Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
FORMULATIONS OF A SOMATOSTATIN MODULATOR
Document Type and Number:
WIPO Patent Application WO/2022/055880
Kind Code:
A1
Abstract:
A spray-dried solid dispersion comprising: (a) 3 - [4 -(4 -amino -piperidin-I-y l)-3 -(3,5 -difluoro-phenyI)- quinolin-6 -yl] -2 -hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof; and (b) a pharmaceutically acceptable polymer; wherein said API is dispersed in a polymer matrix formed from the pharmaceutically acceptable polymer. A tablet comprising said the spray-dried solid dispersion and one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants. Said tablet for use in the treatment of acromegaly or neuroendocrine tumors, or both, in a human by oral administration.

Inventors:
BURKE GERALD (US)
YATES IAN (US)
BULOVSKY HANNAH (US)
KYBURZ KYLE (US)
TYLER CLAYTON (US)
Application Number:
PCT/US2021/049282
Publication Date:
March 17, 2022
Filing Date:
September 07, 2021
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CRINETICS PHARMACEUTICALS INC (US)
International Classes:
A61K31/472; A61K9/14; A61K9/16; A61K9/20; A61K9/48; A61K31/4709; A61P5/08
Domestic Patent References:
WO2019143718A12019-07-25
Foreign References:
US20190382367A12019-12-19
CN108473489A2018-08-31
CN110913840A2020-03-24
Other References:
BRAZEAU ET AL., SCIENCE, vol. 179, 1973, pages 77 - 79
PRADAYROL, FEBSLETTERS, vol. 109, 1980, pages 55 - 58
ESCH ET AL., PROC. NATL. ACAD. SCI. USA, vol. 77, 1980, pages 6827 - 6831
REISINEBELL, ENDOCR REV, vol. 16, 1995, pages 427 - 442
PATELSRIKANT, TRENDS ENDOCRINOLMETAB, vol. 8, 1997, pages 398 - 405
P. H. STAHLC. G. WERMUTH: "Handbook of Pharmaceutical Salts: Properties, Selection and Use. International Union of Pure and Applied Chemistry", 2002, WILEY-VCH/VHCA
S.M. BERGEL.D. BIGHLEYD.C. MONKHOUSE, J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
"Impurities: Guidelines for Residual Solvents, Q3 C(R3", INTERNATIONAL CONFERENCE ON HARMONIZATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE (ICH, November 2005 (2005-11-01)
"Remington: The Science and Practice of Pharmacy", 1995, MACK PUBLISHING COMPANY
HOOVER, JOHN E.: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
"Pharmaceutical Dosage Forms", 1980, MARCEL DECKER
"Pharmaceutical Dosage Forms and Drug Delivery Systems", 1999, LIPPINCOTT WILLIAMS & WILKINS
Attorney, Agent or Firm:
ZUNIC, Valentin B. (US)
Download PDF:
Claims:
CLAIMS

WHAT IS CLAIMED IS:

1. A spray-dried solid dispersion comprising:

(a) 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy- benzonitrile, or a pharmaceutically acceptable salt or solvate thereof; and

(b) a pharmaceutically acceptable polymer; wherein 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy- benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, is dispersed in a polymer matrix formed from the pharmaceutically acceptable polymer.

2. The spray-dried solid dispersion of claim 1, wherein: the pharmaceutically acceptable polymer has a high glass transition temp erature (Tg).

3. The spray-dried solid dispersion of claim 1 or claim 2, wherein: the pharmaceutically acceptable polymer comprises polymers of: cellulose optionally functionalized with any combination of alkyl ethers, alkyl esters, phthalate esters; vinyl alcohol; vinyl acetate; propylene glycol; pyrrolidone; vinylpyrrolidone, oxyethylene; oxypropylene; methacrylic acid; methyl methacrylate; ethylene glycol; ethyleneglycol glycerides; ethylene oxide; propylene oxide; 2-ethyl-2-oxazoline; maleic acid; methyl vinyl ether; vinyl caprolactam; or combinations thereof.

4. The spray-dried solid dispersion of claim 1 or claim 2, wherein: the pharmaceutically acceptable polymer is hydroxypropyl methylcellulose (HPMC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), hydroxypropyl cellulose (HPC), methyl cellulose, hydroxy ethyl methyl cellulose, hydroxy ethyl cellulose acetate, hydroxy ethyl ethyl cellulose, polyvinyl alcohol polyvinyl acetate copolymers, polyethylene glycol, polyethylene glycol polypropylene glycol copolymers, poly vinylpyrrolidone (PVP), polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA), polyethylene polyvinyl alcohol copolymers, polyoxyethylenepolyoxypropylene block copolymers, cellulose acetate phthalate (CAP), hydroxypropyl methyl-cellulose phthalate (HPMCP), co-polymerc of methacrylic acid and methyl methacrylate, polyethylene glycol glycerides composed of mono-, di- and triglycerides and mono- and diesters of polyethylene glycol, hydroxypropylcellulose, copolymers of ethylene oxide and propylene oxide blocks, poly(2-ethyl-2-oxazoline), poly(maleic acid/methyl vinyl ether), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer, ethylene oxide/propylene oxide tetra functional block copolymer, d- alpha tocopheryl polyethylene glycol 1000 succinate, or combinations thereof.

- 52 - The spray-dried solid dispersion of claim 1 or claim 2, wherein: the pharmaceutically acceptable polymer is hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA). The spray-dried solid dispersion of claim 1 or claim 2, wherein: the pharmaceutically acceptable polymer is hydroxypropyl methyl cellulose acetate succinate grade M (HPMCAS-M). The spray-dried solid dispersion of claim 1 or claim 2, wherein: the pharmaceutically acceptable polymer is polyvinylpyrrolidone polyvinyl acetate copolymers in a 6:4 ratio (PVP/VA 64). The spray-dried solid dispersion of any one of claims 1-7, wherein: the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]- 2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptablepolymer is from about 1 :10 to about 10:1. The spray-dried solid dispersion of any one of claims 1-7, wherein: the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]- 2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptable polymer is from about 1 : 1 to about 1 :10. The spray-dried solid dispersion of any one of claims 1-7, wherein: the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]- 2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptablepolymer is from about 1 :4 to about 1 :6. The spray-dried solid dispersion of any one of claims 1 -7, wherein: the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]- 2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptable polymer is from about 1 :1.5 to about 1 :6. The spray-dried solid dispersion of any one of claims 1-7, wherein: the spray -dried solid dispersion comprises at least about 5% by weight of 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof. The spray-dried solid dispersion of any one of claims 1 -7, wherein: the spray -dried solid dispersion comprises at least about 10% by weight of 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof. The spray-dried solid dispersion of any one of claims 1-7, wherein:

- 53 - the spray -dried solid dispersion comprises about 15% by weight of 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof. The spray-dried solid dispersion of any one of claims 1-7, wherein: the spray -dried solid dispersion comprises about 35% by weightof 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof. The spray-dried solid dispersion of any one of claims 1-15, further comprising a nonaqueous solvent. The spray-dried solid dispersion of claim 16, wherein the non-aqueous solvent is selected from the group consisting of tert-butanol, n-propanol, n-butanol, isopropanol, ethanol, methanol, acetone, ethyl acetate, acetonitrile, methyl ethyl ketone, methyl isobutyl ketone, methyl acetate, and mixtures thereof. The spray-dried solid dispersion of claim 16 or claim 17, wherein the non-aqueous solvent is selected from the group consisting of methanol, acetone, and mixtures thereof. The spray-dried solid dispersion of any one of claims 16-18, wherein the non-aqueous solvent is methanol. The spray-dried solid dispersion of any one of claims 1-19, wherein 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, is substantially amorphous. The spray-dried solid dispersion of any one of claims 1-20 wherein 3 -[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, is 3-[4-(4-amino-piperidin-l-yl)-3- (3, 5 -difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof. A tablet comprising: the spray -dried solid dispersion of any one of claims 1-21; one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally one or more film coating agents. A tablet comprising:

- 54 - 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy- benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more disintegrant aid, one or more lubricants, one or more glidants; and optionally one or more film coating agents. The tablet of claim 23, wherein the 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer is a spray-dried solid dispersion. The tablet of any one of claims 22-24, wherein: the one or more pharmaceutical acceptable ingredients comprise microcrystalline cellulose, mannitol, pregelatinized starch croscarmellose sodium crospovidone, sodium chloride, 1 : 1 sodium chloride:potassium chloride, colloidal silicon dioxide, and magnesium stearate. The tablet of any one of claims 22-25, wherein the tablet comprises: about2% by weightto about20% by weight of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5- difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-25, wherein the tablet comprises: the tablet comprises about 2% by weight, about 3% by weight, about 4% by weight, about 5% by weight, about 6% by weight, about 7% by weight, about 8% by weight, about 9% by weight, about 10% by weight, about 11% by weight, about 12% by weight, about 13% by weight, about 14% by weight, or about 15% by weight of 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 23-27, wherein the tablet comprises about 10% by weight to about 35% by weight of the polymer matrix formed from the pharmaceutically acceptable polymer. The tablet of any one of claims 23-28, wherein the tablet comprises: about2% by weightto about 15% by weight of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5- difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable

- 55 - polymer, wherein the dispersed 3-[4-(4-amino-piperidin-l-yl)-3-(3, 5-difluoro-phenyl)- quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, in the polymer matrix is about about 20% by weight to about 35% by weight of the tablet; about 40% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents. The tablet of any one of claims 23-24, wherein the tablet comprises: about20% by weightto about40% of a spray dried dispersion of 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more disintegrant aids, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents. The tablet of claim 30, wherein: the spray dried dispersion comprises an about 15/85 to about 35/65 ratio of 3-[4-(4- amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA). The tablet of claim 23, wherein the tablet comprises: about 20% by weightto about 35% of by weight a spray dried dispersion of 3-[4-(4- amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; wherein the spray dried dispersion comprises an about 15/85 to about 35/65 ratio of 3 -[4- (4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or poly vinylpyrrolidone polyvinyl acetate copolymers (PVP/VA); about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of microcrystalline cellulose, mannitol, pregelatinized starch, croscarmellose sodium, crospovidone, sodium chloride, 1 :1 sodium chloride:potassium chloride, silicon dioxide, and magnesium stearate; optionally less than about 5% by weight of one or more film coating agents. The tablet of any one of claims 23-32, wherein the tablet comprises: about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, or about 35% by weight of a spray dried dispersion of 3-[4-(4-amino-piperidin-l-yl)-3- (3, 5 -difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; wherein the spray dried dispersion comprises an about 15/85 or about 35/65 ratio of 3-[4- (4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or poly vinylpyrrolidone polyvinyl acetate copolymers (PVP/VA); about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more disintegrant aids, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents. The tablet of any one of claims 23-32, wherein the tablet comprises: about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, or about 35% by weight of a spray dried dispersion of 3-[4-(4-amino-piperidin-l-yl)-3- (3, 5 -difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; wherein the spray dried dispersion comprises an about 15/85 or about 35/65 ratio of 3-[4- (4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or poly vinylpyrrolidone polyvinyl acetate copolymers (PVP/VA); about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of microcrystalline cellulose, mannitol, pregelatinized starch, croscarmellose sodium, crospovidone, sodium chloride, 1 :1 sodium chloride:potassium chloride, silicon dioxide, and magnesium stearate; optionally less than about 5% by weight of one or more film coating agents. The tablet of any one of claims 22-34, wherein: the tablet comprises about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, or about 80 mg of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5- difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-35, wherein: the tablet comprises about 10 mg of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-35, wherein: the tablet comprises about 20 mg of 3 -[4-(4-amino-piperidin- 1 -y l)-3 -(3 ,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-35, wherein: the tablet comprises about 30 mg of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-35, wherein: the tablet comprises about 40 mg of 3 -[4-(4-amino-piperidin- 1 -y l)-3 -(3 ,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-35, wherein: the tablet comprises about 50 mg of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. The tablet of any one of claims 22-35, wherein: the tablet comprises about 60 mg of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. A method of treating acromegaly or neuroendocrine tumors, or both, in a human comprising orally administering to the human with acromegaly or neuroendocrine tumors the tablet of any one of claims 22-41. The method of claim 42, wherein: the tablet is administered once daily. The method of claim 42 or claim 43, wherein: the tabletis administered atleast 30 minutes before a meal.

