Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
AMORPHOUS FORM OF 5-BROMOPYRIDIN-3-YL 3-DEOXY-3-[4-(3,4,5-TRIFLUOROPHENYL)-1H-1,2,3-TRIAZOL-1-YL]-1-THIO-ALPHA-D-GALACTOPYRANOSIDE
Document Type and Number:
WIPO Patent Application WO/2020/127461
Kind Code:
A1
Abstract:
The present invention relates to a stabilized amorphous form of a compound of formula (I) as well as compositions for oral administration comprising the compound of formula (I) in a therapeutically effective amount.

Inventors:
GRAVELLE LISE (DK)
TIMMINS PETER (GB)
PEDERSEN ANDERS (DK)
Application Number:
PCT/EP2019/085912
Publication Date:
June 25, 2020
Filing Date:
December 18, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GALECTO BIOTECH AB (DK)
International Classes:
C07H19/056; A61K9/10; A61K31/7056; A61P11/00
Domestic Patent References:
WO2016120403A12016-08-04
WO2016120403A12016-08-04
Other References:
GUY VAN DEN MOOTER: "The use of amorphous solid dispersions: A formulation strategy to overcome poor solubility and dissolution rate", DRUG DISCOVERY TODAY: TECHNOLOGIES, vol. 9, no. 2, 1 June 2012 (2012-06-01), AMSTERDAM, NL, pages e79 - e85, XP055317903, ISSN: 1740-6749, DOI: 10.1016/j.ddtec.2011.10.002
TU VAN DUONG ET AL: "The role of the carrier in the formulation of pharmaceutical solid dispersions. Part II: amorphous carriers", EXPERT OPINION ON DRUG DELIVERY, 17 June 2016 (2016-06-17), GB, pages 1 - 14, XP055283127, ISSN: 1742-5247, DOI: 10.1080/17425247.2016.1198769
TEJA S B ET AL: "Drug-excipient behavior in polymeric amorphous solid dispersions", JOURNAL OF EXCIPIENTS AND FOOD CHEMICALS, INTERNATIONAL PHARMACEUTICAL EXCIPIENTS COUNCIL, AMERICAS, UNITED STATES, vol. 4, no. 3, 1 September 2013 (2013-09-01), pages 70 - 94, XP009176942, ISSN: 2150-2668
NEMANJA KOLASINAC ET AL: "Spray coating as a powerful technique in preparation of solid dispersions with enhanced desloratadine dissolution rate", DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, vol. 39, no. 7, 5 July 2013 (2013-07-05), US, pages 1020 - 1027, XP055653119, ISSN: 0363-9045, DOI: 10.3109/03639045.2012.694890
PAUDEL AMRIT ET AL: "Manufacturing of solid dispersions of poorly water soluble drugs by spray drying: Formulation and process considerations", INTERNATIONAL JOURNAL OF PHARMACEUTICS, ELSEVIER, NL, vol. 453, no. 1, 20 July 2012 (2012-07-20), pages 253 - 284, XP028675204, ISSN: 0378-5173, DOI: 10.1016/J.IJPHARM.2012.07.015
MELERO ET AL., NATURE REVIEWS CANCER, 2015
Attorney, Agent or Firm:
NEX & PHISTER APS (DK)
Download PDF:
Claims:
WE CLAIM:

1. An amorphous form of a compound of formula I

2. A pharmaceutical composition comprising the amorphous form of claim 1 , and op tionally a pharmaceutically acceptable additive.

3. A liquid composition comprising a mixture of a dissolved or dispersed polymer for stabilizing amorphous form of the compound of formula I and a solution of a compound of for mula I

and optionally a solution of a pharmaceutically acceptable additive.

4. An amorphous solid dispersion composition comprising a mixture of an amorphous compound of formula I and a polymer for stabilizing the amorphous form of the compound of formula I, and optionally a pharmaceutically acceptable additive.

5. The composition of claim 4 wherein the mixture comprises a weight ratio of the amorphous compound of formula I to the polymer in the range from 1 :0.5 to 1 :5.

6. The composition of claim 4 or 5 wherein the amorphous compound of formula I is present in a concentration from 0.5 %w/w to 90 %w/w and the polymer is present in a concen tration of at least 0.1 %w/w, and optionally a pharmaceutically acceptable additive adding up to 100 %w/w.

7. A tablet composition for oral administration comprising the amorphous solid disper sion composition of any one of claims 4-6.

8. A drug layered composition comprising a) an inert solid core having an outer surface and b) a mixture of amorphous compound of formula I, a polymer for stabilizing the amor phous form of the compound of formula I and optionally a pharmaceutically acceptable addi tive; wherein the mixture is a layer on the outer surface of the inert solid core.

9. The drug layered composition of claim 8 wherein the mixture dissolved in a suitable solvent is sprayed onto the surface of the inert solid core.

10. The drug layered composition of claim 8 or 9 wherein the mixture comprises a weight ratio of the amorphous compound of formula I to the polymer in the range from 1 :0.5 to 1 :5.

11. The drug layered composition of any one of claims 8-10 wherein the amorphous compound of formula I is present in a concentration from 0.5 %w/w to 20 %w/w, the polymer is present in a concentration from 1 %w/w to 40 %w/w, the inert solid core is present in a con centration of 40 %w/w to 98.5 %w/w, and optionally a pharmaceutically acceptable additive adding up to 100 %w/w.

12. A capsule composition for oral administration comprising the drug layered composi tion of any one of claims 8-11.

13. The composition of any one of claims 3-12 wherein the polymer for stabilizing the amorphous form of the compound of formula I is one or more selected from the group consist ing of polyethylene glycol (PEG), carboxymethylethylcellulose (CMEC), ethyl cellulose, hy- droxyethylcellulose (HEC), methyl cellulose (MC), polyvinylpyrrolidone vinyl acetate (PVP/VA), Polyvinylpyrrolidone, hydroxy propyl methyl cellulose (Hypromellose), Hypro- mellose phthalate, Hypromellose acetate succinate, Eudragit, and Gelucire 44/14, preferably a hydroxy propyl methyl cellulose.

14. The composition of any one of claims 8-13 wherein the pharmaceutically acceptable additive is present and comprises a pore forming excipient.

15. The composition of claim 14 wherein the pore forming excipient is present in a concentration up to 20 %w/w in the layer.

16. The composition of claim 14 or 15 wherein the pore forming excipient is selected from the group consisting of Calcium Dihydrogen Phosphate, Lactose, Hydroxypropyl cellu lose and microcrystalline cellulose.

17. The composition of any one of claims 8-16 wherein the inert solid core is made of a material selected from the group consisting of sugars, such as starch and sucrose, and micro crystalline cellulose (MCC).

18. The composition of any one of claims 8-17 wherein the inert solid core has a size in the range from 50 to 2000 pm.

19. The composition of any one of claims 2-18 comprising the amorphous compound of formula I in an amount of 0.1 mg to lOOOmg, such as 5mg to 900mg, 10 to 800mg, 20 to700mg, 30 to 600mg, 40 to 500mg, 50 to 400mg, 60 to 300mg, 70 to 200mg, 80 to 150mg, or 90 to lOOmg, typically in an amount of 5 to 200mg, preferably in an amount of 10 to lOOmg.

20. A method of preparing the liquid composition of claim 3 comprising the steps of: a) preparing a solution or dispersion of the polymer and dissolving the compound of formula I in the solution or dispersion, and optionally dissolving a pharmaceutically acceptable additive in the solution or dispersion, thereby providing the liquid composition; or

b) preparing a first solution or dispersion of the polymer and a second solution of the com pound of formula I, and mixing the first and second solution, and optionally dissolving a phar maceutically acceptable additive in the first or second solution or dispersion, or the mixture, thereby providing the liquid composition.

