WO2020037189A1 - Formulations of ag10 - Google Patents

Formulations of ag10 Download PDF

Info

Publication number
WO2020037189A1
WO2020037189A1 PCT/US2019/046789 US2019046789W WO2020037189A1 WO 2020037189 A1 WO2020037189 A1 WO 2020037189A1 US 2019046789 W US2019046789 W US 2019046789W WO 2020037189 A1 WO2020037189 A1 WO 2020037189A1
Authority
WO
WIPO (PCT)
Prior art keywords
tablet formulation
tablet
weight
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/US2019/046789
Other languages
French (fr)
Inventor
Jesper JERNELIUS
Mark Michael MENNING
Original Assignee
Eidos Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to AU2019321583A priority Critical patent/AU2019321583A1/en
Priority to KR1020217007760A priority patent/KR20210046708A/en
Priority to EP19849948.5A priority patent/EP3836920A4/en
Priority to JP2021507989A priority patent/JP7469293B2/en
Priority to CN201980054507.6A priority patent/CN112804998A/en
Priority to MX2021001761A priority patent/MX2021001761A/en
Application filed by Eidos Therapeutics, Inc. filed Critical Eidos Therapeutics, Inc.
Priority to SG11202101393PA priority patent/SG11202101393PA/en
Priority to BR112020026493-8A priority patent/BR112020026493A2/en
Priority to EA202190561A priority patent/EA202190561A1/en
Priority to CA3104695A priority patent/CA3104695A1/en
Publication of WO2020037189A1 publication Critical patent/WO2020037189A1/en
Priority to IL280906A priority patent/IL280906A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/28Dragees; Coated pills or tablets, e.g. with film or compression coating
    • A61K9/2806Coating materials
    • A61K9/2833Organic macromolecular compounds

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Inorganic Chemistry (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The present disclosure provides high-load tablet formulations of AG10 or a pharmaceutically acceptable salt thereof. In some aspects, provided herein are table formulations of AG10 or a pharmaceutically acceptable salt thereof that include at least 40% or more AG10 by weight and at least one pharmaceutical excipient selected from one or more fillers, one or more binders, one or more disintegrants, and one or more lubricants.

