SOLID FORMS OF 2-(3,5-DICHLORO-4-((5-ISOPROPYL-6-OXO-1,6-DIHYDROPYRIDAZIN-3-YL)OXY)PHENYL)-3,5-DIOXO-2,3,4,5-TETRAHYDRO-1,2,4-TRIAZINE-6-CARBONITRILE

Information

  • Patent Application
  • 20210122740
  • Publication Number
    20210122740
  • Date Filed
    July 02, 2019
    4 years ago
  • Date Published
    April 29, 2021
    3 years ago
Abstract
The present invention is directed to morphic forms, co-crystals, salts, and amorphous solid dispersions of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile.
Description
INCORPORATION BY REFERENCE OF SEQUENCE LISTING

The contents of the text file named “MDRI-024_SEQ_LISTING.txt”, which was created on May 21, 2019 and is 3.66 KB in size, are hereby incorporated by reference in their entireties.


FIELD OF THE INVENTION

The present invention relates to morphic forms, co-crystals, salts, and amorphous solid dispersions of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile (Compound A).


BACKGROUND OF THE INVENTION

Thyroid hormones are critical for normal growth and development and for maintaining metabolic homeostasis (Paul M. Yen, Physiological reviews, Vol. 81(3): pp. 1097-1126 (2001)). Circulating levels of thyroid hormones are tightly regulated by feedback mechanisms in the hypothalamus/pituitary/thyroid (HPT) axis. Thyroid dysfunction leading to hypothyroidism or hyperthyroidism clearly demonstrates that thyroid hormones exert profound effects on cardiac function, body weight, metabolism, metabolic rate, body temperature, cholesterol, bone, muscle and behavior.


The biological activity of thyroid hormones is mediated by thyroid hormone receptors (TRs or THRs) (M. A. Lazar, Endocrine Reviews, Vol. 14: pp. 348-399 (1993)). TRs belong to the superfamily known as nuclear receptors. TRs form heterodimers with the retinoid receptor that act as ligand-inducible transcription factors. TRs have a ligand binding domain, a DNA binding domain, and an amino terminal domain, and regulate gene expression through interactions with DNA response elements and with various nuclear co-activators and co-repressors. The thyroid hormone receptors are derived from two separate genes, a and (3. These distinct gene products produce multiple forms of their respective receptors through differential RNA processing. The major thyroid receptor isoforms are α1, α2, β1, and β2. Thyroid hormone receptors al, β1, and β2 bind thyroid hormone. It has been shown that the thyroid hormone receptor subtypes can differ in their contribution to particular biological responses. Recent studies suggest that TRβ1 plays an important role in regulating TRH (thyrotropin releasing hormone) and on regulating thyroid hormone actions in the liver. TRβ2 plays an important role in the regulation of TSH (thyroid stimulating hormone) (Abel et. al., J. Clin. Invest., Vol 104: pp. 291-300 (1999)). TRβ1 plays an important role in regulating heart rate (B. Gloss et. al. Endocrinology, Vol. 142: pp. 544-550 (2001); C. Johansson et. al., Am. J. Physiol., Vol. 275: pp. R640-R646 (1998)).


Efforts have been made to synthesize thyroid hormone analogs which exhibit increased thyroid hormone receptor beta selectivity and/or tissue selective action. Such thyroid hormone mimetics may yield desirable reductions in body weight, lipids, cholesterol, and lipoproteins, with reduced impact on cardiovascular function or normal function of the hypothalamus/pituitary/thyroid axis (see, e.g., Joharapurkar et al., J. Med. Chem., 2012, 55 (12), pp 5649-5675). The development of thyroid hormone analogs which avoid the undesirable effects of hyperthyroidism and hypothyroidism while maintaining the beneficial effects of thyroid hormones would open new avenues of treatment for patients with metabolic disease such as obesity, hyperlipidemia, hypercholesterolemia, diabetes and other disorders and diseases such as liver steatosis and NASH, atherosclerosis, cardiovascular diseases, hypothyroidism, thyroid cancer, thyroid diseases, a resistance to thyroid hormone (RTH) syndrome, and related disorders and diseases.


SUMMARY OF THE INVENTION

The present disclosure provides morphic forms, co-crystals, salts, and amorphous solid dispersions of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile (Compound A).


One aspect of the present disclosure relates to a crystalline salt of Compound A. Another aspect of the present disclosure relates to a pharmaceutical composition comprising the crystalline salt disclosed herein.


In some embodiments, the crystalline salt is characterized as having a counter-ion, wherein the counter-ion is selected from L-lysine, L-arginine, 2-hydroxy-N,N,N-trimethylethan-1-aminium, diethylamine, ethanolamine, ethanol-2-diethylamine, Na+, Mg2+, K+, Ca2+, diethanolamine, triethanolamine, L-histidine, and meglumine.