21. A method of preparing the amorphous solid dispersion composition of claim 4 com prising the steps of:

spray drying the liquid composition of claim 3 and collecting the amorphous solid dispersion composition.

22. A method of preparing the drug layered composition of claim 8 comprising the steps of:

spraying the liquid composition of claim 3 onto an inert solid core and collecting the drug lay ered composition.

23. The method of claim 22 wherein the spraying is spray-coating or fluid bed coating.

24. The composition of any one of claims 4-19 for use in a method for treatment of a disease or disorder selected from the group consisting of inflammation; fibrosis, such as pulmo nary fibrosis, liver fibrosis, kidney fibrosis, ophthalmological fibrosis and fibrosis of the skin and heart; scarring; keloid formation; aberrant scar formation; surgical adhesions; septic shock; cancer, such as carcinomas, sarcomas, leukemias and lymphomas, such as T-cell lymphomas; metastasising cancers; autoimmune diseases, such as psoriasis, rheumatoid arthritis, Crohn’s disease, ulcerative colitis, ankylosing spondylitis, systemic lupus erythematosus; metabolic dis orders; heart disease; heart failure; pathological angiogenesis, such as ocular angiogenesis or a disease or condition associated with ocular angiogenesis, e.g. neovascularization related to can cer; and eye diseases, such as age-related macular degeneration and corneal neovascularization; atherosclerosis; metabolic diseases such as diabetes; type 2 diabetes; insulin resistens; obesity; Diastolic HF; asthma and other interstitial lung diseases, including Hermansky-Pudlak syn drome, mesothelioma; liver disorders, such as non-alcoholic steatohepatitis or non-alcoholic fatty liver disease, in a mammal, such as a human.

25. The composition of claim 24 wherein the composition comprising the compound of formula I is administered once daily or twice daily.

26. A method for treatment of a disease or disorder selected from the group consisting of inflammation; fibrosis, such as pulmonary fibrosis, liver fibrosis, kidney fibrosis, ophthal mological fibrosis and fibrosis of the skin and heart; scarring; keloid formation; aberrant scar formation; surgical adhesions; septic shock; cancer, such as carcinomas, sarcomas, leukemias and lymphomas, such as T-cell lymphomas; metastasising cancers; autoimmune diseases, such as psoriasis, rheumatoid arthritis, Crohn’s disease, ulcerative colitis, ankylosing spondylitis, systemic lupus erythematosus; metabolic disorders; heart disease; heart failure; pathological an giogenesis, such as ocular angiogenesis or a disease or condition associated with ocular angiogenesis, e.g. neovascularization related to cancer; and eye diseases, such as age-related macular degeneration and corneal neovascularization; atherosclerosis; metabolic diseases such as diabetes; type 2 diabetes; insulin resistens; obesity; Diastolic HF; asthma and other intersti tial lung diseases, including Hermansky-Pudlak syndrome, mesothelioma; liver disorders, such as non-alcoholic steatohepatitis or non-alcoholic fatty liver disease, in a mammal, comprising administering a therapeutically effective amount of the composition of any one of claims 4-19.

27. The method of claim 26 wherein the composition comprising the compound of for mula I is administered once daily or twice daily.

Description:

Technical field

The present invention relates to an amorphous form of a compound of formula I. The amorphous form of the compound of formula I can be stabilized with polymers when sprayed onto inert particles as well as when prepared as an amorphous solid dispersion.

Background Art

The compound of formula I has been described in international patent application publi cation number W02016120403 as a galectin 3 inhibitor useful for treating various disorders or diseases, as described therein. The compound of formula I is highly crystalline but has rela tively low solubility making it difficult to prepare a suitable formulation for toxicology and clinical studies.

Summary of the invention

The amorphous form approaches described in the present description enable the prepa ration of convenient dosage forms for the treatment of mammalian, such as human disease, not possible without the invention. Thus, all herein described diseases, disorders and/or conditions can be treated with the formulations of the present invention.

Material characterization of the compound of formula I shows that it is highly crystalline by XRPD. A high melting point of 233.7°C suggests that it forms a very stable crystal structure. Thermogravimetric analysis (TGA) data shows no indication of hydration and Dynamic vapor sorption (DVS) shows that the compound of formula I is not hygroscopic and shows no indica tion of hydrate formation.

Excipients to enhance the solubility of the compound of formula I were required, and several iterations of solution formulations were developed for toxicology studies as issues were found with limiting exposure and poor tolerability in earlier solution formulations.

Two different amorphous formulations, both containing amorphous compound of for mula I, were found and investigated, that is 1) an amorphous spray dried dispersion (ASD) and 2) a structured drug composition prepared by spray coating/drug layering the amorphous com pound of formula I onto inert microspheres.

One ASD formulation was found to be stable for 4 weeks at 25°C/60% RH and 40°C/75% RH. One drug layered formulation of the compound of formula I coated onto inert microspheres was achieved and was found to be stable for 12 months at 25°C/60%RH and 6 months at 40°C/75%.

In a broad aspect, the present invention relates to an amorphous form of a compound of formula I

In another aspect, the present invention concerns a pharmaceutical composition com- prising the amorphous form of the compound of formula I, and optionally a pharmaceutically acceptable additive.

In a further aspect, the present invention concerns a liquid composition comprising a mixture of a dissolved or dispersed polymer for stabilizing the amorphous form of the com- pound of formula I and a solution of a compound of formula I, and optionally a solution of a pharmaceutically acceptable additive.

In a still further aspect, the present invention relates to an amorphous solid dispersion composition comprising a mixture of an amorphous compound of formula I and a polymer for stabilizing the amorphous form of the compound of formula I, and optionally a pharmaceuti- cally acceptable additive. In one embodiment the mixture comprises a weight ratio of the amor phous compound of formula I to the polymer in the range from 1 : 0.5 to 1 : 5, such as from 1 : 1 to 1 :3, e.g. 1 :2. In a further embodiment the amorphous compound of formula I is present in a concentration from 0.5 %w/w to 90 %w/w and the polymer is present in a concentration of at least 0.1 %w/w, and optionally a pharmaceutically acceptable additive adding up to 100 %w/w.

In a further aspect, the present invention relates to a tablet or capsule composition for oral administration comprising the amorphous solid dispersion composition of the present in vention. Typically, the present invention relates to a tablet composition. In a particular embodi ment the oral composition, such as a tablet or capsule, is administered once daily. In another embodiment the oral composition, such as a tablet or capsule, is administered twice daily.

In a still further aspect, the present invention relates to a drug layered composition com prising a) an inert solid core having an outer surface and b) a mixture of amorphous compound of formula I, a polymer for stabilizing the amorphous form of the compound of formula I and optionally a pharmaceutically acceptable additive, wherein the mixture is a layer on the outer surface of the inert solid core. Typically, the mixture is deposited on to the surface of the inert solid core by spraying a solution of the mixture in a suitable solvent. In another embodiment the mixture comprises a weight ratio of the amorphous compound of formula I to the polymer in the range from 1 :0.5 to 1 :5, such as from 1 : 1 to 1 :3, e.g. 1 :2. In a further embodiment the amorphous compound of formula I is present in a concentration from 0.5 %w/w to 20 %w/w, the polymer is present in a concentration from 1 %w/w to 40 %w/w, the inert solid core is pre sent in a concentration of 40 %w/w to 98.5 %w/w, and optionally a pharmaceutically accepta ble additive adding up to 100 %w/w.

In a further aspect the present invention relates to a capsule composition for oral admin istration, such as once daily or twice daily, comprising the drug layered composition of the pre sent invention. It was found that the drug layered composition of the present invention would readily be enclosed in a pharmaceutical hard shell capsule either manually or by suitable manu facturing equipment and will allow tailoring of the dose included in the capsule.