Description

FORMULATIONS OF AGIO
CROSS-REFERENCES TO RELATED APPLICATIONS
[0001] This application claims the benefit of priority under 35 U.S.C § 119(e) to U.S. Provisional Application Serial No. 62/765,154 filed August 17, 2018, the disclosure of which is incorporated herein by reference in its entirety.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0002] NOT APPLICABLE
REFERENCE TO A "SEQUENCE LISTING," A TABLE, OR A COMPUTER PROGRAM LISTING APPENDIX SUBMITTED ON A COMPACT DISK
[0003] NOT APPLICABLE
BACKGROUND OF THE INVENTION
[0004] Aberrant protein interaction and aggregation, either through protein misfolding or over activation of a signaling pathway is the underlying cause of a large number of human degenerative diseases. As such, targeting protein protein interactions (PPIs) is of therapeutic interest.
[0005] To date approved inhibitors of PPIs are proteins rather than small-molecule inhibitors. For example, therapeutic monoclonal antibodies (mAbs) are used in treating cancer, autoimmune, infectious, and neuodegenerative diseases. Therapeutic mAbs are costly to manufacture, they require administration by injection, and can illicit an immune-response in the patient. For these reasons the development of small-molecule inhibitors of PPIs remains of interest.
[0006] One example of aberrant protein aggregation is the soluble protein transthyretin (TTR or prealbumin). Wild type (WT) TTR is a 55 kDa homotetrameric protein present in blood and cerebrospinal fluid. When dissociated from its homoterameric form, WT TTR dimers can misfold into amyloidogenic monomers. The formation of amyloidogenic monomers has observed with WT TTR as well as more than 100 different mutated variants. Research has shown that stabilizing the tetrameric form of TTR inhibits the misfolding of amyloidogenic monomers and subsequent TTR amyloid formation.
[0007] Recent work has identified 3-(3-(3,5-dimethyl-lH-pyrazol-4-yl)propoxy)-4- fluorobenzoic acid (AG10) as a promising candidate to treat TTR amyloid related diseases such as TTR amyloid cardiomyopathy and ATTR polyneuropathy. This compound has been disclosed in WO 2014/100227. Despite the disclosure of this compound, improved pharmaceutical formulations that provide increased stability and consistent pharmacokinetic data remain elusive.
[0008] As such, there exists a need to produce pharmaceutical formulations suitable for administration to humans or other animals. The present disclosure addresses these needs and provides related advantages as well.
BRIEF SUMMARY OF THE INVENTION
[0009] The present disclosure provides high-load tablet formulations of AG10 or a pharmaceutically acceptable salt thereof and at least one pharmaceutical excipient selected from one or more fillers, one or more binders, one or more disintegrants, and one or more lubricants. In some embodiments, the tablet formulation is coated with a coating agent.
BRIEF DESCRIPTION OF THE DRAWINGS
[0010] FIG. 1 illustrates the process flow diagram for preparing the AG10 formulations described in Example 2. [0011] FIG. 2 shows images of the AG10 HC1 coated tablets prepared in Example 2.
[0012] FIG. 3 show the dissolution profile for AG10 solid tablets formulations described in
Example 2.
[0013] FIG. 4 illustrates the process flow diagram for preparing the AG10 formulations described in Example 3. [0014] FIG. 5 illustrates the process flow for aqueous coating suspension preparation or preparing the AG10 formulations described in Example 3.
[0015] FIG. 6 shows images illustrating the lack of tablet edge’s erosion after friability test for L018A (High hardness, Left), and L018B (Middle hardness, Right) (at 33.0% AG10). [0016] FIG. 7 shows images of major tablet edge’s erosion after friability test for L016 (Left), and for L017 (Right) (Both formulations have a 40% AG10 load and maximum hardness).
[0017] FIG. 8 illustrates the process Flow Diagram for the 33% AG10 HC1 Tablets described in Example 4.
[0018] FIG. 9 illustrates the process Flow Diagram for the 66.7% AG10 HC1 Tablets described in Example 4.
[0019] FIG. 10 shows the dissolution profile of 33.3% AG10 HC1 tablets after storage under 40°C/75% RH Conditions. T=0 (open triangles); T= 1 Month (open diamonds) T= 3 Months (filled circles); T=6 months (filled squares).
[0020] FIG. 11 shows the dissolution profile of 66.7% AG10 HC1 tablets after storage under 40°C/75% RH Conditions. T=0 (open triangles); T= 3 Months (filled circles); T=6 months (filled squares).
DETAILED DESCRIPTION OF THE INVENTION I. General
[0021] The present disclosure is based, in part, on the discovery that formulations containing 40% or more AG10 can be successfully prepared as tablets. These tablets are particularly well suited for administration to human and animal subjects alike because these amounts meet the necessary stability and pharmacokinetic requirements for oral formulations. Other formulations, such as capsules, fail to meet these needs.
[0022] High-load immediate release AG10 tablets were successfully achieved using a high grade microcrystalline cellulose. In contrast, tablet formulations exceeding 33.3% AG10 using standard grades of microcrystalline cellulose showed signs of tablet erosion after friability tests and reduced dissolution rates after extended storage times. II. Definitions
[0023] Unless specifically indicated otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by those of ordinary skill in the art to which this invention belongs. In addition, any method or material similar or equivalent to a method or material described herein can be used in the practice of the present invention. For purposes of the present invention, the following terms are defined.
[0024] The terms“a,”“an,” or“the” as used herein not only include aspects with one member, but also include aspects with more than one member. For instance, the singular forms “a,”“an,” and“the” include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to“a cell” includes a plurality of such cells and reference to“the agent” includes reference to one or more agents known to those skilled in the art, and so forth.
[0025] As used herein, the term "about" means a range of values including the specified value, which a person of ordinary skill in the art would consider reasonably similar to the specified value. In some embodiments, the term "about" means within a standard deviation using measurements generally acceptable in the art. In some embodiments, about means a range extending to +/- 10% of the specified value. In some embodiments, about means the specified value.
[0026] The term“tablet” refers to solid pharmaceutical formulations with and without a coating. The term“tablet” also refers to tablets having one, two, three or even more layers, wherein each of the before mentioned types of tablets may be without or with one or more coatings. In some embodiments, tablets of the present disclosure can be prepared by roller compaction or other suitable means known in the art. The term“tablet” also comprises mini, melt, chewable, effervescent and orally disintegrating tablets. Tablets include AG10 and at least and one pharmaceutical excipient selected from one or more fillers, one or more binders, one or more disintegrants, and one or more lubricants. Optionally, a coating agent is also included. For the purposes of calculating percent weight of the tablet formulation, the amount of coating agent is not included in the calculation. That is, the percent weights reported herein are of the uncoated tablet.
[0027] The term“salt” refers to acid or base salts of the compounds of the present disclosure. Illustrative examples of pharmaceutically acceptable salts are mineral acid (hydrochloric acid, hydrobromic acid, phosphoric acid, and the like) salts, organic acid (acetic acid, propionic acid, glutamic acid, citric acid and the like) salts, quaternary ammonium (methyl iodide, ethyl iodide, and the like) salts. It is understood that the pharmaceutically acceptable salts are non-toxic. Additional information on suitable pharmaceutically acceptable salts can be found in Remington's Pharmaceutical Sciences, l7th ed., Mack Publishing Company, Easton, Pa., 1985, which is incorporated herein by reference. III. Embodiments of the Disclosure
[0028] The present disclosure provides, inter alia, tablet formulations of AG10 or a pharmaceutically acceptable salt thereof. AG10 is a compound having the formula:
Figure imgf000007_0001
[0029] In some embodiments, a pharmaceutically acceptable salt of AG10 is corresponds to Formula I.
Figure imgf000007_0002
wherein X is a pharmaceutically acceptable anion of a protic acid.
[0030] A variety of protic acids are suitable for making a pharmaceutically acceptable salt of
Formula I. It can be seen that the pharmaceutically acceptable anion of the protic acid is dependent upon the protic acid used. For example, protic acids useful in the present disclosure include hydrochloric acid, hydrobromic acid, sulfonic acid, tosylic acid (p- toluenesulfonic acid), methanesulfonic acid, nitric acid, or acetic acid. Thus,