- 58 - The method of claim 43 or claim 44, wherein: the tablet is administered at least 60 minutes before a meal. The method of any one of claims 42-45, wherein: the tabletis administered with a glass of water on an empty stomach at least 30 minutes before a meal. The method of any one of claims 42-46, wherein: the bioavailability of 3-[4-(4-amino- piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof, is not substantially affected by the coadministration of proton pump inhibitors, histamine H2 -receptor antagonists, or antacids. The method of any one of claims 42-47, wherein: the tablet is coadministered with a proton pump inhibitor, histamine H2 -receptor antagonist, or antacid.

- 59 -

Description:
FORMULATIONS OF A SOMATOSTATIN MODULATOR

CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application claims benefit of U.S. Provisional Patent Application No. 63/076,024, filed on September 9, 2020, which is incoporated herein by reference in its entirety.

FIELD OF THE INVENTION

[0002] Described herein are pharmaceutical compositions and medicaments comprising a somatostatin modulator, methods of making such pharmaceutical compositions and medicaments and methods of using such pharmaceutical compositions and medicaments in the treatment of conditions, diseases, or disorders that would benefit from modulating somatostatin activity.

BACKGROUND OF THE INVENTION

[0003] Somatostatin is a peptide hormone that regulates the endocrine system and affects neurotransmission and cell proliferation via interaction with G-protein-coupled somatostatin receptors and inhibition of the release of numerous secondary hormones. Six subtype somatostatin receptor proteins have been identified (SSTR1, SSTR2a, SSTR2b, SSTR3, SSTR4, SSTR5) and are encoded by five different somatostatin receptor genes. Modulation of a particular subtype somatostatin receptor, or combination thereof, is attractive for the treatment of conditions, diseases, or disorders that would benefit from modulating somatostatin activity.

SUMMARY OF THE INVENTION

[0004] In one aspect, provided herein is a spray -dried solid dispersion comprising: (a) 3-[4-(4- amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl] -2 -hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof; and (b) a pharmaceutically acceptable polymer; wherein 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2- hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, is dispersed in a polymer matrix formed from the pharmaceutically acceptable polymer. In some embodiments, the pharmaceutically acceptable polymer has a high glass transition temperature (Tg). In some embodiments, the pharmaceutically acceptable polymer comprises polymers of cellulose optionally functionalized with any combination of alkyl ethers, alkyl esters, phthalate esters; vinyl alcohol; vinyl acetate; propylene glycol; pyrrolidone; vinylpyrrolidone, oxy ethylene; oxypropylene; methacrylic acid; methyl methacrylate; ethylene glycol; ethyleneglycol glycerides; ethylene oxide; propylene oxide; 2-ethyl-2-oxazoline; maleic acid; methyl vinyl ether; vinyl caprolactam; or combinations thereof. [0005] In some embodiments, the pharmaceutically acceptable polymer is hydroxypropyl methylcellulose (HPMC), hydroxypropyl methyl cellulose acetate succinate (HPMC AS), hydroxypropyl cellulose (HPC), methyl cellulose, hydroxy ethyl methyl cellulose, hydroxy ethyl cellulose acetate, hydroxyethyl ethyl cellulose, polyvinyl alcohol polyvinyl acetate copolymers, polyethylene glycol, polyethylene glycol polypropylene glycol copolymers, polyvinylpyrrolidone (PVP), polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA), polyethylene polyvinyl alcohol copolymers, polyoxy ethylene-poly oxypropylene block copolymers, cellulose acetate phthalate (CAP), hydroxypropyl methyl-cellulose phthalate (HPMCP), co-polymerc of methacrylic acid and methyl methacrylate, polyethylene glycol glycerides composed of mono-, di- and triglycerides and mono- and diesters of polyethylene glycol, hydroxypropylcellulose, copolymers of ethylene oxide and propylene oxide blocks, poly(2-ethyl-2-oxazoline), poly(maleic acid/methyl vinyl ether), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer, ethylene oxide/propylene oxide tetra functional block copolymer, d-alpha tocopheryl polyethylene glycol 1000 succinate, or combinations thereof. In some embodiments, the pharmaceutically acceptable polymer is hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA). In some embodiments, the pharmaceutically acceptable polymer is hydroxypropyl methyl cellulose acetate succinate grade M(HPMCAS-M). In some embodiments, the pharmaceutically acceptable polymer is polyvinylpyrrolidone polyvinyl acetate copolymers in a 6:4 ratio (PVP/VA 64).

[0006] In some embodiments, the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5- difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptable polymer is from about 1 : 10 to about 10 : 1. In some embodiments, the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)- quinolin-6-yl]-2-hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptable polymer is from about 1 : 1 to about 1 :10. In some embodiments, the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2- hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptable polymer is from about 1 :4 to about 1 :6. In some embodiments, the weight ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2-hydroxy- benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, to the pharmaceutically acceptable polymer is from about 1 :1.5 to about 1 :6.

[0007] In some embodiments, the spray-dried solid dispersion comprises at least about 5% by weight of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2-hydroxy- benzonitrile, or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises at least about 10% by weight of 3-[4-(4-amino-piperidin- l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2-hydroxy-benzo nitrile, or a pharmaceutically acceptable salt or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 15% by weight of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)- quinolin-6-yl]-2-hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof. [0008] In some embodiments, the spray-dried solid dispersion further comprises a non-aqueous solvent. In some embodiments, the non-aqueous solvent is selected from the group consisting of tert-butanol, n-propanol, n-butanol, isopropanol, ethanol, methanol, acetone, ethyl acetate, acetonitrile, methyl ethyl ketone, methyl isobutyl ketone, methyl acetate, and mixtures thereof. In some embodiments, the non-aqueous solvent is selected from the group consisting of methanol, acetone, and mixtures thereof. In some embodiments, the non-aqueous solvent is methanol.

[0009] In some embodiments, 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in- 6-yl]-2 -hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, is substantially amorphous.

[0010] In some embodiments, 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in- 6-yl]-2 -hydroxy -benzonitrile, or a pharmaceutically acceptable salt or solvate thereof, is 3-[4-(4- amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinolin-6-yl] -2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof.

[0011] In another aspect, provided herein is a tablet comprising: the spray-dried solid dispersion described herein; one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally one or more film coating agents.

[0012] In another aspect, provided herein is a tablet comprising: 3-[4-(4-amino-piperidin-l-yl)- 3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy-benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally one ormore film coating agents. In some embodiments, 3-[4-(4-amino-piperidin-l-yl)-3-(3,5- difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer is a spray- dried solid dispersion described herein. In some embodiments, the one or more pharmaceutical acceptable ingredients comprise microcrystalline cellulose, mannitol, crospovidone, colloidal silicon dioxide, and magnesium stearate. In some embodiments, the one or more pharmaceutical acceptable ingredients comprise microcrystalline cellulose, mannitol, pregelatinized starch croscarmellose sodium crospovidone, sodium chloride, 1 :1 sodium chloride:potassium chloride, colloidal silicon dioxide, and magnesium stearate.

[0013] In some embodiments, the tablet comprises about 2% by weight to about 20% by weight of 3 -[4-(4-amino-piperidin-l-yl)-3 -(3, 5-difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy- benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 2% by weight to about 15% by weight of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 2% by weight, about 3% by weight, about 4% by weight, about 5% by weight, about 6% by weight, about 7% by weight, about 8% by weight, about 9% by weight, about 10% by weight, about 11 % by weight, about 12% by weight, about 13% by weight, about 14% by weight, or about 15% by weight of 3-[4-(4-amino-piperidin-l-yl)- 3-(3,5-difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy-benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 10% by weightto about 30% by weight of the polymer matrix formed from the pharmaceutically acceptable polymer. In some embodiments, the tablet comprises about 20% by weight to about 35% by weight of the polymer matrix formed from the pharmaceutically acceptable polymer.

[0014] In some embodiments, the tablet comprises about 2% by weight to about 10% by weight of 3 -[4-(4-amino-piperidin-l-yl)-3 -(3, 5-difhioro-phenyl)-quinolin-6-yl]-2 -hydroxy- benzonitrile monohydrochloride, or solvate thereof, dispersed in about 10% by weightto about 30% by weight of a polymer matrix formed from a pharmaceutically acceptable polymer; about 40% by weightto about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents.

[0015] In some embodiments, the tablet comprises: about 2% by weight to about 10% by weight of 3 -[4-(4-amino-piperidin-l-yl)-3 -(3, 5-difhioro-phenyl)-quinolin-6-yl]-2 -hydroxy- benzonitrile monohydrochloride, or solvate thereof, dispersed in about 10% by weightto about 35% by weight of a polymer matrix formed from a pharmaceutically acceptable polymer; about 40% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents. [0016] In some embodiments, the tablet comprises: about 20% by weight to about 40% of a spray dried dispersion of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2- hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more disintegrant aids, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents.

[0017] In some embodiments, the spray dried dispersion comprises an about 15/85 to about 35/65 ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2 -hydroxy- benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA).

[0018] In some embodiments, the tablet comprises: about 20% by weightto about 35% of a spray dried dispersion of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2- hydroxy -benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; wherein the spray dried dispersion comprises an about 15/85 to about 35/65 ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA); about 60% by weightto about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of microcrystalline cellulose, mannitol, pregelatinized starch, croscarmellose sodium, crospovidone, sodium chloride, 1 :1 sodium chloride:potassium chloride, silicon dioxide, and magnesium stearate; optionally less than about 5% by weight of one or more film coating agents. [0019] In some embodiments, the tablet comprises: about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31 %, about 32%, about 33%, about 34%, or about 35% by weight of a spray dried dispersion of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; wherein the spray dried dispersion comprises an about 15/85 or about 35/65 ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6- yl]-2 -hydroxy-benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/V A); about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more di sinte grant aids, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents. [0020] In some embodiments, the tablet comprises: about 20%, about 21%, about 22%, about 23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, or about 35% by weight of a spray dried dispersion of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; wherein the spray dried dispersion comprises an about 15/85 or about 35/65 ratio of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)-quinol in-6- y 1] -2 -hydroxy-benzonitrile monohydrochloride, or solvate thereof to a polymer matrix of hydroxypropyl methyl cellulose acetate succinate (HPMCAS), or polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/V A); about 60% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of microcrystalline cellulose, mannitol, pregelatinized starch, croscarmellose sodium, crospovidone, sodium chloride, 1 :1 sodium chloride :potassium chloride, silicon dioxide, and magnesium stearate; optionally less than about 5% by weight of one or more film coating agents.

[0021] In some embodiments, the tablet comprises about 5 mg, about 10 mg, about 20 mg, about40 mg, about 60 mgor about 80 mgof 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl]-2-hydroxy-benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 10 mg, about 20 mg, about 30 mg, about 40 mg, about 50 mg, about 60 mg, about 70 mg, or about 80 mg of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5- difluoro-phenyl)-quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof. [0022] In some embodiments, the tablet comprises about 10 mgof 3-[4-(4-amino-piperidin-l- yl)-3 -(3, 5 -difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 20 mg of 3-[4-(4-amino-piperidin-l- yl)-3 -(3, 5 -difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 30 mg of 3-[4-(4-amino-piperidin-l- yl)-3 -(3, 5 -difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 40 mg of 3-[4-(4-amino-piperidin-l- yl)-3 -(3, 5 -difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 50 mg of 3-[4-(4-amino-piperidin-l- yl)-3 -(3, 5 -difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof. In some embodiments, the tablet comprises about 60 mg of 3-[4-(4-amino-piperidin-l- yl)-3 -(3, 5 -difluoro-phenyl)-quinolin-6-yl]-2 -hydroxy -benzonitrile monohydrochloride, or solvate thereof.

[0023] In one aspect, described herein is a method of treating acromegaly or neuroendocrine tumors, or both, in a human comprising orally administering to the human with acromegaly or neuroendocrine tumors any one of the spray-dired dispersion tablets described herein.

[0024] In some embodiments, the tablet is administered once daily. In some embodiments, the tablet is administered at least 30 minutes before a meal. In some embodiments, the tablet is administered at least 60 minutes before a meal. In some embodiments, the tablet is administered with a glass of water on an empty stomach at least 30 minutes before a meal. In some embodiments, the bioavailability of 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)- quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, from the tablet is not substatntially affected by the coadministration of proton pump inhibitors, histamine H2- receptor antagonists, or antacids.