In the above aspects and embodiments, where the polymer for stabilizing the amorphous form of the compound of formula I is part of the composition, such polymer is selected from the group consisting of one or more polyethylene glycol (PEG), carboxymethylethylcellulose (CMEC), ethyl cellulose, hydroxyethylcellulose (HEC), methyl cellulose (MC), polyvinylpyr rolidone vinyl acetate (PVPVA), Polyvinylpyrrolidone (PVP), such as (PVP K30), crospovidone, hydroxy propyl methyl cellulose (Hypromellose), e.g. Hypromellose 603 (HPMC 603), Hypromellose 606 (HPMC 606), Hypromellose 615 (HPMC 615), Hypromellose phthalate -55 (HP-55), and Hypromellose acetate succinate -LF (HPMCAS-LF), Eudragit L100-55, poloxamer, and Gelucire 44/14, preferably a hydroxy propyl methyl cellulose, such as HPMC 603.

In a further embodiment the pharmaceutically acceptable additive is present. When a layer is applied in the drug layered composition of the present invention the additive may com prise a pore forming excipient. When the pore forming excipient is present it is typically pre sent in a concentration up to 20 %w/w in the layer. The pore forming excipients as used herein are known to the person skilled in the art and is in one embodiment selected from the group consisting of Calcium Dihydrogen Phosphate, Lactose, Hydroxypropyl cellulose and micro crystalline cellulose also including mixtures hereof.

In the above aspects and embodiments, where the inert solid core is part of the composi tion, the inert solid core is made of a material selected from the group consisting of sugars, such as starch and sucrose, and microcrystalline cellulose (MCC), preferably MCC.

In a further embodiment the inert solid core has a size in the range from 50 to 2000 pm, such as 100 to 1400 pm.

In a further aspect the present invention relates to a method of preparing a liquid com position comprising a mixture of a dissolved or dispersed polymer for stabilizing the amor phous form of the compound of formula I and a solution of a compound of formula I, and optionally a solution of a pharmaceutically acceptable additive, comprising the steps of:

a) preparing a solution or dispersion of the polymer and dissolving the compound of formula I in the solution or dispersion, and optionally dissolving a pharmaceutically acceptable additive in the solution or dispersion, thereby providing the liquid composition; or

b) preparing a first solution or dispersion of the polymer and a second solution of the com pound of formula I, and mixing the first and second solution, and optionally dissolving a phar maceutically acceptable additive in the first or second solution or dispersion, or the mixture, thereby providing the liquid composition. In a still further aspect the present invention relates to a method of preparing an amor phous solid dispersion composition comprising a mixture of the amorphous compound of for mula I and a polymer for stabilizing the amorphous form of the compound of formula I, and op tionally a pharmaceutically acceptable additive, comprising the steps of:

spray drying a liquid composition comprising a mixture of a dissolved or dispersed polymer for stabilizing the amorphous form of the compound of formula I and a solution of a compound of formula I, and optionally a solution of a pharmaceutically acceptable additive; and collecting the amorphous solid dispersion composition.

As used herein and throughout the description and claims‘a solid dispersion’ means a dry composition, such as a dry powder.

In a further aspect the present invention relates to a method of preparing a drug layered composition comprising a) an inert solid core having an outer surface and b) a mixture of the amorphous compound of formula I, a polymer for stabilizing the amorphous form of the com pound of formula I and optionally a pharmaceutically acceptable additive, wherein the mixture is applied as a layer on the outer surface of the inert solid core, comprising the steps of:

spraying a liquid composition comprising a mixture of a dissolved or dispersed polymer for sta bilizing the amorphous form of the compound of formula I and a solution of a compound of formula I, and optionally a solution of a pharmaceutically acceptable additive, onto an inert solid core and collecting the drug layered composition. In one embodiment the spraying is spray-coating or fluid bed coating.

In a further aspect the present invention concerns an oral composition comprising a compound of formula I

and optionally a pharmaceutically acceptable additive, wherein the compound of formula I is present in an amount of 0.1 to lOOOmg, such as 1 to lOOOmg. Preferably, the composition is a solid oral composition, such as a tablet or capsule. Typically, one tablet or one capsule is one unit dosage form which then contains the amount of the compound of formula I. However, for treatment one or more unit dosages may be administered such as 1 to 4 unit dosages, for in stance a capsule containing 5 mg of compound of formula I, or 4 capsules each containing 5mg adding up to 20 mg dosage administered.

In an embodiment the oral composition is a unit dosage form.

In a further embodiment the compound of formula I is present in an amount of 5 to 900mg, such as 10 to 800mg, 20 to 700mg, 30 to 600mg, 40 to 500mg, 50 to 400mg, 60 to 300mg, 70 to 200mg, 80 to lOOmg. in another embodiment the compound of formula I is pre sent in an amount of 0.1 mg to lOOOmg, such as 0.1 to lmg, e.g. 0.2 to 0.8 mg. One example is administration of 5mg compound of formula I, another example is administration of 20 mg compound of formula I, a further example is administration of 50 mg of compound of formula I, and a still further example is administration of 200mg of compound of formula I. Each unit dose may contain from lmg to 200mg, for instance administration of 20 mg may be carried out by giving the human subjects 4 capsules each containing 5 mg compound of formula I.

In a still further embodiment, the pharmaceutically acceptable additive is present in a concentration from 0.1 to 99.5 % w/w, such as 10 %w/w to 99.5 %w/w and the compound of formula I is present in a concentration from 0.5 %w/w to 90 %w/w.

In a further embodiment the pharmaceutically acceptable additive is one or more se lected from the group consisting of polyethylene glycol (PEG), carboxymethylethylcellulose (CMEC), ethyl cellulose, hydroxyethylcellulose (HEC), methyl cellulose (MC), polyvinylpyr rolidone vinyl acetate (PVP/VA), Polyvinylpyrrolidone, hydroxy propyl methyl cellulose (Hypromellose), Hypromellose phthalate, Hypromellose acetate succinate, Eudragit, Gelucire 44/14, sugars, such as starch and sucrose, microcry stalline cellulose (MCC) and a pore forming excipient, such as Calcium Dihydrogen Phosphate, Lactose, Hydroxypropyl cellulose and mi- crocrystalline cellulose as well as mixtures hereof. In a further aspect the present invention relates to a composition of the present invention and any one of the above defined embodiments for use in a method for treatment of a disease or disorder selected from the group consisting of inflammation; fibrosis, such as pulmonary fibro sis, liver fibrosis, kidney fibrosis, ophthalmological fibrosis and fibrosis of the skin and heart; scarring; keloid formation; aberrant scar formation; surgical adhesions; septic shock; cancer, such as carcinomas, sarcomas, leukemias and lymphomas, such as T-cell lymphomas; metasta- sising cancers; autoimmune diseases, such as psoriasis, rheumatoid arthritis, Crohn’s disease, ulcerative colitis, ankylosing spondylitis, systemic lupus erythematosus; metabolic disorders; heart disease; heart failure; pathological angiogenesis, such as ocular angiogenesis or a disease or condition associated with ocular angiogenesis, e.g. neovascularization related to cancer; and eye diseases, such as age-related macular degeneration and corneal neovascularization; athero sclerosis; metabolic diseases such as diabetes; type 2 diabetes; insulin resistance; obesity; Dias tolic HF; asthma and other interstitial lung diseases, including Hermansky-Pudlak syndrome, mesothelioma; liver disorders, such as non-alcoholic steatohepatitis or non-alcoholic fatty liver disease, in a mammal, such as a human. In one embodiment the disease or disorder is NASH.