pharmaceutically acceptable anions of a protic acid include chloride (CT), bromide (Br), sulfonate (HS(0)20 ), tosylate (TsO ), mesylate (MsO ), nitrate (NO3 ) and acetate
(CFbC(O)O ), or combinations thereof.
[0031] In some embodiments, the pharmaceutically acceptable anion of a protic acid is mesylate. [0032] In some embodiments, the pharmaceutically acceptable anion of a protic acid is tosylate.
[0033] In some embodiments, the pharmaceutically acceptable anion of a protic acid is chloride, and the pharmaceutically acceptable salt of Formula I is represented by Formula (la)
Figure imgf000008_0001
[0034] Pharmaceutically acceptable salts of Formula I can be produced using a number of conventional means in the art. For example, the free acid form of a compound of Formula I may be contacted with a stoichiometric amount of the appropriate acid in water, an organic solvent, or a mixture of the two. In some embodiments, pharmaceutically acceptable salts of Formula I are made in nonaqueous media such as an ether, ethyl acetate, ethanol, isopropanol, or acetonitrile. In some embodiments, the pharmaceutically acceptable salts of Formula I are made by dissolving a compound of Formula IX in water, adding a suitable amount of HX to form a mixture, and adding a nonaqueous solvent, such as the nonaqueous media described above to crystallize the salt. In some embodiments, a suitable amount of HX is a stoichiometric amount. It is understood the HX comprises a hydrogen and an X is a pharmaceutically acceptable anion of a protic acid as defined above.
[0035] The tablet formulations of the present disclosure can include, for example, about 40 to 85 or about 50 to 75% by weight of AG10 or a pharmaceutically acceptable salt thereof.
In some embodiments, the tablet formulations contain about 50% to 70% by weight of AG10 or a pharmaceutically acceptable salt thereof. In some embodiments, the tablet formulations contain about 50% by weight of AG10 or a pharmaceutically acceptable salt thereof. In some embodiments, the tablet formulations contain about 66.7% by weight of AG10 or a pharmaceutically acceptable salt thereof. In some embodiments, the tablet formulations contain about 75% by weight of AG10 or a pharmaceutically acceptable salt thereof. In some embodiments, the tablet formulations contain about 80% by weight of AG10 or a pharmaceutically acceptable salt thereof. In some embodiments, the tablet formulations contain about 85% by weight of AG10 or a pharmaceutically acceptable salt thereof.
[0036] The amount of AG10 or a pharmaceutically acceptable salt thereof, in a tablet formulation can be about 0.1 to about 500 mg, about 0.1 to about 250 mg, or about 0.1 to about 100 mg. In some embodiments, the amount of AG10 present in a tablet formulation is about 10, 25, 50, 100, 200, 300, 400, or 500 mg. In some embodiments, the amount of AG10 present in a tablet formulation is about 50, 100, 200, or 400 mg. In some embodiments, the total weight (e.g., active ingredients plus excipients - not including coating) of the tablet formulation is about 50 to about 1500 mg. For example, the total weight of the solid dosage form is about 100, 150, 200, 300, 400, 500, 600, 700, 800, 900, 1000, or 1500 mg.
[0037] The tablet formulations of the present disclosure can include at least one ingredient selected from one or more fillers, one or more binders, one or more disintegrates, and one or more lubricants or other ingredient. In some embodiments, the tablet formulation comprises one or more excipients selected from a high grade microcrystalline filler, an inorganic salt filler, a disintegrant, and a lubricant.
[0038] In some embodiments, the tablet formulations of the present disclosure include one or more fillers. Suitable fillers are described below. In some embodiments, the one or more fillers are present in an amount of about 1 to 60, 5 to 55, 10 to 50, or 15 to 45% by weight. In some embodiments, one or more fillers are present in about 42.5% by weight. In some embodiments, one or more fillers are present in about 25.8% by weight. In some
embodiments, one or more fillers are present in about 17.5% by weight.
[0039] In some embodiments, tablet formulations of the present disclosure include one to three fillers. In some embodiments, tablet formulations of the present disclosure include one to two fillers. In some embodiments, tablet formulations of the present disclosure include two fillers.
[0040] Suitable fillers include, for example, oligosaccharides (e.g., lactose), sugars, starches, modified starches, sugar alcohols (e.g. mannitol, sorbitol, xylitol, lactitol), inorganic salts, cellulose derivatives (e.g. microcrystalline cellulose, silicified microcrystalline cellulose, cellulose, hypromellose), calcium sulfate, aluminum and magnesium silicate complexes and oxides, and the like. Example of inorganic salt fillers include a phosphate salt, such as dibasic calcium phosphate dehydrate, salts of sulfates, and silicon dioxide. In some embodiments, the one or more fillers include cellulose derivatives or alkaline earth metal salts of chloride, phosphates, sulfates, and the like. In some embodiments, the one or more fillers include a cellulose derivative and an inorganic salt. In some embodiments, the one or more fillers are microcrystalline cellulose and silicon dioxide. In some embodiments, the one or more fillers are microcrystalline cellulose. In some embodiments, the
microcrystalline cellulose is a high grade microcrystalline cellulose.
[0041] A high grade microcrystalline cellulose is a cellulose derived product that has specific properties that are not the dominant features in more standard preparations of microcrystalline cellulose. For example, in some embodiments, a high grade microcrystalline cellulose is characterized by cellulose polymers with spherical morphology and porous structure. These properties are found in UF grade microcrystalline cellulose from
CEOLUS™ (e.g. UF-702 and UF-711) and similar available products. In some
embodiments, a high grade microcrystalline cellulose is characterized by cellulose polymers with needle-like particle shape. These properties are found in KG grade microcrystalline cellulose from CEOLUS™ (e.g. KG-802 and KG-1000).
[0042] The high grade cellulose filler can be present in an amount of about 1 to 60% by weight. In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 5 to 55% by weight. In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 10 to 50% by weight. In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 15 to 45% by weight.
In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, or 45% by weight. In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 17%. In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 26%. In some embodiments, the high grade microcrystalline cellulose is present in an amount of about 42%.
[0043] In some embodiments, the tablet formulations of the present disclosure include one or more binders. Suitable binders are described below. In some embodiments, the one or more binders are present in an amount of about 0.5 to 15, about 0.5 to 10, or about 1 to 10% by weight. In some embodiments, the one or more binders are present in an amount of about 3 to 8% by weight. In some embodiments, the one or more binders are present in an amount of about 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10% by weight. In some embodiments, the one or more binders are present in about 5% by weight.
[0044] In some embodiments, tablet formulations of the present disclosure include one to three binders. In some embodiments, tablet formulations of the present disclosure include one binder.
[0045] Suitable binders include, for example, povidone, lactose, starches, modified starches, sugars, gum acacia, gum tragacanth, guar gum, pectin, wax binders,
methylcellulose, carboxymethylcellulose, hydroxypropyl methylcellulose, hydroxyethyl cellulose, hydroxypropyl cellulose, copolyvidone, gelatin, sodium alginate, and the like. Non-cellulosic binders include polymeric and other binders lacking a cellulose backbone. Examples of non-cellulosic binders include povidone, lactose, starches, modified starches, gums, guar gum, pectin, waxes, gelatins, alginates, and the like. In some embodiments, formulations contain a non-cellulosic binder such as povidone or copovidone. In some embodiments, the non-cellulosic binder is copovidone.
[0046] In some embodiments, the tablet formulations of the present disclosure include one or more disintegrants. Suitable disintegrants are described below. In some embodiments, the one or more disintegrants are present in an amount of about 1 to 15, about 1 to about 12, or about 1 to about 10% by weight. In some embodiments, one or more disintegrants are present in about 3-8% by weight. In some embodiments, the formulations contain about 3, 4, 5, 6, 7, or 8% by weight of disintegrant. In some embodiments, the formulations contain about 5% by weight of disintegrant. In some embodiments, the formulations contain about 6% by weight of disintegrant.