In some embodiments, the counter-ion is L-lysine.


In some embodiments, the counter-ion is L-arginine.


In some embodiments, the counter-ion is 2-hydroxy-N,N,N-trimethylethan-1-aminium.


In some embodiments, the crystalline salt (L-lysine salt) is characterized by an X-ray powder diffraction pattern including peaks at about 8.70, 9.22, 11.3, 17.0, and 24.8 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).


In some embodiments, the crystalline salt has an X-ray diffraction pattern substantially similar to that set forth in FIG. 76.


In some embodiments, the crystalline salt has a melting point of about 250° C.


In some embodiments, the crystalline salt has an X-ray diffraction pattern substantially similar to that set forth in any one of FIGS. 67-70.


In some embodiments, the crystalline salt has a melting point of about 200° C.


In some embodiments, the crystalline salt has an X-ray diffraction pattern substantially similar to that set forth in FIG. 66.


In some embodiments, the crystalline salt has a melting point of about 229° C.


In some embodiments, the crystalline salt has purity of Compound A of greater than 90% by weight.


In some embodiments, the crystalline salt has purity of Compound A of greater than 95% by weight.


In some embodiments, the crystalline salt has purity of Compound A of greater than 99% by weight.


Another aspect of the present disclosure relates to a morphic form (Form B) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 5.92, 11.8, and 17.5 degrees 2θ, wherein the X-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form B has an X-ray diffraction pattern substantially similar to that set forth in FIG. 2.


Another aspect of the present disclosure relates to a morphic form (Form C) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 5.74, 11.5, 17.7, 19.3, 19.7, 21.4, 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form C has an X-ray diffraction pattern substantially similar to that set forth in FIG. 3.


Another aspect of the present disclosure relates to a morphic form (Form D) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 5.52, 8.52, 11.0, 16.5, 18.3, 21.0, 21.2, and 24.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form D has an X-ray diffraction pattern substantially similar to that set forth in FIG. 4.


Another aspect of the present disclosure relates to a morphic form (Form E) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 7.13, 10.8, 12.3, 14.1, 14.7, 15.5, 16.1, 17.5, 18.1, 19.9, 20.2, 21.0, 21.2, 22.7, 22.9, 24.4, 25.3, and 26.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form E has an X-ray diffraction pattern substantially similar to that set forth in FIG. 5.


Another aspect of the present disclosure relates to a morphic form (Form F) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.1, 10.4, 11.4, 13.9, 16.2, 16.4, 17.1, 22.0, 23.8, and 29.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form F has an X-ray diffraction pattern substantially similar to that set forth in FIG. 6.


Another aspect of the present disclosure relates to a morphic form (Form G) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 9.50, 12.9, 16.7, 17.3, 19.5, 20.2, 25.6, and 28.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form G has an X-ray diffraction pattern substantially similar to that set forth in FIG. 7.


Another aspect of the present disclosure relates to a morphic form (Form H) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 9.22, 19.8, 23.6, 25.9, and 28.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form H has an X-ray diffraction pattern substantially similar to that set forth in FIG. 8.


Another aspect of the present disclosure relates to a morphic form (Form I) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 5.77, 9.30, 10.2, 11.6, and 21.9 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form I has an X-ray diffraction pattern substantially similar to that set forth in FIG. 9.


Another aspect of the present disclosure relates to a morphic form (Form K) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 8.42, 11.4, 14.5, 18.9, 21.1, and 21.6 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form K has an X-ray diffraction pattern substantially similar to that set forth in FIG. 10.


Another aspect of the present disclosure relates to a morphic form (Form L) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.5, 11.5, 11.9, 15.2, 15.7, 16.0, 16.9, 17.1, 18.4, 18.7, 22.0, 22.8, 23.5, and 26.4 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form L has an X-ray diffraction pattern substantially similar to that set forth in FIG. 11.


Another aspect of the present disclosure relates to a morphic form (Form S+T) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 7.42, 10.5, 11.3, 12.4, 14.3, 15.8, 16.8, 17.7, 18.1, 18.4, 20.1, 20.5, 21.1, 21.9, 23.2, 25.5, 26.9, and 28.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form S+T has an X-ray diffraction pattern substantially similar to that set forth in FIG. 12.


Another aspect of the present disclosure relates to a morphic form (Form S) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.5, 12.3, 14.4, 15.8, 16.7, 17.7, 18.1, 18.4, 20.1, 20.6, 21.2, 21.9, 23.3, 24.4, 25.5, and 27.8 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form S has an X-ray diffraction pattern substantially similar to that set forth in FIG. 13.


Another aspect of the present disclosure relates to a morphic form (Form U) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 5.79, 8.43, 11.4, 11.6, 14.5, 18.9, 21.1, and 21.6 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form U has an X-ray diffraction pattern substantially similar to that set forth in FIG. 14.