[0025] Other features and advantages of the compositions, compounds, and methods described herein will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples, while indicating specific embodiments, are given by way of illustration only, since various changes and modifications within the spirit and scope of the instant disclosure will become apparent to those skilled in the art from this detailed description.

BRIEF DESCRIPTION OF THE DRAWINGS

[0026] Figure 1. Illustrates the observed dose proportionality in humans administered the HMG capsule formulation or the SDD tablet formulation of Compound A-HC1.

[0027] Figure 2. Illustrates the performance of the HMG capsule formulation and the SDD tablet formulation of Compound A-HC1 in dogs with or without pentagastrin pretreatment.

DETAILED DESCRIPTION OF THE INVENTION

[0028] Somatostatin (SST), also known as somatotropin release inhibiting factor (SRIF) was initially isolated as a 14-amino acid peptide from ovine hypothalamii (Brazeaueta/., Science 179, 77-79, 1973). An A-terminal extended 28-amino acid peptide with similar biological activity to 14-amino acid somatostatin was subsequently isolated (Pradayrol et, al., FEBS Letters, 109, 55-58, 1980; Esch etal., Proc. Natl. Acad. Sci. USA, 77, 6827-6831, 1980). SST is a regulatory peptide produced by several cell types in response to other neuropeptides, neurotransmitters, hormones, cytokines, and growth factors. SST acts through both endocrine and paracrine pathways to affect its target cells. Many of these effects are related to the inhibition of secretion of other hormones, most notably growth hormone (GH). They are produced by a wide variety of cell types in the central nervous sstem (CNS) and gut, and have multiple functions including modulation of secretion of growth hormone (GH), insulin, glucagon, as well as many other hormones that are anti-proliferative.

[0029] These pleotropic actions of somatostatins are mediated by six somatostatin receptor proteins (SSTR1, SSTR2a, SSTR2b, SSTR3, SSTR4, SSTR5). The six somatostatin receptor proteins are encoded by five different somatostatin receptor genes (Reisine and Bell, Endocr Rev. 16, 427-442, 1995; Patel and Srikant, Trends Endocrinol Metab 8, 398-405, 1997). All the receptors are members of the class-A subgroup of the G protein-coupled receptor (GPCR) superfamily. SST2A receptor is the most widely expressed subtype in human tumors and is the dominant receptor by which GH secretion is suppressed. Unless otherwise stated, the term SSTR2 means SSTR2a.

[0030] It is possible to selectively modulate any one of the somatostatin receptor subtypes, or combination thereof. In some embodiments, selectively modulating any one of the somatostatin receptor subtypes relative to the other somatostatin receptor subtypes, or combination thereof, is useful in a variety of clinical applications. In some embodiments, selectively modulating any one of the somatostatin receptor subtypes relative to the other somatostatin receptor subtypes reduces unwanted side effects in a variety of clinical applications.

[0031] For example, modulation of SSTR2 activity mediates the inhibition of growth hormone (GH) release from the anterior pituitary and glucagon release from pancreas. SSTR2 is also implicated in many other biological functions such as, but not limited to, cell proliferation, nociception, inflammation, and angiogenesis. In some embodiments, a selective SSTR2 modulator is used in the treatment of acromegaly, gut neuroendocrine tumors, pain, neuropathies, nephropathies, and inflammation, as well as retinopathies resulting from aberrant blood vessel growth. In some other embodiments, a selective SSTR2 modulator is used in the treatment of arthritis, pain, cancer, inflammatory bowel disease, irritable bowel syndrome, Crohn’s disease, Cushing’s disease, acute lung injury, acute respiratory distress syndrome, and ophthalmic disorders such as age-related macular degeneration (AMD), diabetic retinopathy, diabetic macular edema, and Graves ophthalmology, among others.

[0032] In some embodiments, SSTR3 agonists inhibit insulin secretion. In some embodiments, SSTR4 agonists exhibit anti-inflammatory and anti-nociceptive effects. In some embodiments, SSTR5 agonists inhibit insulin secretion. In addition, SSTR5 has also been implicated to modulate the release of growth hormone. [0033] Somatostatin peptide and its receptor subtypes are also widely expressed in the brain and disruption or diminishment of their activity is potentially involved in several psychiatric and neurodegenerative diseases. For example, concentrations of somatostatin in the cebrebral cortex and hippocampus are reduced in schizophrenics and one of the most consistent neuropathologic findings in this patient group is a deficit in cortical inhibitory interneurons expressing somatostatin. Somatostatin is also highly expressed in brain regions associated with seizures and has also been implicated as having an important role in epilepsy. Somatostatin levels are diminshed in the hippocampi of Alzheimer’s and Parkinson’s patients, suggesting that restoration of its signaling as a potential drug target for neurodegeneration.

[0034] In one aspect, the compound 3-[4-(4-amino-piperi din-l-yl)-3-(3, 5-difluoro-phenyl)- quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, or solvate thereof, is a selective nonpeptide SST2 biased agonist that is amenable to oral administration to a mammal in need of treatment with a somatostatin modulator.

[0035] In some embodiments, the somatostatin receptor modulator described herein has utility over a wide range of therapeutic applications. In some embodiments, the somatostatin receptor modulator described herein is used in the treatment of a variety of diseases or conditions such as, but not limited to acromegaly, neuroendocrine tumors, retinopathies and other ophthalmic disorders, neuropathy, nephropathy, respiratory diseases, cancers, pain, neurodegenerative diseases, inflammatory diseases, as well as psychiatric and neurodegenerative disorders. In some embodiments, the somatostatin receptor modulator described herein is used in the treatment of acromegaly, neuroendocrine tumors, or both in a mammal. In some embodiments, the somatostatin receptor modulator described herein is used in the treatment of acromegaly in a mammal. In some embodiments, the somatostatin receptor modulator described herein is used in the treatment of neuroendocrine tumors.

[0036] In some embodiments, the somatostatin receptor modulator described herein inhibits the secretion of various hormones and trophic factors in mammals. In some embodiments, the somatostatin receptor modulator described herein is used to suppress certain endocrine secretions, such as, but not limited to GH, insulin, glucagon and prolactin. The suppression of certain endocrine secretionsis useful in the treatment of disorders such as acromegaly; endocrine tumors such as carcinoids, VIPomas, insulinomas and glucagonomas; or diabetes and diabetes- related pathologies, including retinopathy, neuropathy and nephropathy. In some embodiments, the somatostatin receptor modulator described herein is used to suppress exocrine secretions in the pancreas, stomach and intestines, for the treatment of disorders such as pancreatitis, fistulas, bleeding ulcers and diarrhea associated with such diseases as AIDS or cholera. Disorders involving autocrine or paracrine secretions of trophic factors such as IGF-1 (as well as some endocrine factors) which may be treated by administration of the compounds described herein include cancers of the breast, prostate, and lung (both small cell and non-small cell epidermoids), as well as hepatomas, neuroblastomas, colon and pancreatic adenocarcinomas (ductal type), chondrosarcomas, and melanomas, diabetic retinopathy, and atherosclerosis associated with vascular grafts and restenosis following angioplasty.

[0037] In some embodiments, the somatostatin receptor modulator described herein is used to suppress the mediators of neurogenic inflammation (e.g. substance P or the tachykinins), and may be used in the treatment of rheumatoid arthritis; psoriasis; topical inflammation such as is associated with sunburn, eczema, or other sources of itching; inflammatory bowel disease; irritable bowel syndrome; allergies, including asthma and other respiratory diseases In some other embodiments, the somatostatin receptor modulators described herein function as neuromodulators in the central nervous system and are useful in the treatment of Alzheimer's disease and other forms of dementia, pain, and headaches. In some embodiments, the somatostatin receptor modulator described herein provides cytoprotection in disorders involving the splanchnic blood flow, including cirrhosis and oesophagal varices.

[0038] Compound A is a somatostatin modulator that is useful in the methods of treatment described herein.

Compound A

[0039] As used herein, Compound A refers to 3-(4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro- phenyl)-quinolin-6-yl)-2-hydroxy-benzonitrile, which has the chemical structure shownbelow.

[0040] Compound A is a selective nonpeptide SST2 biased agonist. In clinical studies, Compound A was shown to have an estimated bioavailability of about 70% and an observed half life of about 42 to about 50 hours. In some embodiments, Compound A is used to treat acromegaly, neuroendocrine tumors, or both. In some embodiments, Compound A is used to treat acromegaly. In some embodiments, Compound A is used to treat neuroendocrine tumors.

[0041] In some embodiments, the free base form of Compound A is incorporated into the formulations described herein. In some embodiments, Compound A is incorporated into the formulations described herein as a pharmaceutically acceptable salt. In some embodiments, Compound A is incorporated into the formulations described herein as a pharmaceutically acceptable solvate.

[0042] “Pharmaceutically acceptable,” as used herein, refers a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively nontoxic, i.e., the material is administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.

[0043] The term “pharmaceutically acceptable salt” refers to a form of a therapeutically active agent that consists of a cationic form of the therapeutically active agent in combination with a suitable anion, or in alternative embodiments, an anionic form of the therapeutically active agent in combination with a suitable cation. Handbook of Pharmaceutical Salts: Properties, Selection and Use. International Union of Pure and Applied Chemistry, Wiley -VCH 2002. S.M. Berge, L.D. Bighley, D.C. Monkhouse, J. Pharm. Sci. 1977, 66, 1-19. P. H. Stahl and C. G. Wermuth, editors, Handbook of Pharmaceutical Salts: Properties, Selection and Use, Weinheim/Zurich:Wiley-VCH/VHCA, 2002. Pharmaceutical salts typically are more soluble and more rapidly soluble in stomach and intestinal juices than non-ionic species and so are useful in solid dosage forms. Furthermore, because their solubility often is a function of pH, selective dissolution in one or another part of the digestive tract is possible and this capability can be manipulated as one aspect of delayed and sustained release behaviours. Also, because the saltformingmolecule can be in equilibrium with a neutral form, passage through biological membranes can be adjusted.

[0044] In some embodiments, pharmaceutically acceptable salts are obtained by reacting a compound disclosed herein with an acid. In some embodiments, the compound disclosed herein (i.e. free base form) is basic and is reacted with an organic acid or an inorganic acid. Inorganic acids include, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, nitric acid, and metaphosphoric acid. Organic acids include, but are not limited to, 1 -hydroxy -2-naphthoic acid; 2,2-dichloroacetic acid; 2 -hydroxy ethanesulfonic acid; 2- oxoglutaric acid; 4-acetamidobenzoic acid; 4 -aminosalicylic acid; acetic acid; adipic acid; ascorbic acid (L); aspartic acid (L); benzenesulfonic acid; benzoic acid; camphoric acid (+); camphor- 10-sulfonic acid (+); capric acid (decanoic acid); caproic acid (hexanoic acid); caprylic acid (octanoic acid); carbonic acid; cinnamic acid; citric acid; cyclamic acid; dodecylsulfuric acid; ethane- 1,2-disulfonic acid; ethanesulfonic acid; formic acid; fumaric acid; galactaric acid; gentisic acid; glucoheptonic acid (D); gluconic acid (D); glucuronic acid (D); glutamic acid; glutaric acid; glycerophosphoric acid; glycolic acid; hippuric acid; isobutyric acid; lactic acid (DL); lactobionic acid; lauric acid; maleic acid; malic acid (- L); malonic acid; mandelic acid (DL); methanesulfonic acid; naphthalene- 1,5 -disulfonic acid; naphthalene-2-sulfonic acid; nicotinic acid; oleic acid; oxalic acid; palmitic acid; pamoic acid; phosphoric acid; proprionic acid; pyroglutamic acid (- L); salicylic acid; sebacic acid; stearic acid; succinic acid; sulfuric acid; tartaric acid (+ L); thiocyanic acid; toluenesulfonic acid (/?); andundecylenic acid.

[0045] In some embodiments, Compound A is incorporated into the formulations described herein as a pharmaceutically acceptable salt form that is selected from Compound A hydrochloride and Compound A methanesulfonic acid. In some embodiments, the Compound A salt form is Compound A monohydrochloride. In some embodiments, the Compound A salt form is Compound A dihydrochloride. In some embodiments, the Compound A salt form is Compound A monomethanesulfonic acid. In some embodiments, the Compound A salt form is Compound A dimethanesulfonic acid.