[0047] In some embodiments, tablet formulations of the present disclosure include one to three disintegrants. In some embodiments, tablet formulations of the present disclosure include one disintegrant.
[0048] Suitable disintegrants include, for example, croscarmellose sodium, crospovidone, polyvinylpyrrolidone, sodium starch glycolate, com starch. In some embodiments, the formulations contain a disintegrant such as sodium starch glycolate or crospovidone. In some embodiments, the disintegrant is croscarmellose sodium.
[0049] In some embodiments, the tablet formulations of the present disclosure include one or more lubricants. Suitable lubricants are described below. In some embodiments, the one or more lubricants are present in an amount of about 0.1 to 8, 0.5 to 5, 0.5 to 3% by weight.
In some embodiments, one or more lubricants are present in an amount of about 0.5, 0.75, 1, 1.5, 2, 3, 4, or 5% by weight. In some embodiments, one or more lubricants are present in an amount of about 2% by weight. In some embodiments, one or more lubricants are present in an amount of about 1.5% by weight.
[0050] In some embodiments, tablet formulations of the present disclosure include one to three lubricants. In some embodiments, tablet formulations of the present disclosure include one lubricant.
[0051] Suitable lubricants include, for example, magnesium stearate, stearic acid, palmitic acid, calcium stearate, talc, camauba wax, hydrogenated vegetable oils, mineral oil, polyethylene glycols, and sodium stearyl fumarate. In some embodiments, the one or more lubricants are magnesium stearate and/or sodium stearyl fumarate. In some embodiments, the one or more lubricants is magnesium stearate.
[0052] Other suitable fillers, binders, disintegrants, lubricants and other excipients which may be used are described in Handbook of Pharmaceutical Excipients, 2nd Edition, American Lachman, Leon, 1976; Pharmaceutical Dosage Forms: Tablets Volume 1, 2nd Edition, Lieberman, Herbert A., et al, 1989; Modem Pharmaceutics, Banker, Gilbert and Rhodes, Christopher T, 1979; and Remington's Pharmaceutical Sciences, 15th Edition, 1975, each of which is incorporated herein by reference in its entirety.
[0053] In some embodiments, the tablet is coated with a coating agent. Suitable coating agents include ethylcellulose, polymethacrylates, as well as coating products sold by
OPADRY™. In some embodiments, the coating agent is Opadry Clear, Opadry Blue 13B50579, Opadry White 33628707, Opadrya QX 321A180025, or Opadry II (33G28707).
In some embodiments the coating agent is Opadry White 33628707. In some embodiments the coating agent is Opadry QX 321 Al 80025. In some embodiments the coating agent is Opadry II (33G28707). For the purposes of calculating percent weight of the tablet formulation, the amount of coating agent is not included in the calculation. That is, the percent weights reported herein are of the uncoated tablet.
[0054] In some embodiments, the tablet formulation contains about 40 to 85% by weight of AG10 or a pharmaceutically acceptable salt thereof; about 5 to 55% by weight of one or more fillers; about 0 to 15% by weight of one or more binders; about 1 to 15% by weight of one or more disintegrants; and about 0.1 to 8% by weight of one or more lubricants. In some embodiments, the noted formulation includes a coating agent.
[0055] In some embodiments, the tablet formulation contains about 50 to 75% by weight of AG10 or a pharmaceutically acceptable salt thereof; about 10 to 50% by weight of one or more fillers; about 3 to about 8% by weight of one or more disintegrants; and 0.5 to 3% by weight of one or more lubricants. In some embodiments, the noted formulation includes a coating agent.
[0056] In some embodiments, the tablet formulation contains about 50% by weight of AG10 or a pharmaceutically acceptable salt thereof; about 42.5% by weight of one or more fillers; about 6% by weight of a disintegrant; and about 1.5% by weight of a lubricant. In some embodiments, the noted formulation includes a coating agent. [0057] In some embodiments, the tablet formulation contains about 66.7% by weight of AG10 or a pharmaceutically acceptable salt thereof; about 25.8% by weight of one or more fillers; about 6% by weight of a disintegrant; and about 1.5% by weight of a lubricant. In some embodiments, the noted formulation includes a coating agent.
[0058] In some embodiments, the tablet formulation contains about 75% by weight of AG10 or a pharmaceutically acceptable salt thereof; about 17.5% by weight of one or more fillers; about 6% by weight of a disintegrant; and about 1.5% by weight of a lubricant. In some embodiments, the noted formulation includes a coating agent.
[0059] In some embodiments, the tablet formulation of the present disclosure are at least 75% dissolved after 10 minutes in a solution of 0.1N HC1 at 37±0.5 °C in an Apparatus-II (Paddles) with a paddle speed of about 50 rpm. In some embodiments, the tablet formulation of the present disclosure are at least 85% dissolved after 10 minutes in a solution of 0.1N HC1 at 37±0.5 °C in an Apparatus-II (Paddles) with a paddle speed of about 50 rpm. In some embodiments, the tablet formulation of the present disclosure are at least 95% dissolved after 10 minutes in a solution of 0.1N HC1 at 37±0.5 °C in an Apparatus-II (Paddles) with a paddle speed of about 50 rpm. In some embodiments, the tablet is tested was prepared within a week of the dissolution test. In some embodiments, the tablet is tested was prepared at least a month before performing the dissolution test. In some embodiments, the tablet is tested was prepared at least a three months before performing the dissolution test. In some
embodiments, the tablet is tested was prepared at least six months before performing the dissolution test. In some embodiments, the tablet was incubated for one month at 25°C with 60% relative humidity (RH) before performing the dissolution test. In some embodiments, the tablet was incubated for two months at 25°C with 60% relative humidity (RH) before performing the dissolution test. In some embodiments, the tablet was incubated for three months at 25°C with 60% relative humidity (RH) before performing the dissolution test. In some embodiments, the tablet was incubated for one month at 40°C with 75% relative humidity (RH) before performing the dissolution test. In some embodiments, the tablet was incubated for three months at 40°C with 75% relative humidity (RH) before performing the dissolution test. In some embodiments, the tablet was incubated for six months at 40°C with 75% relative humidity (RH) before performing the dissolution test. IV. Examples
[0060] The following examples are offered to illustrate, but not to limit, the claimed invention.
Example 1: Capsule & Tablet Evaluation, Capsules provide inconsistent oral pharmacokinetic data
[0061] Pharmacokinetics of AG10 were determined when administered once daily to dogs via oral gavage at 20, 60, and 200 mg/kg for 3 days (Study No. 1). Each group consisted of two animals/sex/group. Blood samples were collected from each animal on Day 1 at pre dose, 0.25, 0.5, 1, 2, 4, 8, 12 and 24 hours post-dose, Day 3 at pre-dose, 0.25, 0.5, 1, 2, 4, 8, 12, 24, 48 and 72 hr post-dose. Plasma samples were assayed for AG10 by LC-MS/MS. In general, no sex differences in AG10 mean Cmax and AUC0-24 values were observed; therefore, results for the 20 mg/kg dose group are presented as combined sex values in Table 1 below.
[0062] The pharmacokinetics of AG10 were also determined following oral administration in non-naive male and female beagle dogs (Study No. 2). The study design included three treatment groups (n=2/sex/group). Groups 1 and 2 received 5 mg/kg and 20 mg/kg AG10 in 0.5% methylcellulose (MC) formulations respectively. Group 3 animals received 20 mg/kg AG10 in gelatin capsule form. Blood samples were collected at pre-dose and approximately 2, 4, 6, 8, 12, and 24 hours post-dose. Plasma samples were assayed for AG10 by LC- MS/MS. Plasma exposures (AUC0-24) of AG10 in dogs administered 20 mg/kg AG10 as a suspension in 0.5% methylcellulose were similar to those obtained in Study No. 1 (Table 1). Plasma exposures of AG10 were also similar in dogs which were administered the same dose of AG10 either as a suspension in 0.5% methylcellulose or as a gelatin capsule without any excipients.
[0063] AG10 was administered orally to 4 male beagle dogs each as a 50 mg tablet, 200 mg tablet, and a 200 mg capsule (No. 3). Blood samples were collected at pre-dose and approximately 0.25, 0.5, 1, 2, 4, 8, 24, 48, 72 and 96 hours post-dose. Plasma samples were assayed for AG10 by LC-MS/MS. The Cmax and AUCo-inf values for dogs dosed with 200 mg tablets and 200 mg capsules were not significantly different (P < 0.05) as determined by an unpaired t-test (P values of 0.0788 and 0.0995 for Cmax and AUCo-inf respectively).