In another embodiment the disease or disorder is a cancer. In a further embodiment the disease or disorder is kidney fibrosis. In an embodiment the oral composition is administered once daily, such as one to four unit dosages once daily. In another embodiment the oral composition is administered twice daily, such as one to four unit dosages twice daily. In particular, the once daily amount is from 5mg to 900mg. In another embodiment the once daily amount is from 0.1 to 5 mg, such as 0.1 to 1 mg. In particular, the twice daily amount is from lmg to 500mg, such as 5mg to 200 mg.

In a still further aspect the present invention relates to a method for treatment of a dis ease or disorder selected from the group consisting of inflammation; fibrosis, such as pulmo nary fibrosis, liver fibrosis, kidney fibrosis, ophthalmological fibrosis and fibrosis of the skin and heart; scarring; keloid formation; aberrant scar formation; surgical adhesions; septic shock; cancer, such as carcinomas, sarcomas, leukemias and lymphomas, such as T-cell lymphomas; metastasising cancers; autoimmune diseases, such as psoriasis, rheumatoid arthritis, Crohn’s disease, ulcerative colitis, ankylosing spondylitis, systemic lupus erythematosus; metabolic disorders; heart disease; heart failure; pathological angiogenesis, such as ocular angiogenesis or a disease or condition associated with ocular angiogenesis, e.g. neovascularization related to cancer; and eye diseases, such as age-related macular degeneration and corneal neovasculariza tion; atherosclerosis; metabolic diseases such as diabetes; type 2 diabetes; insulin resistance; obesity; Diastolic HF; asthma and other interstitial lung diseases, including Hermansky-Pudlak syndrome, mesothelioma; liver disorders, such as non-alcoholic steatohepatitis or non-alcoholic fatty liver disease, in a mammal, comprising administering a therapeutically effective amount of the composition of the present invention, such as the amorphous solid dispersion composi tion or the drug layered composition, and any one of the above defined embodiments. In a fur ther embodiment the oral composition is administered once daily or twice daily, such as one to four unit dosages once daily or twice daily. In particular, the once daily amount is from 5mg to 900mg. In particular, the twice daily amount is from lmg to 500mg, such as 5mg to 200 mg.

In an embodiment of the method for treatment the composition comprises the compound of formula I in an amount of 5 to 900mg, such as 10-800mg, 20-700mg, 30 to 600mg, 40 to 500mg, 50 to 400mg, 60 to 300mg, 70 to 200mg. In a further embodiment 1 to 4 unit dosages of compound of formula I is administered, such as 1 to 4 capsules or tablets each containing from 5 to 200mg compound of formula I, such as 10 to lOOmg compound of formula I.

Another aspect of the present invention concerns combination therapy involving admin istering a compound of formula (I) of the present invention together with a therapeutically ac tive compound different from the compound of formula (I) (interchangeable with“a different therapeutically active compound”). In one embodiment the present invention relates to a com bination of a compound of formula (I) and a different therapeutically active compound for use in treatment of a disorder relating to the binding of a galectin-3 to a ligand in a mammal. Such disorders are disclosed below.

In an embodiment of the present invention, a therapeutically effective amount of at least one compound of formula (I) of the present invention is administered to a mammal in need thereof in combination with a different therapeutically active compound. In a further embodi ment, said combination of a compound of formula (I) together with a different therapeutically active compound is administered to a mammal suffering from a disorder selected from the group consisting of inflammation; fibrosis, such as pulmonary fibrosis, liver fibrosis, kidney fibrosis, ophthalmological fibrosis and fibrosis of the skin and heart; scarring; keloid formation; aberrant scar formation; surgical adhesions; septic shock; cancer, such as carcinomas, sarco mas, leukemias and lymphomas, such as T-cell lymphomas; metastasising cancers; autoim mune diseases, such as psoriasis, rheumatoid arthritis, Crohn’s disease, ulcerative colitis, anky losing spondylitis, systemic lupus erythematosus; metabolic disorders; heart disease; heart fail ure; pathological angiogenesis, such as ocular angiogenesis or a disease or condition associated with ocular angiogenesis, e.g. neovascularization related to cancer; and eye diseases, such as age-related macular degeneration and corneal neovascularization; atherosclerosis; metabolic diseases such as diabetes; type 2 diabetes; insulin resistance; obesity; Diastolic HF; asthma and other interstitial lung diseases, including Hermansky-Pudlak syndrome, mesothelioma; liver disorders, such as non-alcoholic steatohepatitis or non-alcoholic fatty liver disease.

A non-limiting group of cancers given as examples of cancers that may be treated, man aged and/or prevented by administration of a compound of formula (I) in combination with a different therapeutically active compound is selected from: colon carcinoma, breast cancer, pancreatic cancer, ovarian cancer, prostate cancer, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lym- phangeosarcoma, lymphangeoendothelia sarcoma, synovioma, mesothelioma, Ewing's sar coma, leiomyosarcoma, rhabdomyosarcoma, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, pa pillary adenocarcinomas, cystandeocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilms' tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung car cinoma, bladder carcinoma, epithelial carcinoma, glioblastomas, neuronomas, craniopharingio- mas, schwannomas, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroama, oligodendroglioma, meningioma, mela noma, neuroblastoma, retinoblastoma, leukemias and lymphomas, acute lymphocytic leukemia and acute myelocytic polycythemia vera, multiple myeloma, Waldenstrom's macroglobuline- mia, and heavy chain disease, acute nonlymphocytic leukemias, chronic lymphocytic leukemia, chronic myelogenous leukemia, Hodgkin's Disease, non-Hodgkin's lymphomas, rectum cancer, urinary cancers, uterine cancers, oral cancers, skin cancers, stomach cancer, brain tumors, liver cancer, laryngeal cancer, esophageal cancer, mammary tumors, childhood-null acute lymphoid leukemia (ALL), thymic ALL, B-cell ALL, acute myeloid leukemia, myelomonocytoid leuke mia, acute megakaryocytoid leukemia, Burkitt's lymphoma, acute myeloid leukemia, chronic myeloid leukemia, and T cell leukemia, small and large non-small cell lung carcinoma, acute granulocytic leukemia, germ cell tumors, endometrial cancer, gastric cancer, cancer of the head and neck, chronic lymphoid leukemia, hairy cell leukemia and thyroid cancer.

In some aspects of the present invention, the administration of at least one compound of formula (I) of the present invention and at least one additional therapeutic agent demonstrates therapeutic synergy. In some aspects of the methods of the present invention, a measurement of response to treatment observed after administering both at least one compound of formula (I) of the present invention and the additional therapeutic agent is improved over the same measure ment of response to treatment observed after administering either the at least one compound of formula (I) of the present invention or the additional therapeutic agent alone.

A further aspect of the present invention concerns combination therapy involving ad ministering a compound of formula (I) of the present invention together with an anti-fibrotic compound different form the compound of formula (I) to a mammal in need thereof. In a fur ther embodiment, such anti-fibrotic compound may be selected from the following non-limiting group of anti-fibrotic compounds: pirfenidone, nintedanib, simtuzumab (GS-6624, AB0024), BG00011 (STX100), PRM-151, PRM-167, PEG-FGF21, BMS-986020, FG-3019, MN-001, IW001, SARI 56597, GSK2126458, PAT-1251 and PBI-4050.

A still further aspect of the present invention concerns combination therapy involving administering a compound of formula (I) in combination with a further conventional cancer treatment such as chemotherapy or radiotherapy, or treatment with immunostimulating sub stances, gene therapy, treatment with antibodies and treatment using dendritic cells, to a mam mal in need thereof.

