the same viscosity quality (Fig 7).

Example 13: PK evaluation of three oral slow-release formulations of pioglitazone in the beagle dog The PK characteristics of three oral slow-release formulations of Pioglitazone of Example 11, in comparison to an immediately available micro-suspension of the drug, were determined following single oral administrations to beagle dogs with a minimum five-day wash-out period between doses. The study followed a four-sequence and four period crossover (4x4) replicate study design, to compare the concentration profiles of three slow release tablets vs. pioglitazone-HCl micro-suspension as reference.

Methods

Formulations

Table 10: Formulations evaluated in a dog PK study

i. Pioglitazone HC1 immediate release oral suspension (5 mL) for comparison with the three slow-release tablets

ii. F 1 according to Example 11

ii. F 2 according to Example 11

ii. F 3 according to Example 11

Animal procedures

The study was performed as a single randomized four-period, four sequence crossover design. Two beagle dogs were assigned to each sequence and crossover according to a 4X4 Williams design in order to receive each of the four formulations (three ER tablets, and an oral suspension). All dogs in a period were administered on the same day. The treatment was performed once a week with an interval of at least five days between treatments. Test formulations were delivered orally for tablets or by gastric gavage using a semi-rigid rubber tube and a graduated plastic syringe for the suspension. Blood was collected by venipuncture while each animal was manually restrained. The area selected for venipuncture was shaved and disinfected. At the end of sampling, pressure was applied to stop bleeding. Samples were taken at pre-dose, and 0.5h, lh, l.5h, 2h, 3h, 4h, 6h, 8h, l2h, 24h and 36h after dosing.

Blood samples were collected into tubes containing potassium-EDTA. Samples were kept at room temperature until centrifugation (10 min, l200g, +4°C) which was performed within 45 minutes of collection. The resultant plasma was divided in 2 aliquots (approx. 250 pL each) and stored in a freezer at -20°C until analysis.

Bioanalytics and PK analysis

Laboratory concentration values for three analytes (pioglitazone and the two metabolites M- III and M-IV) were determined by a validated LC-MS method in each sample. The molar concentrations of pioglitazone, M-III and M-IV were then calculated. Pharmacokinetic analysis was performed according to standard non-compartmental methods using Phoenix WinNonlin software (v. 6.3, Certara Company, USA). The following parameters were calculated: Pioglitazone: Primary parameters AUCO-Tlast, AUCO-inf, Cmax, Tmax,

Secondary parameters Tlag, Kel, Tl/2, AUCO-inf%extrap. For metabolites M-III and M-IV: AUCO-Tlast, AUCO-inf, AUCO-inf%extrap, Cmax, Tmax, Kel, Tl/2. Individual raw data and pharmacokinetic parameters were reported to two decimal places.

Results

The extended release forms result in a lower Cmax, extended Tl/2, and smoother

concentration/time profile.

Example 14: Stability Data

Formulations Fl and F2 (c.f. example 11) were packaged into HDPE bottles with desiccant, stored under standard ICH conditions, and thereafter tested on stability. The corresponding data are shown in Tab. 11-12 (for Fl) and 13-14 (for F2).

Except for a minimal increase in dissolution rate, with formulation Fl no major changes could be seen up to 6 months when stored at 25°C. At 40°C, some increase of the impurity level could be observed with time. The dissolution rate increased somewhat more pronounced compared to the samples stored at 25°C.

Formulation F2 showed an excellent chemical stability when stored at 25°C. The dissolution rate did not change to a major extent over time. At 40°C, again a more pronounced increase in impurity level, and a more pronounced increase of the dissolution rate could be observed. Overall, the stability of the two formulations is considered as sufficient in order to develop a drug product for the market.

Table 11: Stability Summary - Pioglitazone 5 mg Film-Coated Tablets - 25°/60 % r.H. (Type FI - Table 8) Packaging Configuration: HDPE bottle (with desiccant)

n.r. = not reported

*) paddle / lOOrpm / 900ml / phosphate buffer (pH 6.8)+3%SDS Table 12: Stability Summary - Pioglitazone 5 mg Film-Coated Tablets - 40°/75 % r.H.

(Type FI - Table 8) Packaging Configuration: HDPE bottle (with desiccant)

n.r. = not reported

*) paddle / lOOrpm / 900ml / phosphate buffer (pH 6.8)+3%SDS

Table 13: Stability Summary - Pioglitazone 5 mg Film-Coated Tablets - 25°/60 % r.H. (Type F2 - Table 8) Packaging Configuration: HDPE bottle (with desiccant)

n.r. = not reported

*) paddle / lOOrpm / 900ml / phosphate buffer (pH 6.8)+3%SDS Table 14: Stability Summary - Pioglitazone 5 mg Film-Coated Tablets - 40°/75 % r.H. (Type F2 - Table 8) Packaging Configuration: HDPE bottle (with desiccant)

n.r. = not reported

*) paddle / lOOrpm / 900ml / phosphate buffer (pH 6.8)+3%SDS