[0046] In one aspect, Compound A monohydrochloride (Compound A-HC1) is incorporated into the pharmaceutical compositions described herein. Compound A monohydrochloride (Compound A-HC1), also known as 3-[4-(4-amino-piperidin-l-yl)-3-(3,5-difluoro-phenyl)- quinolin-6-yl]-2-hydroxy -benzonitrile monohydrochloride, has the following structure:

[0047] In some embodiments, the Compound A salt form is amorphous. In some embodiments, Compound A monohydrochloride is amorphous

[0048] In some embodiments, the Compound A salt form is crystalline. In some embodiments, Compound A monohydrochloride is crystalline.

[0049] It should be understood that a reference to a pharmaceutically acceptable salt includes the solvent addition forms. In some embodiments, solvates contain either stoichiometric or non - stoichiometric amounts of a solvent, and are formed during the process of crystallization with pharmaceutically acceptable solvents such as water, ethanol, and the like. Hydrates are formed when the solvent is water, or alcoholates are formed when the solvent is alcohol. Solvates of compounds described herein are conveniently prepared or formed during the processes described herein. In addition, the compounds provided herein optionally exist in unsolvated as well as solvated forms. [0050] Therapeutic agents that are administrableto mammals, such as humans, must be prepared by following regulatory guidelines. Such government regulated guidelines are referred to as Good Manufacturing Practice (GMP). GMP guidelines outline acceptable contamination levels of active therapeutic agents, such as, for example, the amount of residual solvent in the final product. Preferred solvents are those that are suitable for use in GMP facilities and consistent with industrial safety concerns. Categories of solvents are defined in, for example, the International Conference on Harmonization of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), “Impurities: Guidelines for Residual Solvents, Q3C(R3), (November 2005).

[0051] Solvents are categorized into three classes. Class 1 solvents are toxic and are to be avoided. Class 2 solvents are solvents to be limited in use during the manufacture of the therapeutic agent. Class 3 solvents are solvents with low toxic potential and of lower risk to human health. Data for Class 3 solvents indicate that they are less toxic in acute or short-term studies and negative in genotoxicity studies.

[0052] Class 1 solvents, which are to be avoided, include: benzene; carbon tetrachloride; 1 ,2- dichloroethane; 1,1 -dichloroethene; and 1,1,1-trichloroethane.

[0053] Examples of Class 2 solvents are: acetonitrile, chlorobenzene, chloroform, cyclohexane, 1,2-dichloroethene, dichloromethane, 1 ,2-dimeth oxy ethane, N,N-dimethylacetamide, N,N- dimethylformamide, 1,4-dioxane, 2-ethoxy ethanol, ethyleneglycol, formamide, hexane, methanol, 2-methoxyethanol, methylbutyl ketone, methylcyclohexane, N-methylpyrrolidine, nitromethane, pyridine, sulfolane, tetralin, toluene, 1,1,2-trichloroethene and xylene.

[0054] Class 3 solvents, which possess low toxicity, include: acetic acid, acetone, anisole, 1 - butanol, 2-butanol, butyl acetate, tert-butylmethyl ether (MTBE), cumene, dimethyl sulfoxide, ethanol, ethyl acetate, ethyl ether, ethyl formate, formic acid, heptane, isobutyl acetate, isopropyl acetate, methyl acetate, 3 -methyl- 1-butanol, methylethyl ketone, methylisobutyl ketone, 2- methyl-1 -propanol, pentane, 1 -pentanol, 1 -propanol, 2-propanol, propyl acetate, and tetrahydrofuran.

[0055] Residual solvents in active pharmaceutical ingredients (APIs) originate from the manufacture of API. In most cases, the solvents are not completely removed by practical manufacturing techniques. Appropriate selection of the solvent for the synthesis of APIs may enhance the yield, or determine characteristics such as crystal form, purity, and solubility. Therefore, the solvent is a critical parameter in the synthetic process.

[0056] In some embodiments, compositions comprising Compound A-HC1 include a residual amount of an organic solvent(s). In some embodiments, compositions comprising Compound A- HC1 comprise a residual amount of a Class 2 or Class 3 solvent. In some embodiments, compositions comprising Compound A-HC1 comprise a residual amount of a solvent selected from ethyl acetate, isopropyl acetate, tert-butylmethylether, heptane, isopropanol, methanol, acetone, dimethylformamide, tetrahydrofuran, 1,4-dioxane, 2-methyltetrahydrofuran, toluene, and ethanol.

[0057] Unless otherwise stated, the following terms used in this application have the definitions given below. The use of the term “including” as well as other forms, such as “include”, “includes,” and “included,” is not limiting. The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. [0058] The term “modulate” as used herein, means to interact with a target either directly or indirectly so as to alter the activity of the target, including, by way of example only, to enhance the activity of the target, to inhibit the activity of the target, to limit the activity of the target, or to extend the activity of the target.

[0059] The term “modulator” as used herein, refers to a molecule that interacts with a target either directly or indirectly. The interactions include, but are not limited to, the interactions of an agonist, partial agonist, an inverse agonist, antagonist, degrader, or combinations thereof. In some embodiments, a modulator is an agonist.

[0060] The terms "administer," "administering", "administration," and the like, as used herein, refer to the methods that may be used to enable delivery of compounds or compositions to the desired site of biological action. These methods include, but are not limited to oral routes of administration. Those of skill in the art are familiar with administration techniques that can be employed with the compounds and methods described herein. In some embodiments, the compounds and compositions described herein are administered orally.

[0061] The terms “co-administration” or the like, as used herein, are meant to encompass administration of the selected therapeutic agents to a single patient, and are intended to include treatment regimens in which the agents are administered by the same or different route of administration or at the same or different time.

[0062] The terms “effective amount” or “therapeutically effective amount,” as used herein, refer to a sufficient amount of an agent or a compound being administered, which will relieve to some extent one or more of the symptoms of the disease or condition being treated. The result includes reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. For example, an “effective amount” for therapeutic uses is the amount of the composition comprising a compound as disclosed herein required to provide a clinically significant decrease in disease symptoms. An appropriate “effective” amount in any individual case is optionally determined using techniques, such as a dose escalation study.

[0063] The terms “enhance” or “enhancing,” as used herein, means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system.

[0064] The term “pharmaceutical combination” as used herein, means a product that results from the mixing or combining of more than one active ingredient and includes both fixed and non-fixed combinations of the active ingredients. The term “fixed combination” means that the active ingredients, e.g. a compound disclosed herein, or a pharmaceutically acceptable salt thereof, and a co-agent, are both administered to a patient simultaneously in the form of a single entity or dosage. The term “non-fixed combination” means that the active ingredients, e.g. a compound disclosed herein, or a pharmaceutically acceptable salt thereof, and a co-agent, are administered to a patient as separate entities either simultaneously, concurrently or sequentially with no specific intervening time limits, wherein such administration provides effective levels of the two compounds in the body of the patient. The latter also applies to cocktail therapy, e.g. the administration of three or more active ingredients.

[0065] The terms “article of manufacture” and “kit” are used as synonyms.

[0066] The term “subject” or “patient” encompasses mammals. Examples of mammals include, but are not limited to, any member of the Mammalian class: humans, non -human primates such as chimpanzees, and other apes and monkey species; farm animals such as cattle, horses, sheep, goats, swine; domestic animals such as rabbits, dogs, and cats; laboratory animals including rodents, such as rats, mice and guinea pigs, and the like. In one aspect, the mammal is a human. [0067] The terms “treat,” “treating” or “treatment,” as used herein, include alleviating, abating or ameliorating at least one symptom of a disease or condition, preventing additional symptoms, inhibiting the disease or condition, e.g., arresting the development of the disease or condition, relieving the disease or condition, causing regression of the disease or condition, relieving a condition caused by the disease or condition, or stopping the symptoms of the disease or condition either prophy lactically and/or therapeutically.

Pharmaceutical Compositions

[0068] In some embodiments, the compounds described herein are formulated into pharmaceutical compositions. Pharmaceutical compositions are formulated in a conventional manner using one or more pharmaceutically acceptable inactive ingredients that facilitate processing of the active compounds into preparations that are used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. A summary of pharmaceutical compositions described herein is found, for example, in Remington: The Science and Practice of Pharmacy, Nineteenth Ed (Easton, Pa.: Mack Publishing Company, 1995); Hoover, John E., Remington’s Pharmaceutical Sciences, Mack Publishing Co., Easton, Pennsylvania 1975; Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms, Marcel Decker, New York, N.Y., 1980; and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed. (Lippincott Williams & Wilkinsl999), herein incorporated by reference for such disclosure.

[0069] In some embodiments, the compounds described herein are administered either alone or in combination with pharmaceutically acceptable carriers, excipients or diluents, in a pharmaceutical composition. Administration of the compounds and compositions described herein can be effected by any method that enables delivery of the compounds to the site of action. These methods include, though are not limited to delivery via enteral routes (including oral) administration, although the most suitable route may depend upon for example the condition and disorder of the recipient.

[0070] In some embodiments, pharmaceutical compositions suitable for oral administration are presented as discrete units such as capsules or tablets each containing a predetermined amount of the active ingredient; or as a powder or granules.

[0071] Pharmaceutical compositions which can be used orally include tablets, push -fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. Tablets may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets maybe prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with binders, inert diluents, or lubricating, surface active or dispersing agents. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. In some embodiments, the tablets are coated or scored and are formulated so as to provide slow or controlled release of the active ingredient therein. All formulations for oral administration should be in dosages suitable for such administration. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In some embodiments, stabilizers are added. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets for identification or to characterize different combinations of active compound doses.

[0072] Conventional techniques to manufacture solid oral dosage forms include, but are not limited to, one or a combination of methods: (1) dry mixing, (2) direct compression, (3) milling, (4) dry or non-aqueous granulation, or (5) wet granulation. See, e.g., Lachman etal., The Theory and Practice of Industrial Pharmacy (1986). Other methods include, e.g., spray drying, pan coating, melt granulation, granulation, fluidizedbed spray drying or coating (e.g., wurster coating), tangential coating, top spraying, tableting, extruding and the like.

[0073] It should be understood that in addition to the ingredients particularly mentioned above, the compounds and compositions described herein may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.

[0074] Provided herein are tablets comprising Compound A, or a pharmaceutically acceptable salt thereof. In some embodiments, the tablet comprises: Compound A-HC1, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally one or more film coating agents.

[0075] In some embodiments, described herein is a spray-dried solid dispersion comprising (a) Compound A-HC1, or solvate thereof; and (b) a pharmaceutically acceptable polymer; wherein Compound A-HC1, or solvate thereof, is dispersed in a polymer matrix formed from the pharmaceutically acceptable polymer.

[0076] In some embodiments, described herein are tablets prepared with the spray-dried solid dispersions described herein.

Spray-Dried Solid Dispersion (SDD)

[0077] The amorphous state for most small molecule drugs is thermodynamically unstable and, unless the glass transition temperature (Tg) is sufficiently high, also kinetically unstable. However, the amorphous state can be stabilized by dilution of the drug in an excipient matrix. When an amorphous molecule is dispersed in a high Tg matrix, low molecular mobility provides a diffusion barrier which inhibits molecular mobility that is required for phase separation upon storage. Phase separation into drug rich domains is the precursor to forming crystal nuclei and eventually widespread crystallization which results in a lost solubility advantage. In some embodiments, pharmaceutically acceptable polymers for use in preparing spray-dried solid dispersions are polymers with a high Tg. In cases when the active pharmaceutical ingredient (API) and excipient are not thermodynamically miscible with one another in the solid state, the spray-dried dispersion (SDD) is formulated such that the resulting Tg of the mixture, including absorbed water, is at least 10 °C to 20 °C greater than typical storage conditions. Further, considerations must be made with respect to water uptake during storage, either through selection of a non-hygroscopic polymer or packaging configuration, as adsorbed water will plasticize the dispersion and lower the Tg.

[0078] In some embodiments, Compound A-HC1, or solvate thereof, in the spray-dried solid dispersions described herein is substantially amorphous.