In an embodiment the compound of formula (I) is administered together with at least one additional therapeutic agent selected from an antineoplastic chemotherapy agent. In a further embodiment, the antineoplastic chemotherapeutic agent is selected from: all-trans retin oic acid, Actimide, Azacitidine, Azathioprine, Bleomycin, Carboplatin, Capecitabine, Cisplatin, Chlorambucil, Cyclophosphamide, Cytarabine, Daunorubicin, Docetaxel, Doxifluridine, Doxo rubicin, Epirubicin, Etoposide, Fludarabine, Fluorouracil, Gemcitabine, Hydroxyurea, Idarubi- cin, Irinotecan, Lenalidomide, Leucovorin, Mechlorethamine, Melphalan, Mercaptopurine, Methotrexate, Mitoxantrone, Oxabplatin, Paclitaxel, Pemetrexed, Revlimid, Temozolomide, Teniposide, Thioguanine, Valrubicin, Vinblastine, Vincristine, Vindesine and Vinorelbine. In one embodiment, a chemotherapeutic agent for use in the combination of the present agent may, itself, be a combination of different chemotherapeutic agents. Suitable combinations in clude FOLFOX and IFF. FOFFOX is a combination which includes 5 -fluorouracil (5-FU), leu covorin, and oxabplatin. IFF treatment includes irinotecan, 5-FU, and leucovorin.

In a further embodiment of the present invention, the further conventional cancer treat ment includes radiation therapy. In some embodiments, radiation therapy includes localized ra diation therapy delivered to the tumor. In some embodiments, radiation therapy includes total body irradiation.

In other embodiments of the present invention the further cancer treatment is selected from the group of immunostimulating substances e.g. cytokines and antibodies. Such cytokines may be selected from the group consisting of, but not limited to: GM-CSF, type I IFN, interleu kin 21, interleukin 2, interleukin 12 and interleukin 15. The antibody is preferably an im munostimulating antibody such as anti-CD40 or anti-CTFA-4 antibodies. The immunostimula- tory substance may also be a substance capable of depletion of immune inhibitory cells (e.g. regulatory T-cells) or factors, said substance may for example be E3 ubiquitin ligases. E3 ubiq- uitin ligases (the HECT, RING and U-box proteins) have emerged as key molecular regulators of immune cell function, and each may be involved in the regulation of immune responses dur ing infection by targeting specific inhibitory molecules for proteolytic destruction. Several HECT and RING E3 proteins have now also been linked to the induction and maintenance of immune self-tolerance: c-Cbl, Cbl-b, GRAIF, Itch and Nedd4 each negatively regulate T cell growth factor production and proliferation. In some embodiments of the present invention the compound of formula (I) is adminis tered together with at least one additional therapeutic agent selected from a checkpoint inhibi tor. In some embodiments of the invention, the checkpoint inhibitor is acting on one or more of the following, non-limiting group of targets: CEACAM1, galectin-9, TIM3, CD80, CTLA4, PD-1, PD-L1, HVEM, BTLA, CD160, VISTA, B7-H4, B7-2, CD155, CD226, TIGIT, CD96, LAG3, GITF, 0X40, CD137, CD40, IDO, and TDO. These are known targets and some of these targets are described in Melero et al, Nature Reviews Cancer (2015). Examples of check point inhibitors administered together with the compound of formula (1) are Anti -PD-1 :

Nivolumab, Pembrolizumab, Cemiplimab. Anti-PD-Ll : Atezolizumab, Avelumab, Durval- umab and one Anti-CTLA-4: Ipilimumab. Each one of these check point inhibitors can be made the subject of an embodiment in combination with any one of the compounds of formula (1).

In some embodiments of the present invention the compound of formula (I) is adminis tered together with at least one additional therapeutic agent selected from an inhibitor of in- doleamine-2, 3-dioxygenase (IDO).

In some embodiments of the present invention the compound of formula (I) is adminis tered together with at least one additional therapeutic agent selected from one or more inhibi tors of the CTLA4 pathway. In some embodiments, the inhibitor of the CTLA4 pathway is se lected from one or more antibodies against CTLA4.

In some embodiments of the present invention the compound of formula (I) is adminis tered together with at least one additional therapeutic agent selected from one or more inhibi tors of the PD-l/PD-L pathway. In some embodiments, the one or more inhibitors of the PD- 1/PD-L pathway are selected from one or more antibodies against PD-1, PD-L1, and/or PD-L2.

Detailed Description

The compound of formula I is tested in a Phase 1, randomized, double-blind, placebo- controlled, first-in-human study in which the safety, tolerability, and pharmacokinetics of orally administered compound of formula I is evaluated in healthy adult subjects and adult subjects with indication of suspected Nonalcoholic steatohepatitis (NASH) and liver fibrosis. This study aims to obtain safety, tolerability and pharmacokinetic data when the com pound of formula I is administered orally as single doses and as multiple doses to healthy sub jects. In addition, an optional part has been included to obtain safety and tolerability data when the compound of formula I is administered orally as multiple doses to subjects with suspected Nonalcoholic steatohepatitis (NASH) and liver fibrosis.

Based on results from a first in human study with single and multiple ascending doses it was concluded that the compound of formula (I) is both safe and well tolerated in man and fa vorable PK parameters support once daily or twice daily dosing for a specific dose range, typi cally from 5mg to 200mg once or twice daily, such as from lOmg tolOOmg twice daily.

The compound of formula (I) has the chemical name (IUPAC) 5-Bromopyri din-3 -yl 3- deoxy-3 - [4-(3 ,4, 5-trifluorophenyl)- 1 H- 1 ,2,3 -triazol- 1 -yl] - 1 -thio-a-D-galactopyranoside.

The compound is herein referred to as’’the compound of formula I”.

In the liquid composition comprising a mixture of a dissolved or dispersed polymer for stabilizing the amorphous form of the compound of formula I and a solution of a compound of formula I, the polymer is dissolved or dispersed in a suitable solvent which is dependent on the polymer used. The person skilled in the art can easily prepare a screening of various solvents as well as solvent mixtures to find the optimal solvent, for instance when HPMC is used dichloro- methane, ethanol, water or a mixture of water and acetone are suitable. Furthermore, the com- pound of formula I can be dissolved in various solvents, such as acetone, tetrahydrofuran,

DMSO, methanol, ethanol, dichloromethane, and here the skilled person can perform a screen ing of suitable solvents or solvent mixtures to find the optimal solvent. For instance, for the compound of formula I acetone and THF are preferred, and in particular a mixture of acetone in water is preferred. It should be noted that the polymer and compound of formula I may be dis solved in the same solvent.

When preparing the liquid composition comprising a mixture of a dissolved or dispersed polymer for stabilizing amorphous form of the compound of formula I and a solution of a com pound of formula I, and optionally a solution of a pharmaceutically acceptable additive, the polymer is dissolved or dispersed in a first solvent and the compound of formula I is dissolved in a second solvent. The first and second solvent may be the same or different. For instance, the polymer may be dissolved in a solvent such as a mixture of acetone and water and the com pound of formula I is then dissolved in the same solvent solution comprising the polymer. The order of dissolving the polymer and the compound of formula I is not critical, however, in one embodiment it is preferred to first dissolve the polymer and then the compound of formula I in the solvent comprising the polymer, and in a second embodiment it is preferred to dissolve the polymer in one solvent and the compound of formula I in a different solvent and the mix the two solutions. Optionally a pharmaceutically acceptable additive is dissolved in any one of the solutions, such as the first or second solution or dispersion, or in the mixture of the first and second solution or dispersion, or in the solution or dispersion comprising the polymer and/or the compound of formula I. The pharmaceutically acceptable additive is typically selected from suitable surfactants and/ or pore forming excipients.

The amorphous compound of formula I can be stabilized as an amorphous solid disper sion or as a drug layered composition.

Thus, the present invention relates to an amorphous solid dispersion composition com prising a mixture of an amorphous compound of formula I and a polymer for stabilizing the amorphous form of the compound of formula I. Optionally a pharmaceutically acceptable addi tive may be comprised by the composition.