Another aspect of the present disclosure relates to a morphic form (Form V) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 6.35, 10.6, 15.6, 16.5, 16.8, and 18.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form V has an X-ray diffraction pattern substantially similar to that set forth in FIG. 15.


Another aspect of the present disclosure relates to a morphic form (Form W) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.1, 10.4, 10.7, 11.7, 13.9, 24.4, and 29.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form W has an X-ray diffraction pattern substantially similar to that set forth in FIG. 16.


Another aspect of the present disclosure relates to a morphic form (Form X) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 9.66, 10.2, 10.5, 11.2, 18.7, and 24.7 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form X has an X-ray diffraction pattern substantially similar to that set forth in FIG. 17.


Another aspect of the present disclosure relates to a morphic form (Form Y) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 6.51, 13.0, 13.3, 19.5, and 24.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form Y has an X-ray diffraction pattern substantially similar to that set forth in FIG. 18.


Another aspect of the present disclosure relates to a morphic form (Form Z) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.6, 11.2, 11.6, 12.0, 14.3, 15.6, 16.2, 17.6, 18.1, 18.7, 24.1, and 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form Z has an X-ray diffraction pattern substantially similar to that set forth in FIG. 19.


Another aspect of the present disclosure relates to a morphic form (Form α) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 7.26, 10.1, 10.4, 10.6, 11.9, 13.9, 16.5, 21.9, 22.4, and 24.1 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form α has an X-ray diffraction pattern substantially similar to that set forth in FIG. 20.


Another aspect of the present disclosure relates to a morphic form (Form β) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 7.36, 10.5, 14.3, 15.7, 18.3, 20.4, 21.0, 21.8, and 23.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form β has an X-ray diffraction pattern substantially similar to that set forth in FIG. 21.


Another aspect of the present disclosure relates to a morphic form (Form χ) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 8.53, 11.2, 18.4, 20.1, and 21.4 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form χ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 22.


Another aspect of the present disclosure relates to a morphic form (Form δ) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.9, 12.1, 14.4, 18.1, 19.6, 24.5, and 27.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form δ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 23.


Another aspect of the present disclosure relates to a morphic form (Form ε) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 5.73, 11.4, 16.6, 17.6, 23.2, and 24.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form ε has an X-ray diffraction pattern substantially similar to that set forth in FIG. 24.


Another aspect of the present disclosure relates to a morphic form (Form ϕ) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 6.95, 13.9, 20.9, 22.3, and 27.9 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form ϕ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 25.


Another aspect of the present disclosure relates to a morphic form (Form η) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 6.87, 7.69, 20.5, 23.0, 23.9, and 28.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form η has an X-ray diffraction pattern substantially similar to that set forth in FIG. 26.


Another aspect of the present disclosure relates to a morphic form (Form λ) of Compound A, characterized by an X-ray powder diffraction pattern including peaks at about 10.6, 12.0, 14.3, 16.2, 17.6, 18.0, and 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, Form λ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 27.


Another aspect of the present disclosure relates to a co-crystal of Compound A and glutaric acid, characterized by an X-ray powder diffraction pattern including peaks at about 9.74, 10.8, 11.0, 12.2, 16.1, 17.0, 19.2, 21.9, and 23.1 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the co-crystal has an X-ray diffraction pattern substantially similar to that set forth in FIG. 28.


Another aspect of the present disclosure relates to an amorphous solid dispersion of Compound A, wherein the amorphous solid dispersion comprises a polymer. In some embodiments, the polymer is polyvinylpyrrolidone. In some embodiments, the weight ratio of Compound A over the polymer is about 1:2 or 1:4.


The crystalline salt, morphic form, co-crystal, or amorphous solid dispersion of the present disclosure can be used in (a) a method for treating a resistance to thyroid hormone (RTH) syndrome; (b) a method for treating non-alcoholic steatohepatitis; (c) a method for treating familial hypercholesterolemia; (d) a method for treating fatty liver disease; and (e) a method for treating dyslipidemia. In some embodiments, the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion is administered daily.


Another aspect of the disclosure relates to crystalline salt, morphic form, co-crystal, or amorphous solid dispersion of the present disclosure for the manufacture of a medicament for treating: (a) resistance to thyroid hormone (RTH) syndrome; (b) non-alcoholic steatohepatitis; (c) familial hypercholesterolemia; (d) fatty liver disease; and (e) treating dyslipidemia, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion of the present disclosure is for administration to the subject in at least one therapeutically effective amount. In some embodiments, the the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion is administered daily.