Table 1: Comparison of the Pharmacokinetics of Various Formulations of AG10
Administered by Oral Gavage to Dogs
Figure imgf000015_0001
[0064] In dog studies comparing methylcellulose formulations of AG10, time to maximal concentration (Tmax) was 0.44 ± 0.38 hr in study No. 1 and 2.5 ± 1 hr in study No. 2. The capsule in study No. 2 was formulated without excipients and showed lower variability (Tmax = 2 ± 0 hr) than the methylcellulose comparator. In study No. 3, even though the maximal exposure of AG10 tablets was comparable to that of the capsule, animal to animal variability in absorption of AG10 was greater in the four animals orally dosed with capsule containing excipients. For the 50 mg tablets, time to maximal concentration (Tmax) was 0.500 ± 0 hr, for the 200 mg tablets, Tmax was at 1.00 ± 0 hr. For 200 mg capsules with excipient, Tmax was more variable at 1.38 ± 0.750 hr. Thus, in the head to head comparison, tablets produced more consistent oral absorption of AG10.
Example 2: High-Load Immediate Release Tablet Formulations of AGIO
[0065] The following Example describes the successful preparation of tablet formulations containing high amounts of AG10. [0066] Three tablet formulations containing differing amounts of AG10 were prepared.
Table 2 provides information on the relative amounts of components used in each formulation.
Table 2: High-Load AGIO Tablet Formulations
Figure imgf000015_0002
Figure imgf000016_0004
[0067] After compression, the tablets were film coated with OpadryQX white.
[0068] Tablets were prepared using the general diagram provided in FIG. 1. Table 3 below, provides an exemplary amounts used in formulating a tablet formulation with 66.7% AG10 HC1 (Batch 2), and Table 4 and Table 5 provide a list of equipment used and a summary of the steps performed to prepare the tablet formulation. Similar processes were performed when preparing Batch 1 and Batch 3, referenced in Table 2.
Table 3: High-Load AGIO Tablet Formulations
Figure imgf000016_0005
Table 4: Equipment Used
Items Used
blender Sterile
qt tote Bosch
le thief
Figure imgf000016_0001
esh siev
Figure imgf000016_0003
Tablet
Figure imgf000016_0002
Figure imgf000017_0001
Table 5: Summary of Procedure
Figure imgf000017_0002
[0069] A picture of a 50% (w/w) and a 66.7 (w/w) AG10 HC1 coated tablets are shown in FIG. 2. The 50 % tablet was compressed with 8 x 17.5 mm capsule shaped tooling, while 66.7 % tablet was compressed at 7.5 x 15 mm capsule tooling. Physical properties of the two displayed tablets are summarized in Table 6. Table 6: Summary of Physical Properties
Figure imgf000018_0001
[0070] Measurement of friability : Friability of the tablets, as reported in Table 6, was evaluated according to USP method <1216> from the percentage weight loss of NLT 6.5g of tablets tumbled in a friabilator (model EF-2, Electrolab) for 100 rounds at 25 rpm. The tablets were dedusted, and the loss in weight caused by fracture or abrasion was recorded as the percentage weight loss. Friability below 1% is considered acceptable.
[0071] Next, dissolution profiles of a 50% (w/w) and a 66.7 (w/w) AG10 HC1 coated tablets were determined. The dissolution experiments were performed by placing a tablet formulation in a solution of 0.1N HC1 described in Table 7. Table 7: Dissolution Parameters
Figure imgf000018_0002
[0072] FIG. 3 shows the dissolution profiles of a 50% (w/w) and a 66.7 (w/w) AG10 HC1 coated tablets. The dissolution experiment was performed with no significant incubation time after the tablet was formulated. As seen in FIG. 3 dissolution reached 100% released within 10 minutes for both tablets.
Example 3: Tablet Formulations Exceeding 33.3% AGIO Exhibited Tablet Erosion During Friability Test
[0073] The following example describes tablet formulations of AG10 where 33.3% drug loading could not be exceeded without experiencing tablet erosion during friability test.
[0074] Formulations of AG10 were prepared as generally outlined in FIG. 4 and FIG. 5. The amount of AG10 and other components are area described in Table 8. Table 8: AGIO Tablet Formulations Prepared
Figure imgf000019_0001
[0075] Each of the above referenced formulations were prepared as 200 mg tablets and underwent a friability test as described in Example 2. Tablets from L018A and L018B prepared at 33.0% drug load (and compressed at high and middle hardness kP values, respectively) were resistant to crumbling, presenting only minor (if any) tablet edge erosion after friability test. See, FIG. 6. Comparatively, L016 and L017, prepared at 40% drug load and compressed at the maximum hardness that could be reached, presented major tablet edge erosion after friability test. See, FIG. 7.
[0076] The formulations discussed above used a standard grade of microcrystalbne cellulose, and the resulting tablets with greater than 33.0% AG10 had friability issues that compromise their clinical use. Comparatively, the formulations of Example 2 used high grade microcrystalbne cellulose, and reliably provided tablets that had favorable physical properties and were not susceptible to crumbling.
Example 4:“Accelerated Stability Condition” Dissolution Test Demonstrates High- Load AGIO Tablet Formulation Stability
[0077] The following Example describes the preparation and subsequent dissolution tests of immediate release tablet formulations containing 33% AG10 HC1 (200 mg) with standard microcrystalbne cellulose and tablet formulations containing 66.7% AG10 HC1 (400 mg) with a high grade microcrystalbne cellulose. [0078] The formulations for each of the tablets are shown in Table 9 and Table 10, respectively.
Table 9. Quantitative Composition of 33% AGIO HC1 Tablets
Figure imgf000020_0001
1 Actual amount of AGIO hydrochloride is adjusted based on drug substance potency and corresponds to 177.82 mg of AGIO free base. The actual amount of silicified microcrystalline cellulose is based on a concomitant reduction such that the target core weight remains 606 mg.
b Prosolv HD 90
c Pearlitol 100SD
4 Solutab type A
' Plasdone S-630
f Ligamed MF-2-V
E Purified water is used in the film coating process and is removed during processing
h Represents 3% weight gain on the tablet core weight. Opadry White, Colorcon 33G28707 contains Hypromellose (Ph. Eur.), titanium dioxide (Ph. Eur.), and triacetin (Ph. Eur.).
Table 10. Quantitative Composition of 66.7% AGIO HC1 Tablets
Figure imgf000020_0005
Intragranular
Figure imgf000020_0002
Extragranular
Figure imgf000020_0003
Film Coat
Figure imgf000020_0004
1 Actual amount of AGIO hydrochloride is adjusted based on drug substance potency and corresponds to 355.64 mg of AGIO free base.
The actual amount of microcrystalline cellulose is based on a concomitant reduction such that the target core weight remains 600 mg b Ceolus UF-711 or equivalent
c Ac-Di-Sol SD711 or equivalent
4 Syloid 244 EP or equivalent
' Hyqual 5712, Ligamed MF-2-K, or equivalent
f Purified water is used in the film coating process and is removed during processing
E Represents 4% weight gain on the tablet core weight. Opadry QX White, Colorcon 321A180025 contains GMCC type 1/mono- and diglycerides, polyethylene glycol polyvinyl alcohol graft copolymer, polyvinyl alcohol (partially hydrolyzed), talc, and titanium dioxide. [0079] The manufacturing process for the two tablet formulations is shown in FIG. 8 and FIG. 9
[0080] 33% AG10 HC1 tablets were bottled and placed under accelerated storage (40°C/75 % relative humidity (RH)) conditions. FIG. 10 shows that storage under accelerated storage conditions significantly reduced the dissolution rate of 33% AG10 HC1 tablets.
[0081] 66.7% AG10 HC1 tablets were also bottled and placed under accelerated storage conditions. FIG. 11 shows that 400 mg AG10 HC1 tablets did not show a reduction in dissolution rate after storage for 6 months. [0082] Both formulations of AG10 HC1 tablets were evaluated using the same dissolution method (USP 2 Apparatus (Paddles), 900 mL 0.1 N HC1, 75 RPM, 37°C) for release and for stability studies. The 66.7% AG10 HC1 tablet formulation was superior relative to the 33% AG10 HC1 tablet formulation in terms of dissolution rate after storage, indicating that the 66.7% AG10 HC1 tablet has improved storage capacity. [0083] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, one of skill in the art will appreciate that certain changes and modifications may be practiced within the scope of the appended claims. In addition, each reference provided herein is incorporated by reference in its entirety to the same extent as if each reference was individually incorporated by reference. Where a conflict exists between the instant application and a reference provided herein, the instant application shall dominate.