The weight ratio of the amorphous compound of formula I to the polymer can be modi fied dependent on the polymer employed to provide the most stable amorphous compound of formula I and a suitable range is from 1 :0.5 to 1 :5. To prevent recrystallization over time of the amorphous form of the compound of formula I in the amorphous solid dispersion a more pre ferred weight ratio is from 1 : 1 to 1 :3, and it was found that the most optimal weight ratio is about 1 :2. In addition, or independently, the amorphous compound of formula I is present in a concentration from 0.5 %w/w to 90 %w/w and the polymer is present in a concentration of at least 0.1 %w/w, and optionally a pharmaceutically acceptable additive adding up to 100 %w/w. For instance, the amorphous compound of formula I is present in a concentration selected from the range consisting of 1 %w/w to 80 %w/w, 2 to 70 %w/w, 5 to 60 %w/w, 10 to 50 %w/w, and 20 to 40 %w/w. Preferably, the amorphous compound of formula I is present in a concen tration in the range from 30-40 %w/w. For instance, the polymer is present in a concentration selected from the range consisting of 0.1 %w/w to 99 %w/w, 1 to 80 %w/w, 5 to 70 %w/w, 10 to 60 %w/w, 20 to 50% w/w, and 30 to 40 %w/w. Preferably, the polymer is present in a con centration in the range from 60-70 %w/w.

The amorphous solid dispersion composition of the present invention can be filled into capsules or be compressed into tablets for oral administration.

Typically, the present invention relates to a tablet composition for oral administration comprising the amorphous solid dispersion composition of the present invention. The above- mentioned weight ratios of the amorphous compound of formula I to the polymer and concen trations of the amorphous compound of formula I and the polymer before adding excipients, such as binding agents, disintegrants, lubricants, bulking agents etc, also apply to the tablet of the present invention.

Further, the present invention relates to a drug layered composition comprising a) an in ert solid core having an outer surface and b) a mixture of amorphous compound of formula I, a polymer for stabilizing the amorphous form of the compound of formula I, wherein the mixture is applied as a layer, for instance by spraying the mixture dissolved in a suitable solvent onto the outer surface of the inert solid core and evaporating off the solvent. The weight ratio of the amorphous compound of formula I to the polymer can be modified to provide the most stable drug layered composition and a suitable range is from 1 :0.5 to 1 :5. To prevent recrystallization over time of the amorphous form of the compound of formula I in the drug layered composition a more preferred weight ratio is from 1 : 1 to 1 :3, and a most preferred weight ratio is about 1 :2. In addition, or independently, the amorphous compound of formula I is present in a concentra tion from 0.5 %w/w to 20 %w/w, the polymer is present in a concentration from 1 %w/w to 40 %w/w, the inert solid core is present in a concentration of 40 %w/w to 98.5 %w/w, and option ally a pharmaceutically acceptable additive adding up to 100 %w/w. For instance, the amor phous compound of formula I is present in a concentration selected from the range consisting of 1 %w/w to 18 %w/w, 5 %w/w to 15 %w/w, 7 %w/w to 15 %w/w, and 10 %w/w to 14 %w/w. For instance, the polymer is present in a concentration selected from the range consist ing of 1% w/w to 40% w/w, 5 %w/w to 35 %w/w, 10 %w/w to 30 %w/w, and 15 %w/w to 25 %w/w. For instance, the inert solid core is present in a concentration selected from the range consisting of 45 %w/w to 90 %w/w, 50 %w/w to 80 %w/w, and 60 %w/w to 70 %w/w. Prefer ably, the drug layered composition comprises the amorphous compound of formula I in a con centration from 1-12 %w/w, the polymer in a concentration from 2-24 %w/w and the inert solid core in a concentration from 64-97 %w/w. One example is 1.2 %w/w amorphous compound of formula I, 2.4 %w/w polymer and 96.4 %w/w inert solid core and another example is 12 %w/w amorphous compound of formula I, 24 %w/w polymer and 64 %w/w inert solid core. Another preferred embodiment concerns the drug layered composition comprising the amorphous com pound of formula I in a concentration from 1-12 %w/w, the polymer in a concentration from 2- 24 %w/w, the inert solid core in a concentration from 45-90 %w/w and the pore forming excip ient in a concentration from 5-20 %w/w. One example is 12 %w/w amorphous compound of formula I, 24 %w/w polymer, 56.8 %w/w inert solid core and 7.2 %w/w pore forming excipi ent.

The mixture of amorphous compound of formula I and the polymer may be layered on the whole surface of the inert solid core, or just parts of the surface, since the inert solid core merely serves as a carrier.

The drug layered composition of the present invention is typically filled into a capsule suitable for oral administration. Such capsule may be a gelatin capsule, such as a size-0 Licap hard shell gelatin capsule.

In the above aspects and embodiments, where the polymer for stabilizing the amorphous form of the compound of formula I is part of the composition, such polymer is selected from the group consisting of one or more polyethylene glycol (PEG), carboxymethylethylcellulose (CMEC), ethyl cellulose, hydroxyethylcellulose (HEC), methyl cellulose (MC), polyvinylpyrrolidone vinyl acetate (PVPVA), Polyvinylpyrrolidone (PVP), hydroxy propyl me thyl cellulose (Hypromellose or HPMC)), Hypromellose phthalate, and Hypromellose acetate succinate (HPMC-AS), Eudragit, poloxamer and Gelucire.

In preferred embodiments the polymer is selected from the group consisting of one or more polyethylene glycol (PEG300 to PEG10000), carboxymethylethylcellulose (CMEC), ethyl cellulose, hydroxyethylcellulose (HEC), methyl cellulose (MC), polyvinylpyrrolidone vi nyl acetate (PVP/VA), Polyvinylpyrrolidone, such as (PVP K30), hydroxy propyl methyl cel lulose (Hypromellose), e.g. Hypromellose 603 (HPMC 603), Hypromellose 606 (HPMC 606), Hypromellose 615 (HPMC 615), Hypromellose phthalate -55 (HP-55), and Hypromellose ace tate succinate -LF (HPMCAS-LF), Eudragit LI 00-55, and Gelucire 44/14, preferably a hy droxy propyl methyl cellulose, such as HPMC 603.

In a further embodiment the amorphous compound of formula I is present in the compo sition, such as the drug layered composition or the amorphous solid dispersion composition, in an amount of 0.1 mg to lOOOmg, such as 5mg to 900mg. In a further embodiment the com pound of formula I is present in an amount of 10-800mg, 20-700mg, 30 to 600mg, 40 to 500mg, 50 to 400mg, 60 to 300mg, 70 to 200mg, 80 to 150mg, or 90 to lOOmg. Preferably, one tablet contains the compound of formula I in an amount of l-200mg, such as from 5 to lOOmg. Preferably one capsule contains the compound of formula I in an amount of l-200mg, such as from 5 to lOOmg. In a typical embodiment this amount is administered once daily or twice daily.

In the above aspects and embodiments, where the inert solid core is part of the composi tion, the inert solid core is made of a material selected from the group consisting of sugars and microcrystalline cellulose (MCC).

Typically, the solid core is made of starch, sucrose, and microcrystalline cellulose (MCC), preferably MCC.

In a further embodiment the inert solid core has a size which is sufficiently large to carry the amorphous compound of formula I in a therapeutically relevant amount, and the size is typically at least 40 pm, such as at least 50 pm. Since the drug layered composition should fit into a capsule for oral administration the size should not exceed 3000 pm, such large particles may also carry less active drug and thus not be optimal for a capsule. Preferred inert cores should be in the range from 50 to 2000 pm, such as from 100 to 1400 pm, 100 to 200 pm, 200 to 355 pm, 355-500 pm, 500 to 710 pm, 710 to 1000 pm, and 1000 to 1400 pm.