Another aspect of the disclosure relates to crystalline salt, morphic form, co-crystal, or amorphous solid dispersion of the present disclosure for use in treating: (a) resistance to thyroid hormone (RTH) syndrome; (b) non-alcoholic steatohepatitis; (c) familial hypercholesterolemia; (d) fatty liver disease; and (e) treating dyslipidemia, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion of the present disclosure is for administration to the subject in at least one therapeutically effective amount. In some embodiments, the the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion is administered daily.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is an X-ray powder diffraction (XRPD) pattern of an anhydrous crystalline form (Form A) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile (Compound A).



FIG. 2 is an XRPD pattern of a methanol solvate (Form B) of Compound A.



FIG. 3 is an XRPD pattern of an ethanol solvate (Form C) of Compound A.



FIG. 4 is an XRPD pattern of an acetone solvate (Form D) of Compound A.



FIG. 5 is an XRPD pattern of a tetrahydrofuran solvate (Form E) of Compound A.



FIG. 6 is an XRPD pattern of an ethyl acetate desolvate (Form F) of Compound A.



FIG. 7 is an XRPD pattern of a methyl isobutyl ketone (MIBK) solvate (Form G) of Compound A.



FIG. 8 is an XRPD pattern of an isopropyl acetate (IPAc) solvate (Form H) of Compound A.



FIG. 9 is an XRPD pattern of an acetic acid solvate (Form I) of Compound A.



FIG. 10 is an XRPD pattern of a dimethyl acetamide solvate (Form K) of Compound A.



FIG. 11 is an XRPD pattern of an acetonitrile solvate (Form L) of Compound A.



FIG. 12 is an XRPD pattern of a MIBK desolvate (Form S+T) of Compound A.



FIG. 13 is an XRPD pattern of an IPAc desolvate (Form S) of Compound A.



FIG. 14 is an XRPD pattern of an acetic acid desolvate (Form U) of Compound A.



FIG. 15 is an XRPD pattern of an acetonitrile desolvate (Form V) of Compound A.



FIG. 16 is an XRPD pattern of an ethyl acetate desolvate (Form W) of Compound A.



FIG. 17 is an XRPD pattern of an acetonitrile solvate (Form X) of Compound A.



FIG. 18 is an XRPD pattern of an ethanol desolvate (Form Y) of Compound A.



FIG. 19 is an XRPD pattern of an acetic acid desolvate (Form Z) of Compound A.



FIG. 20 is an XRPD pattern of an acetone desolvate (Form α) of Compound A.



FIG. 21 is an XRPD pattern of an N-methylpyrrolidone (NMP) solvate (Form β) of Compound A.



FIG. 22 is an XRPD pattern of a dimethyl sulfoxide (DMSO) solvate (Form χ) of Compound A.



FIG. 23 is an XRPD pattern of a possible THF solvate (Form δ) of Compound A.



FIG. 24 is an XRPD pattern of a mixture of Form C and a possible acetone solvate (Form ε) of Compound A.



FIG. 25 is an XRPD pattern of an acetone solvate (Form ϕ) of Compound A.



FIG. 26 is an XRPD pattern of an IPA solvate (Form η) of Compound A.



FIG. 27 is an XRPD pattern of an IPAc desolvate (Form λ) of Compound A.



FIG. 28 is an XRPD pattern of the cocrystal obtained from heating a mixture of Form A and glutaric acid showing no change before and after drying. The patterns of Form A and the co-former glutaric acid are provided for comparison.



FIG. 29 is an XRPD pattern of the Calcium salt of Compound A from the experiment L100110-68-1 along with peak positions (Form 1-A).



FIG. 30 is an XRPD pattern of the Calcium salt of Compound A from the experiment L100110-68-3-Wet along with peak positions (Form 1-B).



FIG. 31 is an XRPD pattern of the Calcium salt of Compound A from the experiment L100110-68-8 along with peak positions (Form 2-B).



FIG. 32 is an XRPD pattern of the Calcium salt of Compound A from the experiment L100110-68-10 after exposing to saturated humidity environment along with peak positions (Form 2-D).



FIG. 33 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-11-Dry along with peak positions (3-A).



FIG. 34 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-11 upon exposing it to saturated humidity environment at room temperature along with peak positions (3-C).



FIG. 35 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-13 before drying (3-B).



FIG. 36 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-13, dried after deliquescing (3-D).



FIG. 37 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-17, before drying (Form 4-B).



FIG. 38 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-20, before drying (Form 4-D).



FIG. 39 is an XRPD pattern of the Magnesium salt of Compound A from the experiment L100110-68-20, after drying (Form 4-E).



FIG. 40 is an XRPD pattern of the Sodium salt of Compound A from the experiment L100110-68-21 along with peak positions (Form 5-A).



FIG. 41 is an XRPD pattern of the Sodium salt of Compound A from the experiment L100110-68-24 along with peak positions (Form 5-B).



FIG. 42 is an XRPD pattern of the Sodium salt of Compound A from the experiment L100110-68-25 along with peak positions (Form 5-C).