Claims

WHAT IS CLAIMED IS:
1. A tablet formulation comprising AG10 or a pharmaceutically acceptable salt thereof and at least one pharmaceutical excipient selected from one or more fillers, one or more binders, one or more disintegrants, and one or more lubricants, wherein said tablet comprises at least 40% or more by weight of AG10 or a pharmaceutically acceptable salt thereof.
2. The tablet formulation of claim 1, comprising about 40 to 85% by weight of AG10 or a pharmaceutically acceptable salt thereof.
3. The tablet formulation of claim 1, comprising about 50 to 75% by weight of AG10 or a pharmaceutically acceptable salt thereof.
4. The tablet formulation of claim 1, comprising about 50% by weight of AG10 or a pharmaceutically acceptable salt thereof.
5. The tablet formulation of claim 1, comprising about 66.7% by weight of AG10 or a pharmaceutically acceptable salt thereof.
6. The tablet formulation of claim 1, comprising about 75% by weight of AG10 or a pharmaceutically acceptable salt thereof.
7. The tablet formulation of any one of claims 1 to 6, comprising about 1 to 60% by weight of one or more fillers.
8 . The tablet formulation of claim 7, wherein said one or more fillers comprises about 5 to 55% by weight of said tablet formulation.
9 . The tablet formulation of claim 7, wherein said one or more fillers comprises about 10 to 50% by weight of said tablet formulation.
10. The tablet formulation of claim 7, wherein said one or more fillers comprises about 15 to 45% by weight of said tablet formulation.
11. The tablet formulation of any one of claims 1 to 10, wherein said tablet comprises a high grade microcrystalline cellulose as a filler component.
12. The tablet formulation of claim 11, wherein said high grade microcrystalline cellulose is characterized by cellulose polymers with spherical morphology and porous structure.
13. The tablet formulation of claim 12, wherein said high grade microcrystalline cellulose is selected from the group consisting of UF-702 and UF-711.
14. The tablet formulation of claim 11, wherein said high grade microcrystalline cellulose is characterized by cellulose polymers with needle-like particle shape.
15. The tablet formulation of claim 14, wherein said high grade microcrystalline cellulose is selected from the group consisting of KG-802 and KG- 1000.
16. The tablet formulation of any one of claims 1 to 10, wherein said one or more fillers are selected from a cellulose derivative and an inorganic salt.
17. The tablet formulation of any one of claims 1 to 10, wherein said one or more fillers are microcrystalline cellulose and silicon dioxide.
18. The tablet formulation of any one of claims 1 to 17, comprising about 1 to about 15% by weight of one or more disintegrants.
19. The tablet formulation of claim 18, wherein said one or more disintegrants comprises about 3 to 8% by weight of said tablet formulation.
20. The tablet formulation of claim 18, wherein said one or more disintegrants comprises about 6% by weight of said tablet formulation.
21. The tablet formulation of any one of claims 18 to 20, wherein said one or more disintegrants is croscarmellose sodium.
22. The tablet formulation of any one of claims 1 to 21, comprising about 0.1 to 8% by weight of a lubricant.
23. The tablet formulation of claim 22, wherein said one or more lubricants comprises about 1.5% by weight of said tablet formulation.
24. The tablet formulation of claim 22 or claim 23, wherein said one or more lubricants is magnesium stearate.
25. The tablet formulation of any one of claims 1 to 24, wherein said tablet is at least 75% dissolved after 10 minutes in a solution of 0.1N HC1 at 37±0.5 °C in an Apparatus-II (Paddles) with a paddle speed of about 50 rpm.
26. The tablet formulation of any one of claims 1 to 24, wherein said tablet is at least 85% dissolved after 10 minutes in a solution of 0.1N HC1 at 37±0.5 °C in an Apparatus-II (Paddles) with a paddle speed of about 50 rpm.
27. The tablet formulation of any one of claims 1 to 24, wherein said tablet is at least 95% dissolved after 10 minutes in a solution of 0.1N HC1 at 37±0.5 °C in an Apparatus-II (Paddles) with a paddle speed of about 50 rpm.
28. The tablet formulation of any one of claims 25 to 27, wherein said tablet was prepared at least three months before performing the dissolution test.
29. The tablet formulation of any one of claims 1 to 28, further comprising a coating agent.
30. The tablet formulation of claim 29, wherein said coating agent is Opadry QX 321A180025.
31. The tablet formulation of any one of claims 1 to 30, wherein AG10 is the pharmaceutically acceptable salt form of Formula la
Figure imgf000024_0001
PCT/US2019/046789 2018-08-17 2019-08-16 Formulations of ag10 WO2020037189A1 (en)

Priority Applications (11)

Application Number Priority Date Filing Date Title
KR1020217007760A KR20210046708A (en) 2018-08-17 2019-08-16 Formulation of AG10
EP19849948.5A EP3836920A4 (en) 2018-08-17 2019-08-16 Formulations of ag10
JP2021507989A JP7469293B2 (en) 2018-08-17 2019-08-16 Preparation of AG10
CN201980054507.6A CN112804998A (en) 2018-08-17 2019-08-16 Formulation of AG10
MX2021001761A MX2021001761A (en) 2018-08-17 2019-08-16 Formulations of ag10.
AU2019321583A AU2019321583A1 (en) 2018-08-17 2019-08-16 Formulations of AG10
SG11202101393PA SG11202101393PA (en) 2018-08-17 2019-08-16 Formulations of ag10
BR112020026493-8A BR112020026493A2 (en) 2018-08-17 2019-08-16 AG10 FORMULATIONS
EA202190561A EA202190561A1 (en) 2018-08-17 2019-08-16 AG10 PREPARATIONS
CA3104695A CA3104695A1 (en) 2018-08-17 2019-08-16 Formulations of ag10
IL280906A IL280906A (en) 2018-08-17 2021-02-16 Formulations of ag10

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862765154P 2018-08-17 2018-08-17
US62/765,154 2018-08-17

Publications (1)

Publication Number Publication Date
WO2020037189A1 true WO2020037189A1 (en) 2020-02-20

Family

ID=69524257

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2019/046789 WO2020037189A1 (en) 2018-08-17 2019-08-16 Formulations of ag10

Country Status (15)

Country Link
US (2) US11260047B2 (en)
EP (1) EP3836920A4 (en)
JP (1) JP7469293B2 (en)
KR (1) KR20210046708A (en)
CN (1) CN112804998A (en)
AU (1) AU2019321583A1 (en)
BR (1) BR112020026493A2 (en)
CA (1) CA3104695A1 (en)
CL (1) CL2021000289A1 (en)
EA (1) EA202190561A1 (en)
IL (1) IL280906A (en)
MA (1) MA53238A (en)
MX (1) MX2021001761A (en)
SG (1) SG11202101393PA (en)
WO (1) WO2020037189A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023052652A1 (en) 2021-10-01 2023-04-06 Sandoz Ag Crystalline form of acoramidis hydrochloride

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EA201991921A1 (en) 2017-02-17 2020-02-06 Эйдос Терапьютикс, Инк. METHODS FOR PRODUCING AG-10, ITS INTERMEDIATE COMPOUNDS AND THEIR SALTS

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130095181A1 (en) 2010-04-07 2013-04-18 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions of 3-(6-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl) cyclopropanecarboxamido)-3-methylpyriodin-2-yl)benzoic acid and administration thereof
WO2014100227A1 (en) 2012-12-21 2014-06-26 The Board Of Trustees Of The Leland Stanford Junior University Transthyretin stabilizers and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
WO2018071678A1 (en) 2016-10-12 2018-04-19 Global Blood Therapeutics, Inc. Tablets comprising 2-hydroxy-6-((2-(1-isopropyl-1h-pyrazol-5-yl)pyridin-3-yl)methoxy)benzaldehyde