In the method of preparing the amorphous solid dispersion composition of the present invention the spray drying of the liquid composition comprising the mixture of the dissolved or dispersed polymer for stabilizing amorphous form of the compound of formula I and a dis solved compound of formula I, can be performed using a ProCepT 4M8 Trix spray drier with the following parameters:

In the method of preparing the drug layered composition of the present invention the spraying of the liquid composition comprising the mixture of the dissolved or dispersed poly mer for stabilizing amorphous form of the compound of formula I and a dissolved compound of formula I, onto the inert solid core can be performed using a fluid bed fitted with a Wurster insert such as an Aeromatic STREA-1 or a Glatt GPCG3 with the following coating solution and parameters (wherein‘API’ is used interchangebly with the‘compound of formula G):

The term“treatment” and“treating” as used herein means the management and care of a patient for the purpose of combating a condition, such as a disease or a disorder. The term is intended to include the full spectrum of treatments for a given condition from which the patient is suffering, such as administration of the active compound to alleviate the symptoms or com plications, to delay the progression of the disease, disorder or condition, to alleviate or relief the symptoms and complications, and/or to cure or eliminate the disease, disorder or condition as well as to prevent the condition, wherein prevention is to be understood as the management and care of a patient for the purpose of combating the disease, condition, or disorder and includes the administration of the active compounds to prevent the onset of the symptoms or complica tions. The treatment may be performed in a chronic way. The patient to be treated may be a hu man subject diagnosed with pulmonary fibrosis or other types of lung fibrosis, kidney fibrosis or NASH.

As used herein“pharmaceutically acceptable additive” is intended without limitation to include carriers, excipients, diluents, adjuvant, colorings, aroma, preservatives etc. that the skilled person would consider using when formulating a compound of the present invention in order to make a pharmaceutical composition.

The adjuvants, diluents, excipients and/or carriers that may be used in the composition of the invention must be pharmaceutically acceptable in the sense of being compatible with the compound of formula (I) and the other ingredients of the pharmaceutical composition, and not deleterious to the recipient thereof. It is preferred that the compositions shall not contain any material that may cause an adverse reaction, such as an allergic reaction. The adjuvants, diluents, excipients and carriers that may be used in the pharmaceutical composition of the in vention are well known to a person within the art.

As mentioned above, the compositions and particularly pharmaceutical compositions as herein disclosed may, in addition to the compounds herein disclosed, further comprise at least one pharmaceutically acceptable adjuvant, diluent, excipient and/or carrier. The combined amount of the active ingredient and of the pharmaceutically acceptable adjuvant, diluent, excip ient and/or carrier may not constitute more than 100% by weight (100 %w/w) of the composi tion, particularly the pharmaceutical composition.

Further embodiments of the process are described in the experimental section herein, and each individual process as well as each starting material constitutes embodiments that may form part of embodiments.

The above embodiments should be seen as referring to any one of the aspects (such as ‘method for treatment’,‘pharmaceutical composition’,‘compound for use as a medicament’, or ‘compound for use in a method’) described herein as well as any one of the embodiments de scribed herein unless it is specified that an embodiment relates to a certain aspect or aspects of the present invention.

All references, including publications, patent applications and patents, cited herein are hereby incorporated by reference to the same extent as if each reference was individually and specifically indicated to be incorporated by reference and was set forth in its entirety herein.

All headings and sub-headings are used herein for convenience only and should not be construed as limiting the invention in any way.

Any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contra dicted by context.

The terms“a” and“an” and“the” and similar referents as used in the context of describ ing the invention are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context.

Recitation of ranges of values herein are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless other- wise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. Unless otherwise stated, all exact values provided herein are repre sentative of corresponding approximate values ( e.g ., all exact exemplary values provided with respect to a particular factor or measurement can be considered to also pro-vide a correspond ing approximate measurement, modified by "about," where appropriate).

All methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context.

The use of any and all examples, or exemplary language (e.g.,“such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise indicated. No language in the specification should be construed as indicating any element is essential to the practice of the invention unless as much is explicitly stated.

The citation and incorporation of patent documents herein is done for convenience only and does not reflect any view of the validity, patentability and/or enforceability of such patent documents.

The description herein of any aspect or embodiment of the invention using terms such as“comprising”,“having”,“including” or“containing” with reference to an element or ele ments is intended to provide support for a similar aspect or embodiment of the invention that “consists of’,“consists essentially of’, or“substantially comprises” that particular element or elements, unless otherwise stated or clearly contradicted by context (e.g., a composition de scribed herein as comprising a particular element should be understood as also describing a composition consisting of that element, unless otherwise stated or clearly contradicted by con text).

This invention includes all modifications and equivalents of the subject matter recited in the aspects or claims presented herein to the maximum extent permitted by applicable law.

The present invention is further illustrated by the following examples that, however, are not to be construed as limiting the scope of protection. The features disclosed in the foregoing description and in the following examples may, both separately and in any combination thereof, be material for realizing the invention in diverse forms thereof. Experimental

Amorphous Solid Dispersion

A small trial batch of API-HPMC 603 (1 :2) Spray Dried Dispersion (SDD) was prepared using a 4M8 TriX spray drier from ProCepT. One gram of HPMC 603 was dissolved in 22.2 mL of DCM/ethanol (1/1) solution (45 mg/mL) and 500 mg API was dissolved in 10 ml of tetrahydro- furan (THF) (50 mg/mL). The two solutions were mixed at room temperature, filtered through 45 -pm filters followed by spray drying using the parameters listed below.

Parameters used for spray drying:

The prepared SDD was characterized by X-ray powder diffraction (XRPD) and modu lated Differential Scanning Calorimetry (mDSC). The XRPD data shows that the SDD was amorphous and a single Tg at 109°C derived from the mDSC scan indicated the formation of a miscible system between API and HPMC 603 without phase separation. The yield of spray dry ing was 78% with an HPLC assay of 97%.

Figure 1 is an XRPD diffractogram of the API/HPMC 603 (1 :2) SDD.

Figure 2 is an mDSC thermogram (reversing signal) of the API/HPMC 603 (1 :2) SDD showing a Tg at 109°C. Dissolution results of the API-HPMC 603 SDD compared to non-formulated API are shown in figure 3. Due to the low solubility of API, less than 12% API (i.e. 0.011 mg/mL in FaSSIF) from capsules containing API only was released in the dissolution media over 18 hours. Compared to the non-formulated API, API-HPMC603 SDD filled capsules showed at least 8 times higher solubility of API and a much-improved dissolution profile with over 97% API released (0.096 mg/mL in FaSSIF) within 18 hours.

As plug formation was observed for the SDD, a third formulation composed of a physi cal mixture of Poloxamer 188 with the SDD was prepared. The addition of poloxamer 188 pre vented plug formation and resulted in a faster rate of disintegration/dissolution of API (i.e. 74% with Poloxamer 188 (0.073 mg/mL) vs. 43% without Poloxamer 188 (0.042 mg/mL) after 2 hours of dissolution).

A 4- week stability study was conducted on the prepared SDD’s with and without polox amer. Following storage at 25°C/60% RH and 40°C/75% RH for up to 4 weeks there was no change in assay or dissolution profile. The SDD’s remained amorphous at both conditions, however, the Tg appeared to shift to 60-70°C during storage.

As an alternative to poloxamer 188, the surfactant SLS was also evaluated to aid in the dispersion of the SDD. SDD’s with both poloxamer and Sodium Lauryl Sulfate (SLS) yielded similar release profiles, however, in the presence of SLS, the SDD powder was visually dis persed completely in the dissolution medium within 10 minutes, which is faster than with the use of Poloxamer 188 (~30 minutes).