[0079] In some embodiments, the pharmaceutically acceptable polymer comprises polymers of : cellulose optionally functionalized with any combination of alkyl ethers, alkyl esters, phthalate esters; vinyl alcohol; vinyl acetate; propylene glycol; pyrrolidone; vinylpyrrolidone, oxyethylene; oxypropylene; methacrylic acid; methyl methacrylate; ethylene glycol; ethylene glycol glycerides; ethylene oxide; propylene oxide; 2-ethyl-2-oxazoline; maleic acid; methyl vinyl ether; vinyl caprolactam; or combinations thereof.

[0080] In some embodiments, the pharmaceutically acceptable polymer is hydroxypropyl methylcellulose (HPMC), hydroxypropyl methyl cellulose acetate succinate (HPMCAS), hydroxypropyl cellulose (HPC), methyl cellulose, hydroxy ethyl methyl cellulose, hydroxy ethyl cellulose acetate, hydroxyethyl ethyl cellulose, polyvinyl alcohol polyvinyl acetate copolymers, polyethylene glycol, polyethylene glycol polypropylene glycol copolymers, polyvinylpyrrolidone (PVP), polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA), polyethylene polyvinyl alcohol copolymers, poly oxy ethylene-poly oxypropylene block copolymers, cellulose acetate phthalate (CAP), hydroxypropyl methyl-cellulose phthalate (HPMCP), co-polymerc of methacrylic acid and methyl methacrylate, polyethylene glycol glycerides composed of mono-, di- and triglycerides and mono- and diesters of polyethylene glycol, hydroxypropylcellulose, copolymers of ethylene oxide and propylene oxide blocks, poly(2-ethyl-2-oxazoline), poly(maleic acid/methyl vinyl ether), polyvinyl caprolactam-polyvinyl acetate-polyethylene glycol graft copolymer, ethylene oxide/propylene oxide tetra functional block copolymer, d-alpha tocopheryl polyethylene glycol 1000 succinate, or combinations thereof.

[0081] In some embodiments, the spray-dried dispersion further comprises a dispersion polymer. Dispersion polymers are selected from hydroxypropyl methylcellulose (HPMC), hypromellose acetate succinate (hydroxypropyl methyl cellulose acetate succinate; HPMCAS, such as HPMCAS-H, HPMCAS-L, or HPMCAS-M), hydroxypropyl cellulose (HPC), methyl cellulose, hydroxy ethyl methyl cellulose, hydroxy ethyl cellulose acetate, hydroxy ethyl ethyl cellulose, polyvinyl alcohol polyvinyl acetate copolymers, polyethylene glycol, polyethylene glycol polypropylene glycol copolymers, polyvinylpyrrolidone (PVP), polyvinylpyrrolidone polyvinyl acetate copolymers (PVP/VA), polyethylene polyvinyl alcohol copolymers, poly oxy ethylene-poly oxypropylene block copolymers, and combinations thereof.

[0082] HPMCAS is a cellulosic polymer with four types of substituents semirandomly substituted on the hydroxyls: methoxy, hydroxypropyloxy, acetate, and succinate. The polymer is available in three grades: L, M and H, based on the content of acetyl and succinoyl groups (wt%) in the HPMCAS molecule. Grade L: 5-9% by weight acetate, 14-18% by weight succinate, 20-24% by weight methoxy, 5 -9% by weight hydroxypropyloxy. Grade M: 7-11% by weight acetate, 10-14% by weight succinate, 21-25%by weight methoxy, 5-9% by weight hydroxypropyloxy. Grade H: 10-14% by weight acetate, 4-8% by weight succinate, 22-26% by weight methoxy, 6-10% by weight hydroxypropyloxy.

[0083] In some embodiments, the pharmaceutically acceptable polymer is selected from PVP/VA 64, PVP 30, HPMCAS-L, HPMCAS-M, HPMCAS-H, Eudragit LI 00-55, poly(methacrylic acid-co-methyl methacrylate)(PMMAMA, or trade name Eudragit LI 00), Eudragit EPO, HPMC E15, HPMC E3, HPMC E5, HPMCP-HP55, and Soluplus.

[0084] In some embodiments, the pharmaceutically acceptable polymer is selected from PVP/VA 64 and HPMCAS-M. In some embodiments, the pharmaceutically acceptable polymer is PVP/VA 64. In some embodiments, the pharmaceutically acceptable polymer is HPMCAS-M. [0085] In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is between about 1 : 10 and about 10:1. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is between about 1 : 1 and about 1 : 10. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is between about 1 :3 and about 1 :8. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is between about 1 :4 and about 1 :7. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is between about 1 :4 and about 1 :6. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is between about 1 :5 and about 1 :6. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 : 10. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :9. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :8. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :7. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :6. In some embodiments, the weight ratio of Compound A- HC1, or solvate thereof, to the dispersion polymer is about 1 :5. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :4. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :3. In some embodiments, the weight ratio of Compound A-HC1, or solvate thereof, to the dispersion polymer is about 1 :2. In some embodiments, the weight ratio of Compound A- HC1, or solvate thereof, to the dispersion polymer is about 1 :1.

[0086] In some embodiments, the spray-dried solid dispersion comprises at least 5% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises at least 10% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray -dried solid dispersion comprises at least 15% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises at least 20% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray- dried solid dispersion comprises at least 25% by weight of Compound A-HC1, or solvate thereof. The % amounts are calculated based on the free base, i.e. Compound A.

[0087] In some embodiments, the spray-dried solid dispersion comprises about 5%, about 6%, ab out 7 % , ab out 8 % , ab out 9 % , ab out 10 %, ab out 11 %, ab out 12 %, ab out 13 % , ab out 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about 23%, about 24%, or about 25% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, about 20%, about 21%, about 22%, about23%, about 24%, about 25%, about 26%, about 27%, about 28%, about 29%, about 30%, about 31%, about 32%, about 33%, about 34%, or about 35%by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 15% of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 35% of Compound A-HC1, or solvate thereof. The % amounts are calculated based on the free base, i.e. Compound A.

[0088] In some embodiments, the spray-dried solid dispersion comprises about 5% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 6% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 7% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 8% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 9% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 10% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 11% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 12% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 13% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 14% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 15% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 16% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 17% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 18% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 19% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 20% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 21% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 22% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 23% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 24% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 25% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 25% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 27% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 28% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 29% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 30% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 31% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 32% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 33% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 34% by weight of Compound A-HC1, or solvate thereof. In some embodiments, the spray-dried solid dispersion comprises about 35% by weight of Compound A-HC1, or solvate thereof. The % amounts are calculated based on the free base, i.e. Compound A.

[0089] In some embodiments, the spray-dried solid dispersion further comprises a non-aqueous solvent. In some embodiments, the non-aqueous solvent is present in detectable amounts. In some embodiments, the spray-dried solid dispersion is non-aqueous solvent free.

[0090] In some embodiments, the spray-dried solid dispersion further comprises a nonaqueous solvent selected from the group consisting of tert -butanol, n-propanol, n-butanol, isopropanol, ethanol, methanol, acetone, ethyl acetate, acetonitrile, methyl ethyl ketone, methyl isobutyl ketone, methyl acetate, and mixtures thereof. In some embodiments, the spray-dried solid dispersion further comprises a non-aqueous solvent selected from the group consisting of methanol, acetone, and mixtures thereof. In some embodiments, the spray-dried solid dispersion further comprises methanol.

Tablets

[0091] In one aspect, described herein is a tablet comprising: Compound A-HC1, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer; one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally one or more film coating agents.

[0092] In some embodiments, Compound A-HC1, or solvate thereof, dispersed in a polymer matrix formed from a pharmaceutically acceptable polymer is the spray-dried solid dispersion described herein.

[0093] In some embodiments, tablets comprise about 2% by weight to about 20% by weight of Compound A-HC1, or solvate thereof. In some embodiments, tablets comprise about 2% by weight to about 15% by weight of Compound A-HC1, or solvate thereof.

[0094] In some embodiments, tablets comprise about 10% by weightto about 30% by weight of the polymer matrix formed from the pharmaceutically acceptable polymer. In some embodiments, tablets comprise about 20% by weight to about 35% by weight of the polymer matrix formed from the pharmaceutically acceptable polymer.

[0095] In some embodiments, tablets comprise about 2% by weightto about 10% by weight of Compound A-HC1, or solvate thereof, dispersed in about 10% by weightto about 30% by weight of a polymer matrix formed from a pharmaceutically acceptable polymer. [0096] In some embodiments, tablets comprise about 2% by weight to about 10% by weight of Compound A-HC1, or solvate thereof, dispersed in about 10% by weight to about 30% by weight of a polymer matrix formed from a pharmaceutically acceptable polymer; about 40% by weight to about 80% by weight of one or more pharmaceutical acceptable ingredients selected from the group consisting of one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants; and optionally less than about 5% by weight of one or more film coating agents.

[0097] In some embodiments, in addition to the spray-dried solid dispersion, additional excipients in the tablets comprise one or more diluents, one or more disintegrants, one or more lubricants, one or more glidants, or any combination thereof. In some embodiments, in addition to the spray-dried solid dispersion, additional excipients in the tablets comprise microcrystalline cellulose, mannitol, crospovidone, colloidal silicon dioxide, and magnesium stearate.

[0098] In some embodiments, the tablet comprises one or more fillers/binders/diluents. Fillers/binders/diluents are selected from celluloses (such as microcrystalline cellulose, carboxymethylcellulose, ethyl cellulose and methyl cellulose), starch, gelatin, sugars (such as sucrose, glucose, dextrose, mannitol, and lactose), natural and synthetic gums (such as acacia, sodium alginate, panwar gum, and ghatti gum), poly vinylpyrrolidinone, polyethylene glycol, waxes, and any combinations thereof. In some embodiments, tablets comprise microcrystalline cellulose, and mannitol.

[0099] In some embodiments, the one or more fillers/binders/diluents in the tablets described herein comprise between about 20% and about 80% by weight of the total tablet weight. In some embodiments, the one or more fillers/binders/diluents in the tablets described herein comprise between about 40% and about 65% by weight of the total tablet weight. In some embodiments, the one or more fillers/binders/diluents in the tablets described herein comprise between about 50% and about 65% by weight of the total tablet weight. In some embodiments, the one or more fillers/binders/diluents in the tablets described herein comprise about 45%, about 50%, about 55%, about 60%, about 65%, or about 70% by weight of the total tablet weight. In some embodiments, the one or more fillers/binders/diluents in the tablets described herein comprise about 58% by weight of the total tablet weight. In some embodiments, less than 70% by weight, less than 65% by weight, less than 60% by weight, less than 55% by weight, or less than 50% by weight of the total tablet weight comprise one or more fillers/binders/diluents. In some embodiments, less than 60% by weight of the total tablet weight comprise one or more fillers/binders/diluents. [00100] In some embodiments, tablets comprise one or more disintegrants. Disintegrants are selected from croscarmellose sodium, crospovidone, sodium starch glycolate, veegum HV, methylcellulose, agar, bentonite, cellulose, carboxymethyl cellulose, and any combination thereof. In some embodiments, tablets comprise crospovidone.

[00101] In some embodiments, the one or more disintegrants in the tablets described herein comprise between about 2% and about 30% by weight of the total tablet weight. In some embodiments, the one or more disintegrants in the tablets described herein comprise between about 5% and about 20% by weight of the total tablet weight. In some embodiments, the one or more disintegrants in the tablets described herein comprise between about 10% and about 20% by weight of the total tablet weight. In some embodiments, the one or more disintegrants in the tablets described herein comprise about 5%, about 6%, about 7%, about 8%, about 9%, about 10%, about 11%, about 12%, about 13%, about 14%, about 15%, about 16%, about 17%, about 18%, about 19%, or about 20% by weight of the total tablet weight. In some embodiments, the one or more disintegrants in the tablets described herein comprise about 15% by weight of the total tablet weight. In some embodiments, less than 20% by weight of the total tablet weight comprise one or more disintegrants.

[00102] In some embodiments, tablets comprise one or more lubricants. Lubricants are selected from talc, magnesium stearate, calcium stearate, stearic acid, sodium stearyl fumarate, glyceryl behenate, hydrogenated vegetable oils, polyethylene glycol, and any combinations thereof. In some embodiments, tablets comprise magnesium stearate.

[00103] In some embodiments, the one or more lubricants in the tablets described herein comprise between about 0.1 % and about 5% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise between about 0.1% and about 2% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise between about 0.1% and about 1% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about O.7%, about 0.8%, about 0.9%, or about 1% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise about 0.5% by weight of the total tablet weight. In some embodiments, less than 2% by weight of the total tablet weight comprise one or more lubricants. In some embodiments, less than 1% by weight of the total tablet weight comprise one or more lubricants.