Family Cites Families (73)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
USRE28819E (en) 1972-12-08 1976-05-18 Syntex (U.S.A.) Inc. Dialkylated glycol compositions and medicament preparations containing same
US4255329A (en) 1973-10-02 1981-03-10 Syva Company Double receptor fluorescent immunoassay
US4117149A (en) 1975-09-12 1978-09-26 Pfizer Inc. 4-oxo-4h-benzopyrans as animal growth promotants
US4171365A (en) 1977-08-05 1979-10-16 Sterling Drug Inc. Antiviral aryloxyalkylpyrazoles
US4261928A (en) 1977-08-05 1981-04-14 Sterling Drug Inc. 2-Benzoyl-8-(2-chloro-4-methoxyphenoxy)-1-phenyl-1-octanone
US4232161A (en) 1979-10-24 1980-11-04 Sterling Drug Inc. 4-[6-(2-Chloro-4-methoxyphenoxy)hexyl]-3,5-diethyl-1-(2-pyridinyl)-1H-pyrazole
US4234725A (en) 1979-10-24 1980-11-18 Sterling Drug Inc. 4-[6-(2-Chloro-4-methoxyphenoxy)hexyl]-3,5-diethyl-1-[4-(4-morpholinyl)-1-oxobutyl]-1H-pyrazole
US4410545A (en) 1981-02-13 1983-10-18 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4328245A (en) 1981-02-13 1982-05-04 Syntex (U.S.A.) Inc. Carbonate diester solutions of PGE-type compounds
US4358603A (en) 1981-04-16 1982-11-09 Syntex (U.S.A.) Inc. Acetal stabilized prostaglandin compositions
US4668640A (en) 1981-12-11 1987-05-26 Abbott Laboratories Fluorescence polarization immunoassay utilizing substituted carboxyfluoresceins
US4409239A (en) 1982-01-21 1983-10-11 Syntex (U.S.A.) Inc. Propylene glycol diester solutions of PGE-type compounds
GB2120242A (en) 1982-04-30 1983-11-30 Erba Farmitalia Ergoline derivatives
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US5585112A (en) 1989-12-22 1996-12-17 Imarx Pharmaceutical Corp. Method of preparing gas and gaseous precursor-filled microspheres
IT1246382B (en) 1990-04-17 1994-11-18 Eurand Int METHOD FOR THE TARGETED AND CONTROLLED DELIVERY OF DRUGS IN THE INTESTINE AND PARTICULARLY IN THE COLON
US5543390A (en) 1990-11-01 1996-08-06 State Of Oregon, Acting By And Through The Oregon State Board Of Higher Education, Acting For And On Behalf Of The Oregon Health Sciences University Covalent microparticle-drug conjugates for biological targeting
US6010715A (en) 1992-04-01 2000-01-04 Bertek, Inc. Transdermal patch incorporating a polymer film incorporated with an active agent
US6024975A (en) 1992-04-08 2000-02-15 Americare International Diagnostics, Inc. Method of transdermally administering high molecular weight drugs with a polymer skin enhancer
US5315015A (en) 1992-11-10 1994-05-24 Hoffmann-La Roche Inc. Compounds having improved fluorescence in fluorescence polarization immunoassays and immunoassays utilizing same
US6274552B1 (en) 1993-03-18 2001-08-14 Cytimmune Sciences, Inc. Composition and method for delivery of biologically-active factors
AU668818B2 (en) 1993-04-07 1996-05-16 Taiho Pharmaceutical Co., Ltd. Thiazolidine derivative and pharmaceutical composition containing the same
US5523092A (en) 1993-04-14 1996-06-04 Emory University Device for local drug delivery and methods for using the same
US5985307A (en) 1993-04-14 1999-11-16 Emory University Device and method for non-occlusive localized drug delivery
US6004534A (en) 1993-07-23 1999-12-21 Massachusetts Institute Of Technology Targeted polymerized liposomes for improved drug delivery
US5744368A (en) 1993-11-04 1998-04-28 Research Foundation Of State University Of New York Methods for the detection of soluble amyloid β-protein (βAP) or soluble transthyretin (TTR)
US5759542A (en) 1994-08-05 1998-06-02 New England Deaconess Hospital Corporation Compositions and methods for the delivery of drugs by platelets for the treatment of cardiovascular and other diseases
US5660854A (en) 1994-11-28 1997-08-26 Haynes; Duncan H Drug releasing surgical implant or dressing material
US5983134A (en) 1995-04-23 1999-11-09 Electromagnetic Bracing Systems Inc. Electrophoretic cuff apparatus drug delivery system
US6316652B1 (en) 1995-06-06 2001-11-13 Kosta Steliou Drug mitochondrial targeting agents
US6167301A (en) 1995-08-29 2000-12-26 Flower; Ronald J. Iontophoretic drug delivery device having high-efficiency DC-to-DC energy conversion circuit
US6039975A (en) 1995-10-17 2000-03-21 Hoffman-La Roche Inc. Colon targeted delivery system
TW345603B (en) 1996-05-29 1998-11-21 Gmundner Fertigteile Gmbh A noise control device for tracks
US5985317A (en) 1996-09-06 1999-11-16 Theratech, Inc. Pressure sensitive adhesive matrix patches for transdermal delivery of salts of pharmaceutical agents
WO1998014179A1 (en) 1996-10-01 1998-04-09 Cima Labs Inc. Taste-masked microcapsule compositions and methods of manufacture
US6131570A (en) 1998-06-30 2000-10-17 Aradigm Corporation Temperature controlling device for aerosol drug delivery
US5860957A (en) 1997-02-07 1999-01-19 Sarcos, Inc. Multipathway electronically-controlled drug delivery system
US6120751A (en) 1997-03-21 2000-09-19 Imarx Pharmaceutical Corp. Charged lipids and uses for the same
US6060082A (en) 1997-04-18 2000-05-09 Massachusetts Institute Of Technology Polymerized liposomes targeted to M cells and useful for oral or mucosal drug delivery
US5948433A (en) 1997-08-21 1999-09-07 Bertek, Inc. Transdermal patch
DE69839179T2 (en) 1997-10-28 2009-02-19 Bando Chemical Industries, Ltd., Kobe DERMATOLOGICAL PLASTER AND METHOD FOR PRODUCING ITS BASE LAYER
US6048736A (en) 1998-04-29 2000-04-11 Kosak; Kenneth M. Cyclodextrin polymers for carrying and releasing drugs
US6271359B1 (en) 1999-04-14 2001-08-07 Musc Foundation For Research Development Tissue-specific and pathogen-specific toxic agents and ribozymes
US6256533B1 (en) 1999-06-09 2001-07-03 The Procter & Gamble Company Apparatus and method for using an intracutaneous microneedle array
US6261595B1 (en) 2000-02-29 2001-07-17 Zars, Inc. Transdermal drug patch with attached pocket for controlled heating device
AU2002352706A1 (en) 2001-11-15 2003-06-10 Maxia Pharmaceuticals, Inc. N-substituted heterocycles for the treatment of hypercholesteremia, dyslipidemia and other metabolic disorders, cancer, and other diseases
JP4568603B2 (en) 2002-05-31 2010-10-27 プロテオテック・インコーポレイテッド Compounds, compositions, and methods for treating amyloid disease and synucleinopathies (eg, Alzheimer's disease, type 2 diabetes, and Parkinson's disease)
WO2004096808A1 (en) 2003-04-28 2004-11-11 De Novo Pharmaceuticals Limited Triazine compounds and their use
ATE493973T1 (en) 2004-06-04 2011-01-15 Teva Pharma PHARMACEUTICAL COMPOSITION CONTAINING IRBESARTAN
WO2006009826A1 (en) 2004-06-18 2006-01-26 3M Innovative Properties Company Aryloxy and arylalkyleneoxy substituted thiazoloquinolines and thiazolonaphthyridines
JP2007284350A (en) 2004-07-27 2007-11-01 Takeda Chem Ind Ltd Therapeutic agent for diabetes
WO2006088694A1 (en) 2005-02-14 2006-08-24 Wyeth SUBSTITUTED THIENYL AND FURYL ACYLGUANIDINES AS β-SECRETASE MODULATORS
JP2009501801A (en) 2005-07-18 2009-01-22 ホライゾン セラピューティクス, インコーポレイテッド Medicament containing ibuprofen and famotidine and its administration
BRPI0718874A2 (en) 2006-12-22 2015-06-23 Novartis Ag Organic compounds
KR101119294B1 (en) 2007-05-11 2012-04-12 일라이 릴리 앤드 캄파니 2-[4-pyrazol-4-ylalkylpiperazin-1-yl]-3-phenyl pyrazines and pyridines and 3-[4-pyrazol-4-ylalkylpiperazin-1-yl]-2-phenyl pyridines as 5-ht7 receptor antagonists
CN101687790B (en) 2007-05-25 2015-02-11 Abbvie公司 Heterocyclic compounds as positive modulators of metabotropic glutamate receptor 2 (mglu2 receptor)
PL2061766T3 (en) 2007-06-06 2011-02-28 Torrent Pharmaceuticals Ltd Novel compounds
BRPI0811065A2 (en) 2007-06-08 2014-12-02 Abbott Lab 5-HETEROARYL INDAZOES REPLACED AS KINASE INHIBITORS
EP2219646A4 (en) 2007-12-21 2010-12-22 Univ Rochester Method for altering the lifespan of eukaryotic organisms
WO2009118305A1 (en) 2008-03-26 2009-10-01 Novartis Ag Hydroxamate-based inhibitors of deacetylases b
JO3041B1 (en) 2008-07-25 2016-09-05 Galapagos Nv Novel compounds useful for the treatment of degenerative and inflammatory diseases
US7994171B2 (en) 2008-09-11 2011-08-09 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
US8048887B2 (en) 2008-09-11 2011-11-01 Bristol-Myers Squibb Company Compounds for the treatment of hepatitis C
JP5743897B2 (en) 2008-11-20 2015-07-01 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニーGlaxoSmithKline LLC Compound
EP2488028B1 (en) 2009-10-14 2020-08-19 Merck Sharp & Dohme Corp. Substituted piperidines that increase p53 activity and the uses thereof
AR079022A1 (en) 2009-11-02 2011-12-21 Sanofi Aventis DERIVATIVES OF CYCLIC CARBOXYL ACID SUBSTITUTED WITH ACILAMINE, ITS USE AS PHARMACEUTICAL PRODUCTS, PHARMACEUTICAL COMPOSITION AND PREPARATION METHOD
WO2011140333A1 (en) 2010-05-07 2011-11-10 The Board Of Trustees Of The Leland Stanford Junior University Identification of stabilizers of multimeric proteins
BR112013015260B1 (en) 2010-12-16 2020-03-24 Allergan, Inc. SULFUR DERIVATIVES AS CHEMOCCIN RECEPTOR MODULATORS, COMPOSITION UNDERSTANDING THE SAME AND ITS USE
WO2016025129A1 (en) 2014-08-14 2016-02-18 Alhamadsheh Mamoun M Conjugation of pharmaceutically active agents with transthyretin ligands through adjustable linkers to increase serum half-life
PT3607939T (en) 2015-06-30 2022-09-12 Gilead Sciences Inc Pharmaceutical formulations
BR112018000041A2 (en) 2015-07-31 2018-09-04 Pfizer Inc. 1,1,1-TRIFLUORO-3-HYDROXYPROPAN-2-ILA CARBAMATE DERIVATIVES AND 1,1,1-TRIFLUORO-4-HYDROXYBUTAN-2-ILA CARBAMATE DERIVATIVES AS MAGL INHIBITORS
EA201991921A1 (en) 2017-02-17 2020-02-06 Эйдос Терапьютикс, Инк. METHODS FOR PRODUCING AG-10, ITS INTERMEDIATE COMPOUNDS AND THEIR SALTS
US11058668B2 (en) 2018-03-23 2021-07-13 Eidos Therapeutics, Inc. Methods of treating TTR amyloidosis using AG10