FIG. 43 is an XRPD pattern of the Sodium salt of Compound A from the experiment L100110-68-25 after humidity exposure, that resulted in substantial improvement in crystallinity. along with peak positions (Form 5-D).



FIG. 44 is an XRPD pattern of the Potassium salt of Compound A from the experiment L100110-68-26 after drying along with peak positions (Form 6-A).



FIG. 45 is an XRPD pattern of the Potassium salt of Compound A from the experiment L100110-68-29 along with peak positions (Form 6-B).



FIG. 46 is an XRPD pattern of the Potassium salt of Compound A from the experiment L100110-68-30 after drying along with peak positions (Form 6-C).



FIG. 47 is an XRPD pattern of the Potassium salt of Compound A from the experiment L100110-68-30-H along with peak positions (Form 6-D).



FIG. 48 is an XRPD pattern of the Ethanolamine salt of Compound A from the experiment L100110-68-31 along with peak positions (Form 7-A).



FIG. 49 is an XRPD pattern of the Ethanolamine salt of Compound A from the experiment L100110-68-32 after drying along with peak positions (Form 7-B).



FIG. 50 is an XRPD pattern of the Diethanolamine salt of Compound A from the experiment L100110-68-36 (Form 8-A) along with peak positions.



FIG. 51 is an XRPD pattern of the Diethanolamine salt of Compound A from the experiment L100110-68-38 (Form 8-B) along with peak positions.



FIG. 52 is an XRPD pattern of the Diethanolamine salt of Compound A from the experiment L100110-68-36 after subjecting to saturated humidity environment at RT (Form 8-C) along with peak positions.



FIG. 53 is an XRPD pattern of the Diethanolamine salt of Compound A from the experiment L100110-68-40 before drying (Form 8-D) along with peak positions.



FIG. 54 is an XRPD pattern of the Diethanolamine salt of Compound A from the experiment L100110-68-40 after drying followed by exposure to saturated humidity environment (Form 8-E) (Essentially Form 8-B with extra peaks) along with peak positions.



FIG. 55 is an XRPD pattern of the Triethanolamine salt of Compound A from the experiment L100110-68-42 (Form 9-A) along with peak positions.



FIG. 56 is an XRPD pattern of the Triethanolamine salt of Compound A from the experiment L100110-68-44 (Form 9-B) along with peak positions.



FIG. 57 is an XRPD pattern of the Triethanolamine salt of Compound A from the experiment L100110-68-41 after drying and subjecting it to saturated humidity environment at room temperature (RT) (Form 9-C) along with peak positions.



FIG. 58 is an XRPD pattern of the Triethanolamine salt of Compound A from the experiment L100110-68-44 after drying, subjecting it to saturated humidity environment at RT (Form 9-D) along with peak positions.



FIG. 59 is an XRPD pattern of the Triethanolamine salt of Compound A from the experiment L100110-68-45 (Form 9-E) along with peak positions.



FIG. 60 is an XRPD pattern of the Diethylamine salt of Compound A from the scale-up experiment L100110-85-9 (Form 10-A) along with peak positions.



FIG. 61 is an XRPD pattern of the Diethylamine salt of Compound A from the experiment L100110-68-46 followed by drying (Form 10-C) along with peak positions.



FIG. 62 is an XRPD pattern of the Diethylamine salt of Compound A from the experiment L100110-68-49 (Form 10-B+extra minor peaks) along with their peak positions.



FIG. 63 is an XRPD pattern of the Ethanol-2-diethylamine salt of Compound A from the experiment L100110-68-56 (Form 12-A) along with their peak positions.



FIG. 64 is an XRPD pattern of the Ethanol-2-diethylamine salt of Compound A from the experiment L100110-68-60 (Form 12-B) along with their peak positions.



FIG. 65 is an XRPD pattern of the Ethanol-2-diethylamine salt of Compound A from the experiment L100110-68-60 after subjecting it to saturated humidity environment at RT (Form 12-C) along with their peak positions.



FIG. 66 is an XRPD pattern of the Choline hydroxide salt of Compound A from the experiment L100110-68-64 (Form 13-A) along with their peak positions.



FIG. 67 is an XRPD pattern of the L-Arginine salt of Compound A from the experiment L100110-68-66 (Form 14-A) along with their peak positions.



FIG. 68 is an XRPD pattern of the L-Arginine salt of Compound A from the experiment L100110-68-68 (Form 14-B) along with their peak positions,



FIG. 69 is an XRPD pattern of the L-Arginine salt of Compound A from the experiment L100110-68-69 (Form 14-C) along with their peak positions.



FIG. 70 is an XRPD pattern of the L-Arginine salt of Compound A from the experiment L100110-68-70 after drying (Form 14-E) along with their peak positions.



FIG. 71 is an XRPD pattern of the L-Histidine salt of Compound A from the experiment L100110-68-71 after drying (Form 15-A) along with their peak positions.