[00104] In some embodiments, tablets comprise one or more glidants. A glidantis a substance that is added to a powder to improve its flowability. Examples of glidants include magnesium stearate, colloidal silicon dioxide, starch and talc. In some embodiments, tablets comprise colloidal silicon dioxide.

[00105] In some embodiments, the one or more lubricants in the tablets described herein comprise between about 0. 1 % and about 5% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise between about 0.1% and about 2% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise between about 0.5% and about 1.5% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise about 0.1%, about 0.2%, about 0.3%, about 0.4%, about 0.5%, about 0.6%, about 0.7%, about 0.8%, about 0.9%, about 1%, about 1 .1%, about 1 .2%, about 1.3%, about 1 .4%, about 1.5%, about 1 .6%, about 1.7%, about 1.8%, about 1.9%, or about 2% by weight of the total tablet weight. In some embodiments, the one or more lubricants in the tablets described herein comprise about 1% by weight of the total tablet weight. In some embodiments, less than 2% by weight of the total tablet weight comprise one or more lubricants. In some embodiments, less than 1.5% by weight of the total tablet weight comprise one or more lubricants.

Additional Excipients

[00106] In some embodiments, the tablet described herein comprises additional excipients including, but not limited, to buffering agents, glidants, preservatives, and coloring agents. Additional excipients such as bulking agents, tonicity agents, and chelating agents are within the scope of the embodiments.

[00107] Non-limiting examples of buffering agents include, but are not limited to, sodium bicarbonate, potassium bicarbonate, magnesium hydroxide, magnesium lactate, magnesium glucomate, aluminum hydroxide, aluminum hydroxide/sodium bicarbonate co precipitate, a mixture of an amino acid and a buffer, a mixture of aluminum glycinate and a buffer, a mixture of an acid salt of an amino acid and a buffer, and a mixture of an alkali salt of an amino acid and a buffer. Additional buffering agents include sodium citrate, sodium tartarate, sodium acetate, sodium carbonate, sodium polyphosphate, potassium polyphosphate, sodium pyrophosphate, potassium pyrophosphate, disodium hydrogenphosphate, dipotassium hydrogenphosphate, trisodium phosphate, tripotassium phosphate, sodium acetate, potassium metaphosphate, magnesium oxide, magnesium hydroxide, magnesium carbonate, magnesium silicate, calcium acetate, calcium glycerophosphate, calcium chloride, calcium hydroxide, calcium lactate, calcium carbonate, calcium bicarbonate, and other calcium salts. [00108] In some embodiments, the tablet described herein comprises a preservative. Preservatives include anti-microbials, anti-oxidants, and agents that enhance sterility. Exemplary preservatives include ascorbic acid, ascorbyl palmitate, BHA, BHT, citric acid, erythorbic acid, fumaric acid, malic acid, propyl gallate, sodium ascorbate, sodium bisulfate, sodium metabisulfite, sodium sulfite, parabens (methyl-, ethyl-, butyl-), benzoic acid, potassium sorbate, vanillin, and the like.

[00109] In some embodiments, the tablet described herein comprises a coloring agent for identity and/or aesthetic purposes of the resultant liquid form. Suitable coloring agents illustratively include FD&C Red No. 3, FD&C Red No. 20, FD&C Red No. 40, FD&C Yellow No. 6, FD&C Blue No. 2, D&C Green No. 5, D&C Orange No. 5, caramel, ferric oxide and mixtures thereof.

[00110] Additional excipients are contemplated in the tablet embodiments. These additional excipients are selected based on function and compatibility with the tablet compositions described herein and may be found, for example in Remington: The Science andPractice of Pharmacy, Nineteenth Ed (Easton, PA: Mack Publishing Company, 1995); Hoover, John E., Remington ’s Pharmaceutical Sciences, (Easton, PA: Mack Publishing Co 1975); Liberman, H. A. and Lachman, L., Eds., Pharmaceutical Dosage Forms (New York, NY : Marcel Decker 1980); and Pharmaceutical Dosage Forms and Drug Delivery Systems, Seventh Ed (Lippincott Williams & Wilkins 1999), herein incorporated by reference in their entirety.

[00111] In further embodiments, the tablets described herein are coated tablets, such as enteric- coated tablets, sugar-coated, or film-coated tablets.

[00112] In one embodiment, the individual unit dosages also include film coatings, which disintegrate upon oral ingestion or upon contact with diluent. In one embodiment, these formulations are manufactured by conventional techniques.

[00113] Compressed tablets are solid dosage forms prepared by compacting the bulk blend formulations described above. In various embodiments, compressed tablets which are designed to dissolve in the mouth will include one or more flavoring agents. In other embodiments, the compressed tablets will include a film surrounding the final compressed tablet. In some embodiments, the film coating aids in patient compliance (e.g., Opadry® coatings or sugar coating). Film coatings comprising Opadry® typically range from about 1% to about 5% of the tablet weight. In other embodiments, the compressed tablets include one or more excipients.

[00114] Provided herein are film -coated tablets forms, which comprise: a combination of an active ingredient (e.g. Compound A-HC1) and one or more tabletting excipients to form a tablet core and subsequently coating the core. The tablet cores are produced using conventional tabletting processes and with subsequent compression and coating.

[00115] Enteric-coatings are coatings that resist the action of stomach acid but dissolve or disintegrate in the intestine.

[00116] In one aspect, the oral solid dosage form disclosed herein include an enteric coating(s). Enteric coatings include one or more of the following: cellulose acetate phthalate; methyl acrylate-methacrylic acid copolymers; cellulose acetate succinate; hydroxy propyl methyl cellulose phthalate; hydroxy propyl methyl cellulose acetate succinate (hypromellose acetate succinate); polyvinyl acetate phthalate (PVAP); methyl methacrylate-methacrylic acid copolymers; methacrylic acid copolymers, cellulose acetate (and its succinate and phthalate version); styrol maleic acid co-polymers; polymethacrylic acid/acrylic acid copolymer; hydroxy ethyl ethyl cellulose phthalate; hydroxypropyl methyl cellulose acetate succinate; cellulose acetate tetrahydrophtalate; acrylic resin; shellac.

[00117] An enteric coating is a coating put on a tablet, pill, capsule, pellet, bead, granule, particle, etc. so that it doesn’t dissolve until it reaches the small intestine.

[00118] Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation.

[00119] Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxy ethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets. In some embodiments, tablets are coated with water soluble, pH independent film coating which allows for immediate disintegration for fast, active release (e.g. Opadry products).

Dosage in the Tablet

[00120] In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is between about 5 mg and about 100 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is between about 5 mg and about 80 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is between about 5 mg and about 60 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is between about 10 mg and about 40 mg. [00121] In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 10 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 20 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 30 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 40 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 50 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 60 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 70 mg. In some embodiments, the amount of Compound A-HC1, or solvate thereof, in the tablet is about 80 mg.

Methods of Dosing and Treatment Regimens

[00122] In one embodiment, the pharmaceutical compositions disclosed herein are used as medicaments for the treatment of diseases or conditions in a mammal that would benefit from modulation of somatostatin activity. Methods for treating any of the diseases or conditions described herein in a mammal in need of such treatment, involves administration of pharmaceutical compositions that include Compound A, or a pharmaceutically acceptable salt thereof, in therapeutically effective amounts to said mammal.

[00123] In certain embodiments, the compositions containing Compound A described herein are administered for prophylactic and/or therapeutic treatments. In certain therapeutic applications, the compositions are administered to a patient already suffering from a disease or condition, in an amount sufficient to cure or at least partially arrest at least one of the symptoms of the disease or condition. Amounts effective for this use depend on the severity and course of the disease or condition, previous therapy, the patient's health status, weight, and response to the drugs, and the judgment of the treating physician. Therapeutically effective amounts are optionally determined by methods including, but not limited to, a dose escalation and/or dose ranging clinical trial.

[00124] In general, however, doses employed for adult human treatment are typically in the range of about 10 mg to about 100 mg per day of Compound A. In one embodiment, the desired dose is conveniently presented in a single dose or in divided doses administered simultaneously or at appropriate intervals, for example as two, three, four or more sub-doses per day.

[00125] In any of the aforementioned aspects are further embodiments comprising single administrations of the effective amount of the compound, including further embodiments in which the compound is administered once a day. EXAMPLES

[00126] The following examples are provided for illustrative purposes only and not to limit the scope of the claims provided herein.

Example 1: Oral Capsules

[00127] Representative capsules are described below in Table 1 .

Table 1. aAmount corrected for assay and moisture, chloride and isopropyl alcohol content; bCapsule fill weight adjusted based upon blend assay; cComposed of red iron oxide, titanium dioxide and gelatin.

[00128] A representative description of the manufacturing process for the hot melt granulation capsules is as follows:

[00129] Stage 1 : High Shear Wet Granulation: Melt the Vitamin E Polyethylene Glycol Succinate (TPGS). Compound A-HC1, mannitol, microcrystalline cellulose, croscarmellose sodium and silicon dioxide are charged into a high shear wet granulator and mixed. The melted Vitamin E TPGS is sprayed onto the granulation components.

[00130] Stage 2: Milling: The wet granulation is milled through a screening mill using an appropriately sized screen.

[00131] Stage 3 : Blending: The sodium stearyl fumarate is sieved using an appropriately sized screen. The milled granulation is charged into the diffusion mixer (tumble) along with the sodium stearyl fumarate and blended.

[00132] Stage d: Encapsulation: The 10 mg capsules were automatically encapsulated in Size 2 gelatin capsules. Example 2: Spray-Dried Solid Dispersions

[00133] Spray -dried solid dispersions were prepared with 15% by weight Compound A-HC1: 15/85 Compound A-HC1/HPMCAS-M and 15/85 Compound A-HC1/PVP VA64 formulations. The manufactures were completed using the BLD-150, the Bend Lab Dryer with 150 kg/hr drying gas capacity. The parameters varied were solution solids loading for the HPMCAS-M SDD formulation to help reduce nozzle bearding, and dryer outlet temperature for the PVP VA64 SDD to de-risk fluctuations that may occur during clinical manufacturing. The original process parameter screening plan specified manufacturing the decreased dryer outlet temperature condition with a larger orifice nozzle to produce larger particles, however based on the results of the first spray it was determined that the atomization pressure needed to achieve the desired solution flow rate would have been too low to fully atomize the solution with the larger orifice. As dryer outlet temperature tends to have more variability than solution flow rate, the parameter screen was shifted to focus on de-risking the dryer outlet temperature alone while ensuring fully atomized droplets. Dryer outlet temperature variation could affect residual solvent levels in the SDDs, which can affect physical and chemical stability. All sprays were completed successfully with good yields, and indicate a robust processing space for both formulations.

15/85 Compound A HPMCAS-M SDD Formulation Manufacturing Details

[00134] Three sub batches of 15/85 Compound A-HC1/HPMCAS-M SDDs were sprayed to explore the manufacture processing space and prepare for clinical trial manufacturing. A sub batch was sprayed first at 10 wt% solids, and significant nozzle bearding that appe ared to affect the spray plume was observed after about 45 minutes on solution.

[00135] The solution was diluted to 8 wt%, and a sub batch was manufactured with a duration of 1 hourto ensure bearding was reduced. A very small amount of bearding was observed during this batch after about 50 minutes on solution, but did not appear to affect the atomization plume and the 8 wt% solids loading was selected.

[00136] Cooling water at 2 GPM and approximately 7 °C was run through the spray dryer lid throughout all sprays to keep the lid cool and prevent sticking and browning. No significant lid buildup or browning was seen throughout manufacturing. No cleaning was performed between sprays, and all sprays were completed from one solution with additional solvent added prior to manufacturing batches 2 A and 2C. A summary of the manufacturing parameters used for all three sub batches is shown in Table 2. Table 2. Manufacturing summary for 15/85 Compound A-HC1/HPMCAS-M SDDs.

' Dry yield is calculated as SDD samples and dry bulk collected divided by the amount of solids sprayed.

15/85 Compound A-HC1/PVP VA64 SDD Formulation Manufacturing Details

[00137] Process parameter screening sprays and an FPN demonstration batch were also completed for the 15/85 Compound A-HC1/PVP VA64 SDD formulation. The dryer outlet temperature was varied to de-risk process parameter variability to prepare for clinical trial manufacturing.