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130095181A1 (en) 2010-04-07 2013-04-18 Vertex Pharmaceuticals Incorporated Pharmaceutical compositions of 3-(6-(1-(2,2-difluorobenzo[d][1,3]dioxol-5-yl) cyclopropanecarboxamido)-3-methylpyriodin-2-yl)benzoic acid and administration thereof
WO2014100227A1 (en) 2012-12-21 2014-06-26 The Board Of Trustees Of The Leland Stanford Junior University Transthyretin stabilizers and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
US9913826B2 (en) 2012-12-21 2018-03-13 The Board Of Trustees Of The Leland Stanford Junior University Compounds and compositions that bind and stabilize transthyretin and their use for inhibiting transthyretin amyloidosis and protein-protein interactions
WO2018071678A1 (en) 2016-10-12 2018-04-19 Global Blood Therapeutics, Inc. Tablets comprising 2-hydroxy-6-((2-(1-isopropyl-1h-pyrazol-5-yl)pyridin-3-yl)methoxy)benzaldehyde

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"Handbook of Pharmaceutical Excipients", 1976
"Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING COMPANY
BANKER, GILBERTRHODESCHRISTOPHER T: "Modern Pharmaceutics", 1979
LIEBERMAN, HERBERT A ET AL.: "Pharmaceutical Dosage Forms: Tablets", vol. 1, 1989
See also references of EP3836920A4

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023052652A1 (en) 2021-10-01 2023-04-06 Sandoz Ag Crystalline form of acoramidis hydrochloride

Also Published As

Publication number Publication date
US11260047B2 (en) 2022-03-01
EP3836920A4 (en) 2022-04-13
BR112020026493A2 (en) 2021-03-23
TW202021583A (en) 2020-06-16
CL2021000289A1 (en) 2021-08-27
US20220313661A1 (en) 2022-10-06
US20200054607A1 (en) 2020-02-20
EP3836920A1 (en) 2021-06-23
CA3104695A1 (en) 2020-02-20
JP7469293B2 (en) 2024-04-16
JP2021534189A (en) 2021-12-09
MX2021001761A (en) 2021-04-19
AU2019321583A1 (en) 2021-01-21
CN112804998A (en) 2021-05-14
EA202190561A1 (en) 2021-05-26
MA53238A (en) 2022-04-13
KR20210046708A (en) 2021-04-28
IL280906A (en) 2021-04-29
SG11202101393PA (en) 2021-03-30

Similar Documents

Publication Publication Date Title
US8221792B2 (en) Sustained release pharmaceutical compositions for highly water soluble drugs
US20110218216A1 (en) Extended release pharmaceutical composition of donepezil
WO2008064202A2 (en) Modified-release formulations of calcium receptor-active compounds
JP2011516613A (en) An oral pharmaceutical composition in a solid dispersion, preferably comprising posaconazole and HPMCAS
US20220313661A1 (en) Formulations of ag10
US9387166B2 (en) Controlled release oral dosage form comprising oxycodone
US20180000792A1 (en) Modified release compositions of epalrestat or a derivative thereof and methods for using the same
US20130143897A1 (en) Oral controlled release pharmaceutical compositions of blonanserin
US20100172984A1 (en) tablet dosage form comprising cetirizine and pseudoephedrine
JP2014504633A (en) Controlled release pharmaceutical composition for oral administration
WO2021074808A1 (en) Pharmaceutical composition comprising sacubitril and valsartan and process for preparation thereof
JP2023071921A (en) Lenalidomide oral tablet composition in various doses
WO2018211336A2 (en) Solid dosage form containing sorafenib tosylate
TWI834708B (en) Formulations of ag10
US20220226246A1 (en) Pharmaceutical composition comprising axitinib
US20050059719A1 (en) Solid dosage formulation containing a Factor Xa inhibitor and method
US20080081069A1 (en) Novel controlled release formulations of divalproex sodium
JP7271869B2 (en) Tablets containing levocetirizine
WO2005065662A1 (en) Solid dosage formulations of galantamine
KR102330953B1 (en) Pharmaceutical dosage forms containing sodium-1-[6-(morpholin-4-yl)pyrimidin-4-yl]-4-(1h-1,2,3-triazol-1-yl)-1h-pyrazol-5-olate
EP4205730A1 (en) Pharmaceutical composition of single dosage form for treating or preventing hypertension and hyperlipidemia
US20230073216A1 (en) Pharmaceutical Compositions of Raltegravir
JP2019510071A (en) Tadalafil-containing and tamsulosin-containing capsule composites with improved stability and dissolution rate
WO2024100682A1 (en) Sustained release composition of hydroxyzine and it&#39;s process
WO2024095137A1 (en) Pharmaceutical composition of empagliflozin and process thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 19849948

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3104695

Country of ref document: CA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112020026493

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2019321583

Country of ref document: AU

Date of ref document: 20190816

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2021507989

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20217007760

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019849948

Country of ref document: EP

Effective date: 20210317

ENP Entry into the national phase

Ref document number: 112020026493

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20201223