FIG. 72 is an XRPD pattern of the L-Histidine salt of Compound A from the experiment L100110-68-71 after drying, followed by subjecting it to saturated humidity environment at RT (Form 15-B) along with its peak positions.



FIG. 73 is an XRPD pattern of the L-Histidine salt of Compound A from the experiment L100110-68-72 after drying (Form 15-C) along with its peak positions.



FIG. 74 is an XRPD pattern of the L-Histidine salt of Compound A from the experiment L100110-68-75 before drying (Form 15-D) along with its peak positions.



FIG. 75 is an XRPD pattern of the L-Histidine salt of Compound A from the experiment L100110-68-75 (Form 15-E) along with their peak positions.



FIG. 76 is an XRPD pattern of the L-Lysine salt of Compound A from the experiment L100110-68-79 (Form 16-A) along with their peak positions.



FIG. 77 is an XRPD pattern of the Meglumine salt of Compound A from the experiment L100110-68-82 (Form 17-A) along with their peak positions.



FIG. 78 is an XRPD pattern of the Meglumine salt of Compound A from the experiment L100110-68-85 (Form 17-B) along with their peak positions.



FIG. 79 is a graph showing a series of XRPD patterns of amorphous solids that were produced during the trials with 1:4 mixtures of Compound A with polymer.



FIG. 80 is a graph showing a series of XRPD patterns of amorphous solids after 1 month of storage that were produced during the trials with 1:4 mixtures of Compound A with polymer.



FIG. 81 is a graph showing a series of XRPD patterns of amorphous solids that were produced during the trials with 1:2 mixtures of Compound A with polymer.



FIG. 82 is a graph showing a series of XRPD patterns of amorphous solids after 1 month of storage that were produced during the trials with 1:2 mixtures of Compound A with polymer.



FIG. 83 is a dynamic vapor sorption (DVS) isotherm of the glutaric acid co-crystal of Compound A showing a total mass gain of 1.12% by weight between 2% and 95% relative humidity environments.



FIG. 84 is an XRPD pattern of the glutaric acid co-crystal after subjecting it to DVS analysis (L100129-24-Dry-Post DVS), compared with the starting material, no change was observed.



FIG. 85 is an XRPD pattern of a mixture of Form B and Form M.



FIG. 86 is an XRPD pattern of a mixture of Form F and Form N.



FIG. 87 is an XRPD pattern of a mixture of Form A and Form O.



FIG. 88 is an XRPD pattern of a mixture of Form B and Form P.



FIG. 89 is an XRPD pattern of a mixture of Form C and Form Q.



FIG. 90 is an XRPD pattern of a mixture of Form F and Form R.





DETAILED DESCRIPTION OF THE INVENTION

Provided herein are various morphic forms, co-crystals, salts, and amorphous solid dispersions of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile (Compound A). The synthetic methods for Compound A can be found at U.S. Pat. Nos. 7,452,882 and 9,266,861, the contents of each of which are incorporated herein by reference. U.S. Pat. No. 9,266,861 also discloses Form A (FIG. 1) of Compound A and methods of production thereof.


All the XRPD patterns described herein are based on a Cu Kα radiation wavelength (1.54 Å).


In one aspect, the present disclosure provides a morphic form of Compound A. In some embodiments, the morphic form is a solvate such as a methanol solvate, an ethanol solvate, an acetone solvate, a tetrahydrofuran solvate, an N-methylpyrrolidone solvate, a methyl isobutyl ketone solvate, an isopropyl acetate solvate, an acetic acid solvate, a dimethyl acetamide solvate, a dimethyl sulfoxide solvate, an isopropanol solvate, and an acetonitrile solvate.


In some embodiments, the morphic form is a desolvate such as an acetic acid desolvate, an acetonitrile desolvate, an ethyl acetate desolvate, an ethanol desolvate, and an acetone desolvate.


In some embodiments, the morphic form (Form B) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 5.92, 11.8, and 17.5 degrees 2θ, wherein the X-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form B can further include a peak at about 15.0 degrees 2θ. In some embodiments, Form B has an X-ray diffraction pattern substantially similar to that set forth in FIG. 2.


In some embodiments, the morphic form (Form C) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 5.74, 11.5, 17.7, 19.3, 19.7, 21.4, 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form C can further include one or more peaks from Table 5. In some embodiments, Form C has an X-ray diffraction pattern substantially similar to that set forth in FIG. 3.


In some embodiments, the morphic form (Form D) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 5.52, 8.52, 11.0, 16.5, 18.3, 21.0, 21.2, and 24.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form D can further include one or more peaks from Table 6. In some embodiments, Form D has an X-ray diffraction pattern substantially similar to that set forth in FIG. 4.