[00138] The process space was constrained by a maximum dryer outlet temperature, the dryer outlet temperature, and the minimum desired solution flow rate. A maximum inlet temperature of 160 °C was specified to avoid sticking or browning of SDD on the spray dryer lid, and the minimum flow rate was set at 100 g/min to ensure sufficient throughput. The minimum and maximum dryer outlet temperatures were chosen to be 40 °C and 65 °C respectively to ensure adequate particle drying and that the dryer outlet temperature will not be above the wet particle Tg.

[00139] The process parameter screening sprays for the PVP VA64 formulation explored the manufacturing space by varying the dryer outlet temperature. This allowed investigation of the effect of dryer relative saturation on particle residual solvent content, morphology, density, and stability. [00140] Cooling water was run at 2 GPM and approximately 7 °C throughout all sprays to prevent lid buildup and browning, and neither were noted. No nozzle bearding was observed throughout all three manufactures. All sprays were completed from one solution. The manufacturing details of each sub batch are summarized in Table 3.

Table 3. 15/85 Compound A-HC1*/PVP VA64 SDD process parameters

* Compound A-HC1, formulated on a basis offreebase. xDry yield is calculated as SDD samples and dry bulk collected divided by the amount of solids sprayed.

SDD Characterization

[00141] Particle Properties: The particle size distribution and bulk and tapped densities were measured for each batch of Compound A-HC1 SDD. The HPMCAS-M SDDs have larger particle sizes, which is expected because the HPMCAS-M solution is more viscous than the PVP VA64 solution which leads to larger droplets for a given nozzle configuration. The increased solids loading in solution of lot 2B led to larger particles than batches 2 A and 2C, which is also due to the higher viscosity of the spray solution. The particle size distributions of all PVPVA-64 batches are similar, as expected.

[00142] The bulk and tapped densities of 2 A and 2C are similar, while batch 2B has a slightly lower density potentially due to the larger particles. The bulk and tapped densities of all PVPVA- 64 batches are similar, indicating a robust process with respect to the dryer outlet temperature effects on powder properties. [00143] Residual Solvent and Water Content: The residual methanol and water of the six SDDs were measured using GC and KF respectively. All SDDs contained residual methanol below the ICH guideline of 0.3 wt% after secondary drying, suggesting adequate drying at 40 °C/15%RH.

[00144] Morphology by SEM: The particle morphology of all six SDDs was evaluated via SEM. Each SDD showed typical morphology with no evidence of irregular particles suggesting adequate atomization for all conditions tested. The HPMCAS-M particles were primarily collapsed spheres while the PVP VA64 SDDs contain a larger fraction of spherical particles.

[00145] Crystallinity by PXRD: All six SDDs were evaluated for crystallinity using PXRD. All SDDs were amorphous by PXRD as evident in the absence of sharp diffraction peaks.

[00146] Thermal Characteristics by DSC: All six SDDs were characterized by modulatedDSC. The results are tabulated in Table 4. The manufactured SDDs were all amorphous and homogenous by DSC as evident by the presence of a single glass transition in the reversing heat signal. Neither formulation shows signs of crystallization after the Tg suggesting low propensity f or crystallization of Compound A at those temperatures for both formulations. Furthermore, both formulations showed high Tg relative to ambient temperatures suggesting low physical stability risks in dry conditions. Packaging to minimize humidity will be necessary for the PVP VA64 formulation.

Table 4. Tabulated thermal characteristics of the six batches as measured by DSC.

Summary

[00147] Physical Stability observations: PVP VA64 SDD appeared to deliquesce upon storage with crystals observed at 3 months (40 °C/75%RH open). Storage with desiccant is recommended. HPMCAS-M SDD was physically stable through 6 months at 40 °C/75%RH open.

[00148] Chemical Stability observations: Possible acid catalyzed degradation in the HPMCAS- M formulation on stability. Some degradation in the PVP VA64 formulation as well, but not as significant as the HPMCAS-M SDD. Packaging will be required for the PVPVA SDD which will be driven by physical stability concerns.

[00149] The 15% by weight Compound A/PVP VA64 was selected as the lead SDD formulation.

12-month stability: 15% Compound A-HC1/PVP-VA64 SDD

[00150] 12-month SDD samples were held at 5 °C, 25 °C/60%RH, and 40 °C/75% RHwith desiccant. Samples for water analysis by Karl Fisher titration were prepared and analyzed immediately; the remaining samples were vacuum desiccated overnight to remove residual moisture and preserve the physical state of SDD’ s for further characterization. A list of the analytical tests performed for characterization included: appearance, water content by Karl Fisher titration, Powder x-ray diffraction (PXRD), scanning electron microscopy (SEM), thermal characterization by modulated differential scanning calorimetry (mDSC), dissolution performance by micro-centrifuge (MCT) test, assay and related substances by HPLC.

[00151] Conclusion from PXRD analysis on the 12-month SDD stability samples: There was no evidence of crystallinity in samples stored at 12 months at each of the stability conditions.

[00152] Conclusion from SEM analysis on the 12-month SDD stability samples: No particle fusion was observed across all stability conditions at 12 month. There was no evidence of crystallinity in samples stored at 12 months at each of the stability conditions.

[00153] Conclusion from SEM analysis on the 12-month SDD stability samples: No particle fusion is observed across all stability conditions at 12 months. There was no evidence of crystallinity in samples stored at 12 months at each of the stability conditions.

[00154] Conclusions from mDSC analysis on the 12-month SDD stability samples: Repeated analysis of the 12 month SDD sample held at 5 °C affords non-reproducible thermograms, the cause for this result is not known at this time. The 5 °C sample was determined to be physically stable by all other characterization techniques. The 12 month SDD samples held at 25 °C/60%RH and 40 °C/75%RH showeda single, reproducible Tgat 124-125 °C, supporting the conclusion that the SDD is stable after 12 months of storage with desiccant at these conditions. [00155] Conclusion from MCT dissolution analysis on the 12-month SDD stability samples: Non-sink dissolution performance of the 12 month stability samples is consistent with the initial (to) sample stored at -20 °C.

35/65 Compound A-HC1/PVP VA64 SDD Formulation Manufacturing Details

[00156] Spray -dried solid dispersions were prepared with 35% by weight Compound A-HC1: 35/65 Compound A-HC1/PVP VA64 formulation. [00157] The manufacture of the SDD was completed using the SD-180 lab dryer. Secondary drying was completed using the Binder Convection Dryer. The manufacturing details are summarized in Table 5.

[00158] The spray was completed successfully with good yield.

Table 5. 35/65 Compound A-HC1*/PVP VA64 SDD process parameters

* Compound A-HC1, formulated on a basis offreebase

Example 3: Oral Tablets

[00159] Representative 10 mg, 20 mg, 30 mg, 40 mg, and 60 mg spray-dried dispersion tablets are presented in Tables 6, 7, 8, 9, 10, 11, 12, and 13.

[00160] Typical excipients used to prepare the tablets included: microcrystalline cellulose, mannitol, crospovidone, colloidal silicon dioxide, magnesium stearate, and Opadry White 03K184116 (film coating).

Table 6. Representative 10 mg spray-dried dispersion tablets.

aMethanol removed on drying during the spray drying process. bComposed of Hypromellose 2910, titanium dioxide and triacetin. cPurified water removed on drying during the film coating process.

Table 7. Representative 20 mg spray-dried dispersion (HPMCAS-M) tablets. Table 8. Representative 20 mg spray-dried dispersion (PVPVA 64) tablets.

Table 9. Additional representative 20 mg spray dried dispersion tablets.

Table 10. Representative 30 mg and 40 mg spray -dried dispersion (PVPVA 64) tablets. Table 11. Additional representative 40 mg spray-dried dispersion (PVPVA 64) tablets.

Table 12. Representative 60 mg spray-dried dispersion (PVPVA 64) tablets.

Table 13. Additional representative 60 mg spray-dried dispersion (PVPVA 64) tablets.

Table 14. Exemplary spray-dried dispersion tablets.

[00161] A representative non-limiting description of the manufacturing process for the SDD tablets is as follows:

[00162] Stage 1 : Spray Drying: Compound A-HC1 and copovidone are dissolved in MeOH. The solution spray-dried. Spray -dried dispersion (Compound A-HC1 SDD) is collected.

[00163] Stage 2: Roller Compaction: Granulation blend consisting of Compound A-HC1 SDD, filler(s), disintegrant(s), glidant(s), and lubricant(s) are blended. In some embodiments, granulation blend consisting of Compound A-HC1 SDD, mannitol, microcrystalline cellulose, crospovidone, colloidal silicon dioxide are prepared and blended. The intra-granular portion of the magnesium stearate is screened and added to the granulation blend. The resulting blend is blended. Granulation blend is charged into hopper of the roller compaction and compacted into ribbons. The ribbons are passed through mesh screen using in-line oscillating mill to break up the ribbons and mill into granules.

[00164] In some embodiments, the granulation blend comprises about 20% to about 35% (w/w of the final tablet weight) Compound A-HC1 SDD. In some embodiments, the granulation blend comprises about21%, about 22%, about28%, 29%, about33%, about 34% (w/w of the final tablet weight) Compound A-HC1 SDD. In some embodiments, the Compound A-HC1 SDD comprises a 15/85 Compound A-HC1 /HPMCAS-M, 15/85 Compound A-HC1 /PVPVA64, or 35/65 Compound A-HC1 /PVPVA 64 SDD.

[00165] Stage 3 : Blending: The intra-granular material is mixed with the extragranular excipients. Extragranular excipients comprise one or more excipients selected from: fillers, disintegrants, glidants, and lubricants. The extragranular components include microcrystalline cellulose, crospovidone, colloidal silicon dioxide. The extragranular lubricant, magnesium stearate, is sieved using an appropriately sized screen, then added to the blend and mixed.

[00166] Stage 4: Compression: The final blend is compressed into tablets.

[00167] Stage 5 : Pan-Coating: Film coating suspension of Opadry White 03K18416.is prepared in purified water, and tablets are coated with Opadry White 03K18416 in a perforated coating pan.

Example 4: Evaluation of Formulation Performance in Dogs

Study Designs

[00168] Evaluated two conditions* in the dog: +Pg pretreatment (mimics human fasted stomach, pH 1 -2) and -Pg pretreatment (mimics humans taking PPIs or antacids, pH 3-5). (* : 1 -week washout between each condition; Pg=pentagastrin.)

Compound A-HC1 Solution

[00169] N=4 non-naive dog. Vehicle: propylene glycol. Conditions: -Pg.

Compound A-HC1 HMG capsule

[00170] N=4 non-naive dog. Conditions: +Pg, -Pg.

Compound A-HC1 Spray-dried dispersion tablets: PVPVA

[00171] 2 groups of N=6 non-naive male dogs.. Conditions: +Pg, -Pg.

[00172] Results from this study is presented in Tables 15 and 16. Table 15. Dog PK evaluation of formulations of Compound A-HC1

[00173] As shown in Table 15 and Figure 2, the HMG capsule formulations performed poorly in dogs not pretreated with pentagastrin, whereas the spray-dried dispersionstablets performed better; For HMG capsule formulation, AUC without pentagastrin was only 11 % of the AUC with pentagastrin (98.2 ng*hr/mL compared to 917 ng*hr/mL). In comparison, the AUC for the PVPVA SDD tablet formulations without pentagastrin was 185% and 124% of with pentagastrin condition, respectively. These data show that PVPVA SDD tablet formulations are superior under high gastric pH environment (e.g., as would be in subjects that are are taking PPI or antacids).

Table 16. Dog PK evaluation of 60 mg 35/65 PVP-VA SDD tablet of Compound A-HC1

Example 5: A Phase 1, Multi-Cohort, Single Dose Study to Assess the Relative Bioavailability, Performance, and Safety of Two Formulations of Compound A [00174] The study was conducted in up to 3 cohorts, each with a specific primary objective: [00175] Cohort 1 : To characterize performance of 10 mg tablets prepared by spray-dried dispersion (SDD) of Compound A-HC1 salt.

[00176] Cohort 2 : To evaluate the relative bioavailability of 10 mg SDD tablets compared to the Compound A-HC1 hot melt granulation (HMG) formulation, 10 mg capsules. To determine the effect of timing of food administration on pharmacokinetics of low dose of the 10 mg SDD tablets.