Spray Coating/Drug Layering of Amorphous Compound of formula I

Drug layering on a small scale (30g of layered pellets) was conducted by using an Aero- matic coater (fluid bed laboratory unit, STREA-1 from GEA Niro Inc). The composition of the coating solution and the parameters used during the drug layering process are provided below.

Coating solution and parameters used for the drug layering process:

The coating solution for applying the drug: polymer mixture over the inert cores was prepared as follows. To prepare 200mL coating solution, 12g of HPMC 603 was added into 40 mL of distilled water in a beaker and the mixture was heated to 70-80°C, stirring using a magnetic stirrer. After the HPMC 603 was fully dispersed, the solution was cooled down to room tem- perature resulting in a clear HPMC 603 solution. 140mL acetone was added into the solution and the mixture was stirred using a magnetic stirrer until a homogenous solution was achieved. To this, 6g of API was added and the solution was stirred until all of the API dissolved. During the process, the beaker was sealed using parafilm to prevent the evaporation of acetone. Fi nally, the coating solution was sieved through 0.25mm mesh prior to use.

30g VIVAPUR ® microcrystalline cellulose (MCC) spheres 700 (710-1000 pm) from

JRS was used for drug layering. The final composition of the drug layered MCC spheres is shown below.

Composition of drug layered pellets:

The resulting drug layered pellets were analysed by UV-HPLC to determine the drug loading as well as by SEM, XRPD and Dissolution in biorelevant media.

The UV-HPLC results showed that the drug loading of the pellets was 12.19% with no degradation of API observed. XRPD showed that API remained amorphous after drug layering.

The dissolution profile of the drug-layered pellets was compared to the dissolution pro files of the API alone as well an ASD formulation (physical mixture of SDD

(API:HPMC603=1 :2) and poloxamer 188) (see figure 4). The drug layered pellets show a simi lar maximum %release to that of the ASD formulation in three hours, whereas the initial release rate was a bit slower with approximately 33% of API released in 30 minutes (as compared to approximately 50% released from the ASD formulation).

To investigate the slower dissolution further, an HPC coating was applied to the surface of the drug layered pellets (7% weight gain). The coating process was conducted using a Ca- leva mini coater drier and 5g of drug layered pellets were sprayed for approximately 1 hour with the coating solution to reach a 7% polymer weight gain. The coating solution used was a 5% w/w HPC aqueous solution and the parameters used as well as the composition of the final

HPC coated drug layered pellets are shown below.

Parameters used for HPC coating:

Composition of HPC coated drug layered pellets:

The dissolution test of the HPC coated pellets showed that although the HPC coated pellets were fully dispersed in 30 minutes in the biorelevant dissolution media, however the dissolution profile was not improved (see figure 5).

A 4-week stability study was conducted on the drug-layered pellets. The drug-layered pellets were manually filled into size-00 Li cap hard shell gelatin capsules (410mg pellets per capsule, equivalent to 50mg API). The capsules were placed in closed glass vials and were in cubated at 25°C/60%RH and 40°C/75%. At each time point (T=0, 7 days and 4 weeks) the samples were analysed/characterized by XRPD, SEM and dissolution.

The results of the stability study show the pellets remain unchanged following storage at 25°C/60%RH and 40°C/75% for 4 weeks. The dissolution profile of the drug layered pellets did not change at either condition. It was also confirmed by XRPD that API remained amor phous with no evidence of recrystallization during the 4-week period when stored at both 25°C/60%RH and 40°C/75%. The SEM results of the drug layered pellets show that no obvious morphology change to the surface of the pellets was observed during the 4-week storage at both

25°C/60%RH and 40°C/75%.

On a larger scale (1-4 kg coated pellets), drug-layering was conducted using a Glatt GPCG3 fluid bed with a 6 or 7-inch Wurster insert. The equipment parameters used during the drug layering process are provided below. Two different drug loadings of MCC spheres were prepared.

Parameters used for the drug layering process:

The coating solution was prepared as follows. Water was placed in a jacketed mixing bowl and heated to 35-37°C. HPMC was slowly added to the heated water while mixing then the mixture was heated to 52-55°C and mixing was continued until a homogenous dispersion was formed. The mixture was cooled to room temperature and mixing was continued until a clear solution was obtained. The aqueous HPMC solution and acetone were mixed together in a glass container suitable for feeding solutions to a fluid bed coating machine. The API was dis solved in the aqueous HPMC/acetone mixture. MCC microspheres were transferred to the bowl of a fluid bed coating machine and spray coated with the API/aqueous HPMC/acetone mixture until the desired amount of drug layering was achieved. The final composition of the drug lay- ered MCC spheres is shown below.

Composition of drug layered pellets:

Use of pore forming agent:

Additional drug layering trials were conducted with the inclusion of pore forming agents in the layering solution. Pore forming agents such as Calcium Dihydrogen Phosphate, Lactose, Hy- droxypropyl cellulose and microcrystalline cellulose are excipients added to the layering solution along with HPMC 603 and the compound of formula I which cause the formation of small pores or channels in the coating. The pores allow for more rapid wetting of the layered coating thus increasing the rate of drug release from the coated microspheres. These excipients can be added at up to 20% w/w cone. The excipient can be either dissolved in the aqueous HPMC 603 polymer solution or in the final acetone/water solution containing the compound of formula I before the spray coating.

One example of a drug layered pellet containing a pore forming excipient has the composition below:

Composition of drug layered pellets with a Pore Forming excipient:

Drug layered pellets were manufactured using the composition above. SEM analysis showed the presence of pores on the surface of the coated pellets. Dissolution testing showed a faster release profile than drug layered pellets without calcium dihydrogen phosphate.

Pharmacokinetic data in humans

The first part of this study aims to obtain safety, tolerability and pharmacokinetic data when the compound of formula I is administered orally as a single dose in healthy subjects us ing the above drug layered formulation.

Experimental data from healthy subject cohort Al: 5 mg compound of formula I (API)

6 healthy subjects received 5 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A2: 20 mg compound of formula I

6 healthy subjects received 20 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A3 Fasted: 50 mg compound of formula I (API)

6 healthy subjects received 50 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A3 Fed: 50 mg compound of formula I (API)

6 healthy subjects received 50 mg of compound of formula I capsules orally as a single dose in the fed state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A4: 50 mg compound of formula I (API) (as 10x5 mg capsules) 6 healthy subjects received 50 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A5: 100 mg compound of formula I (API)

6 healthy subjects received 100 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A6: 200 mg compound of formula I (API)

6 healthy subjects received 200 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder.

Experimental data from healthy subject cohort A7: 400 mg compound of formula I (API)

6 healthy subjects received 400 mg of compound of formula I capsules orally as a single dose in the fasted state, and 2 subjects receive placebo. The first 2 subjects (1 active and 1 pla cebo) were dosed 24 hours before the remaining subjects. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing, and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. See tables hereunder. The second part of this study aims to obtain safety, tolerability and pharmacokinetic data when the compound of formula I is administered orally dosed twice a day for 10 days in healthy subjects using the above drug layered formulation. Experimental data from healthy subject cohort Bl: 50 mg compound of formula I (API)

8 healthy subjects received 50 mg of compound of formula I capsules and 3 subjects re ceive placebo orally twice a day for 10 days. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing on day 1 , and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24, 36, and 48 hours post dose. Blood sampling was repeated on day 10. See tables hereunder.

Experimental data from healthy subject cohort B2: 100 mg compound of formula I

8 healthy subjects received 100 mg of compound of formula I capsules and 3 subjects receive placebo orally twice a day for 10 days. Blood samples for pharmacokinetic analysis of plasma API concentrations were obtained by venipuncture before dosing on day 1 , and then at 0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 16, 24 hours post dose. Blood sampling was repeated at day 10. See tables hereunder.