In some embodiments, the morphic form (Form E) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 7.13, 10.8, 12.3, 14.1, 14.7, 15.5, 16.1, 17.5, 18.1, 19.9, 20.2, 21.0, 21.2, 22.7, 22.9, 24.4, 25.3, and 26.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form E can further include one or more peaks from Table 7. In some embodiments, Form E has an X-ray diffraction pattern substantially similar to that set forth in FIG. 5.


In some embodiments, the morphic form (Form F) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.1, 10.4, 11.4, 13.9, 16.2, 16.4, 17.1, 22.0, 23.8, and 29.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form F can further include one or more peaks from Table 8. In some embodiments, Form F has an X-ray diffraction pattern substantially similar to that set forth in FIG. 6.


In some embodiments, the morphic form (Form G) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 9.50, 12.9, 16.7, 17.3, 19.5, 20.2, 25.6, and 28.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form G can further include one or more peaks from Table 9. In some embodiments, Form G has an X-ray diffraction pattern substantially similar to that set forth in FIG. 7.


In some embodiments, the morphic form (Form H) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 9.22, 19.8, 23.6, 25.9, and 28.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form H can further include one or more peaks from Table 10. In some embodiments, Form H has an X-ray diffraction pattern substantially similar to that set forth in FIG. 8.


In some embodiments, the morphic form (Form I) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 5.77, 9.30, 10.2, 11.6, and 21.9 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form I can further include one or more peaks from Table 11. In some embodiments, Form I has an X-ray diffraction pattern substantially similar to that set forth in FIG. 9.


In some embodiments, the morphic form (Form K) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 8.42, 11.4, 14.5, 18.9, 21.1, and 21.6 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form K can further include one or more peaks from Table 12. In some embodiments, Form K has an X-ray diffraction pattern substantially similar to that set forth in FIG. 10.


In some embodiments, the morphic form (Form L) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.5, 11.5, 11.9, 15.2, 15.7, 16.0, 16.9, 17.1, 18.4, 18.7, 22.0, 22.8, 23.5, and 26.4 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form L can further include one or more peaks from Table 13. In some embodiments, Form L has an X-ray diffraction pattern substantially similar to that set forth in FIG. 11.


In some embodiments, the morphic form (Form S+T) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 7.42, 10.5, 11.3, 12.4, 14.3, 15.8, 16.8, 17.7, 18.1, 18.4, 20.1, 20.5, 21.1, 21.9, 23.2, 25.5, 26.9, and 28.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form S+T can further include one or more peaks from Table 14. In some embodiments, Form S+T has an X-ray diffraction pattern substantially similar to that set forth in FIG. 12.


In some embodiments, the morphic form (Form S) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.5, 12.3, 14.4, 15.8, 16.7, 17.7, 18.1, 18.4, 20.1, 20.6, 21.2, 21.9, 23.3, 24.4, 25.5, and 27.8 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form S can further include one or more peaks from Table 15. In some embodiments, Form S has an X-ray diffraction pattern substantially similar to that set forth in FIG. 13.


In some embodiments, the morphic form (Form U) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 5.79, 8.43, 11.4, 11.6, 14.5, 18.9, 21.1, and 21.6 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form U can further include one or more peaks from Table 16. In some embodiments, Form U has an X-ray diffraction pattern substantially similar to that set forth in FIG. 14.


In some embodiments, the morphic form (Form V) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 6.35, 10.6, 15.6, 16.5, 16.8, and 18.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form V can further include one or more peaks from Table 17. In some embodiments, Form V has an X-ray diffraction pattern substantially similar to that set forth in FIG. 15.


In some embodiments, the morphic form (Form W) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.1, 10.4, 10.7, 11.7, 13.9, 24.4, and 29.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form W can further include one or more peaks from Table 18. In some embodiments, Form W has an X-ray diffraction pattern substantially similar to that set forth in FIG. 16.


In some embodiments, the morphic form (Form X) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 9.66, 10.2, 10.5, 11.2, 18.7, and 24.7 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form X can further include one or more peaks from Table 19. In some embodiments, Form X has an X-ray diffraction pattern substantially similar to that set forth in FIG. 17.


In some embodiments, the morphic form (Form Y) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 6.51, 13.0, 13.3, 19.5, and 24.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form Y can further include one or more peaks from Table 20. In some embodiments, Form Y has an X-ray diffraction pattern substantially similar to that set forth in FIG. 18.


In some embodiments, the morphic form (Form Z) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.6, 11.2, 11.6, 12.0, 14.3, 15.6, 16.2, 17.6, 18.1, 18.7, 24.1, and 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form Z can further include one or more peaks from Table 21. In some embodiments, Form Z has an X-ray diffraction pattern substantially similar to that set forth in FIG. 19.