[00177] Cohort 3 : To determine the effect of timing of food administration on pharmacokinetics of SDD tablets and dose proportionality in doses higher than 20 mg. To determine the optimal dosing regimen that results in adequate systemic exposure with short post-dose fasting duration. Study Design:

[00178] Up to thirty-six (36) healthy male and female subjects were enrolled. Cohorts 1 -2 consisted of four periods each, and Cohort 3 consisted of three periods.

Cohort 1:

[00179] The SDD tablets were evaluated. Up to twelve (12) healthy male and female subjects were enrolled in each cohort. Cohort 1 consisted of 4 periods: In Period 1, subjects were administered a proton-pump inhibitor (lansoprazole, 15 mg BID for 3 days (from Day -3), taken orally at least 30 min prior to a meal, once in the morning and once in the evening). On the f ourth day (Day 1 of study), fasted subjects will take the last dose of lansoprazole (15 mg) 60 min prior to 20 mg Compound A (2x10 mg of the SDD tablets). In Period 2, fasted subjects were administered 20 mg Compound A (2xl0 mg of the SDD tablets). In Period 3, fasted subjects were administered 20 mg Compound A (2x10 mg of the SDD tablets) with a high -fat, high- calorie meal. In Period 4, fasted subjects will take up to 80 mg Compound A (up to 8x10 mg of the SDD tablets). The actual dose was selected based on the pharmacokinetic data from Period 2. [00180] For Period 1 : In the eveningbefore dosing (Day -1), subjects were administered their evening dose of 15 mg lansoprazole, provided an evening meal at least 30 min after administration of lansoprazole, and then were required to fast overnight (> 10 hr) on Day -1 . On Day 1 , they were administered the morning dose (last dose) of 15 mg lansoprazole at least 60 min prior to administration of Compound A (2x10 mg SDD tablets). Subjects continued to fast for 2 hr after Compound A, after which they were allowed to ingest a standard meal.

[00181] For Period 2: Subjects were required to fast overnight (> 10 hr) on Day 7. On Day 8, 20 mg Compound A (2x10 mg SDD tablets) was administered orally. Subjects continued to fast for 2 hr after Compound A, after which they were allowed to ingest a standard meal.

[00182] For Period 3 : Subjects were required to fast overnight (> 10 hr) on Day 14. On Day 15, they were allowed to ingest a high-fat, high-calorie meal within 30 min. Upon completion of the ingestion of the meal, Compound A (2x10 mg SDD tablets) was administered (no more than 30 min after the start of the meal). No additional food was provided for at least 4 hr after administration of Compound A.

[00183] Subjects were not allowed to perform strenuous exercise of > 30 min/day 3 days prior to Day -1 and throughout the study.

[00184] PK and safety assessments including adverse event (AE) monitoring, clinical laboratory tests, vital sign measurements, 12-lead ECGs, Holter and telemetry monitoring (Period 4 only), and physical examinations were conducted at scheduled times throughout the study. Cohort l:

[00185] The cohort consisted of four periods. In each period, a single dose of 20 mg Compound A (2x10 mg SDD) was administered orally.

[00186] For Period 1 : Subjects were required to fast overnight (> 10 hr) on Day -1. On Day 1, a low-fat meal was given 2 hours after administration of 20 mg Compound A (2x 10 mg HMG capsules; reference formulation).

[00187] For Period 2: Subjects were required to fast overnight (> 10 hr) on Day 7. On Day 8, they were given a low-fat meal 2 hr after administration of 20 mg Compound A (2x10 mg SDD tablets; test formulation).

[00188] For Period 3 : Subjects were required to fast overnight (> 10 hr) on Day 14. On Day 15, they were given a low-fat meal 1 hr after administration of 20 mg Compound A (2x10 mg SDD tablets).

[00189] For Period 4: Subjects were required to fast overnight (> 10 hr) on Day 21. On Day 22, they were given a low-fat meal 0.5 hr after administration of 20 mg Compound A (2x10 mg SDD tablets).

[00190] The final study visit occured on Day 29. Subjects were not allowed to perform strenuous exercise of > 30 min/day, 3 days prior to Day -1 and throughout the study. PK and safety assessments including adverse event (AE) monitoring, clinical laboratory tests, vital sign measurements, 12-leadECGs, and physical examinations were conducted at scheduled times throughout the study.

Cohort 3:

[00191] The cohort consisted of three periods. In each period, a single dose of Compound A SDD (40, 60, or 80 mg) was administered orally (4x10 mg SDD tablets, 6x10 mg SDD tablets, or 8x1 Omg SDD tablets). There was a washout period of at least 10 days between each dose of Compound A.

[00192] For Period 1 : Subjects were required to fast overnight (> 10 hr) on Day -1. On Day 1, they were given a standard meal 1 hr after administration of 40 mg Compound A (4x10 mg SDD tablets).

[00193] For Period 2: Subjects were required to fast overnight (> 10 hr) on Day 10. On Day 11, they were given a standard meal 1 hr or 2 hr after administration of 80 Compound A (8x10 mg SDD tablets). The timing of the meal (1 hr or 2 hr post administration of Compound A) was dependent on the mean AUC 0 -24 determined in Period 1.

[00194] For Period 3 : Subjects were required to fast overnight (> 10 hr) on Day 20. On Day 21, they were given a standard meal 1 hr or 4 hr after administration of 60 or 80 mg Compound A (6x10 mg SDD tablets or 8x10 mg SDD tablets). The dose and timing of the standard meal was dependent on the mean AUC 0 -24 determined in Period 2.

[00195] The final study visit occured on Day 29. Subjects were not allowed to perform strenuous exercise of > 30 min/day, 3 days prior to Day -1 and throughout the study. PK and safety assessments including adverse event (AE) monitoring, clinical laboratory tests, vital sign measurements, 12-leadECGs, and physical examinations were conducted at scheduled times throughout the study.

Study Population:

[00196] Up to 36 healthy male or female subjects, between the ages of 18 to 55 years, inclusive, were enrolled. For Cohort 2 only, male and female subjects 18 to 65 years of age, inclusive, at the time of screening.

Inclusion Criteria

[00197] Each subject had to meet all of the following inclusion criteria to be enrolled in the study: Male and female subjects 18 to 55 years of age, inclusive, at the time of screening. For cohort 2 only, male and female subjects 18 to 65 years of age, inclusive, at the time of screening. Body mass index (BMI) of 18 to 30 kg/m 2 , inclusive. Willing to refrain from strenuous, unaccustomed exercise and sports, defined as greater than 30 minutes per day, 3 days priorto Day -1 and throughout the study. If the subject was a heterosexual orbisexual female, she had to be of non-childbearing potential OR must agree to use a highly effective or two clinically acceptable methods of contraception.

Exclusion Criteria

[00198] A healthy subject meeting any of the following criteria was to be excluded from the study: Prior treatment with Compound A. Any uncontrolled or activemajor systemic disease which makes study participation unsafe or could interfere with evaluation of the endpoints of the study. History or presence of malignancy except adequately treated basal cell or squamous cell carcinomas of the skin within the past 5 years. Active acute or chronic infection. Use of any investigational drug within the past 60 days or 5 half -lives, whichever is longer, priorto the first dosing of study drug. Use of tobacco and/or nicotine-containing products, recreational drugs, or alcohol for 48 hr prior to admission and agreement to refrain from use throughout the study. History of or current alcohol abuse and/or other drug addiction < 1 year priorto screening. Used any prescription or over-the-counter (OTC) medication or alternative medicinal products within 14 days ofDay -1. Use of caffeine-containing beverages or food for 48 hrpriorto Day -1 and for 48 hr prior to each check-in day for all subsequent periods. Have ingested foods containing poppy seeds within 7 days before screening until completion of the study assessments. Taking moderate or strong CYP3 A4 inhibitors or inducers. Strenuous exercise for > 30 min/day, 3 days prior to Day - 1 and throughoutthe study. Had a blood loss > 500 mL or donated blood within 3 months prior to admission. Have amylase and/or lipase levels >2xULN, alanine aminotransferase (ALT) and /or aspartate aminotransferase (AST) >2xULN, total bilirubin >1.5xULN (except in the case of known Gilbert’s syndrome), and/or serum creatinine above the upper limit of normal. History of hypersensitivity reactions to any excipients in the study drug. Tested positive at screening for human immunodeficiency virus (HIV), hepatitis B surface antigen (HBsAg), or hepatitis C antibody (HCV-Ab), or has a history of a positive result. Female subjects who have a positive serum pregnancy test or are breastfeeding. For Cohort 1 only, subjects that are classified as CYP2C19 poor metab olizers orultra-rapid metabolizers.

Test Product, Dose, and Mode of Administration:

[00199] 10 mg tablets (SDD). Multiple tablets were swallowed with water depending on dose specified for a given period/cohort.

Reference Therapy, Dose, and Mode of Administration:

[00200] 10 mg HMG capsule formulation served as the reference formulation. Multiple capsules were swallowed with water depending on dose specified for a given period/cohort.

Plasma Pharmacokinetic Parameters:

[00201] Blood PK samples were collected to evaluate Compound A plasma concentrations. [00202] PK parameters were calculated for Compound A and the following are shown in tables below: Area under the plasma concentration curve from 0 to 24 hours (AUC 0-24); Maximum plasma concentration (C max ); Time to achieve maximum plasma concentration (T max );

Results

[00203] Results from this clinical trial showed that the co-administration of proton pump inhibitors only had a small effect on the pharmacokinetics observed with the administration of the SDD tablets, shorter fasting times are realized with the SDD tablets and the SDD tablets provide better dose proportional pharmacokinetics.

[00204] Results from Cohort 1 is presented in Table 17.

Table 17. Results from Cohort 1

Median and range are reported for T ma x. Mean and range are reported for P4/P2 ratios. All dose administered with overnight fast and 2h post dose fast. Food: high fat meal.

[00205] Cohort 1 (SDD 10 mg x2 under different conditions): the observed exposures with and without PPI are fairly comparable. Cohort 1 (SDD 10 mgx 2 vs. 10 mg x 6): Relatively dose proportional increases in exposures were observed.

[00206] In comparison to the SDD tablets, relatively dose proportional increases in exposures were not observed with the HMG capsules. See Figure 1 . Dose proportionality data for the HMG capsules obtained from a previous clinical study is presentedin Table 18.

Table 18. Comparative data: Dose Proportionality Observed for HMG Capsule Formulations after single dose (4 h post dose fast).

Data shown are mean ± standard deviation

[00207] Results from Cohort 2 is presented in Table 19. Table 19. Results from Cohort 2

All dose administered with overnight fast.

[00208] Cohort 2 (SDD 10 mg x2 vs. HMG and under different post-dose fasting durations): SDD tablets did not appear to have better exposures than HMG capsules and the two formulations were relatively comparable. For the SDD tablets the AUC 0 .24 (a measure of extent of absorption) after a 1 hr post dose fast was decreased to 82% of that obsereved with 2 hr postdose fast, which is a relatively small decrease in exposure.

[00209] In comparison to the performance of the SDD tablets under different post-dose fasting duration scenarios, the HMG capsules performed poorly under different post-dose fasting duration scenarios in a previously completed clinical study. Pharmacokinetic data obtained after administering a 20 mg dose (10 mg HMG capsule x2) in 12 subjects (N=4 male, N=8 Female) is presented in Table 20. Table 20. Comparative Data: HMG capsule Performance Under Different Post-Dose Fasting Durations

[00210] With the HMG capsule formulations, approximatley 30% loss in extent of absorption was noted with a 1 -hour post-dose fasting vs. a 2-hour post-dose fast.

[00211] HMG capsules with a 2h fast was evaluated in Phase 2 clinical studies. A 1 hour fast is more desirable than a 2 hour fast. Only 18% loss of AUC (0 -24) was observed with 1 hour fast compared to a 2 hour fast. SDD Ih fast will be utilized for Phase 3. Importantly, SDD tablets appeared to have better dose proportionality than HMG capsules, allowing for 3 ,0x dose (i.e., 60 mg) to be administered in Phase 3 clinical studies.

[00212] The examples and embodiments described herein are for illustrative purposes only and various modifications or changes suggested to persons skilled in the art are to be included within the spirit and purview of this application and scope of the appended claims.