In some embodiments, the morphic form (Form α) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 7.26, 10.1, 10.4, 10.6, 11.9, 13.9, 16.5, 21.9, 22.4, and 24.1 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form α can further include one or more peaks from Table 22. In some embodiments, Form α has an X-ray diffraction pattern substantially similar to that set forth in FIG. 20.


In some embodiments, the morphic form (Form β) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 7.36, 10.5, 14.3, 15.7, 18.3, 20.4, 21.0, 21.8, and 23.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form β can further include one or more peaks from Table 23. In some embodiments, Form β has an X-ray diffraction pattern substantially similar to that set forth in FIG. 21.


In some embodiments, the morphic form (Form χ) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 8.53, 11.2, 18.4, 20.1, and 21.4 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form χ can further include one or more peaks from Table 24. In some embodiments, Form χ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 22.


In some embodiments, the morphic form (Form δ) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.9, 12.1, 14.4, 18.1, 19.6, 24.5, and 27.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form δ can further include one or more peaks from Table 25. In some embodiments, Form δ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 23.


In some embodiments, the morphic form (Form ε) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 5.73, 11.4, 16.6, 17.6, 23.2, and 24.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form ε can further include one or more peaks from Table 26. In some embodiments, Form ε has an X-ray diffraction pattern substantially similar to that set forth in FIG. 24.


In some embodiments, the morphic form (Form ϕ) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 6.95, 13.9, 20.9, 22.3, and 27.9 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form ϕ can further include one or more peaks from Table 27. In some embodiments, Form ϕ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 25.


In some embodiments, the morphic form (Form η) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 6.87, 7.69, 20.5, 23.0, 23.9, and 28.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form η can further include one or more peaks from Table 28. In some embodiments, Form η has an X-ray diffraction pattern substantially similar to that set forth in FIG. 26.


In some embodiments, the morphic form (Form λ) of Compound A is characterized by an X-ray powder diffraction pattern including peaks at about 10.6, 12.0, 14.3, 16.2, 17.6, 18.0, and 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å). In some embodiments, the X-ray powder diffraction pattern of Form λ can further include one or more peaks from Table 29. In some embodiments, Form λ has an X-ray diffraction pattern substantially similar to that set forth in FIG. 27.


In some embodiments, the morphic form has purity of greater than 85% by weight, e.g., greater than 86% by weight, greater than 90% by weight, greater than 92.5% by weight, greater than 95% by weight, greater than 96% by weight, greater than 97% by weight, greater than 97.5% by weight, greater than 98% by weight, greater than 98.5% by weight, greater than 99% by weight, greater than 99.2% by weight, greater than 99.5% by weight, or greater than 99.8% by weight. For example, the content of impurities (i.e., any components of the composition other than Compound A, such as byproducts, starting material, solvent residues, heavy metal, etc.) is less than 15% by weight, less than 14% by weight, less than 10% by weight, less than 8% by weight, less than 5% by weight, less than 4% by weight, less than 3% by weight, less than 2% by weight, less than 1.5% by weight, less than 1% by weight, less than 0.8% by weight, less than 0.5% by weight, or less than 0.2% by weight.


In another aspect, the present disclosure provides an amorphous solid dispersion of Compound A. As used herein, the term “solid dispersion” refers to a system in a solid state comprising at least two components, wherein one component is dispersed throughout the other component or components. The term “amorphous solid dispersion” as used herein, refers to a stable solid dispersion comprising an amorphous drug substance and a stabilizing polymer. Non-limiting examples of the stabilizing polymer are polyvinylpyrrolidone MW 10,000 (PVP-10), polyvinylpyrrolidone MW 40,000 (PVP-40), and poly(1-vinylpyrrolidone-co-vinyl acetate) (PVP-Co-VA), hydroxy-propyl methyl cellulose (Hypromellose), methylcellulose, hydroxy propyl cellulose, and poly ethylene glycol (PEG) 6000. In some embodiments, the stabilizing polymer is polyvinylpyrrolidone.


The amorphous solid dispersion of the present disclosure includes Compound A and a stabilizing polymer. The amount of Compound A in the amorphous solid dispersions of the present disclosure can range from about 0.1% to about 60% by weight relative to the stabilizing polymer. In some embodiments, the amount of Compound A in the amorphous solid dispersions of the present disclosure ranges from about 15% to about 50% by weight relative to the stabilizing polymer. In some embodiments, the amount of Compound A in the amorphous solid dispersions of the present disclosure can be about 50% by weight relative to the stabilizing polymer. In some embodiments, the amount of Compound A in the amorphous solid dispersions of the present disclosure can be about 25% by weight relative to the stabilizing polymer.


In another aspect, the present disclosure provides a co-crystal of Compound A. Co-crystal screening was performed on Compound A with 20 different potential co-formers. The co-formers tested include adipic acid, L-arginine, ascorbic acid, benzoic acid, citric acid, D (+) glucose, glutaric acid, L-histidine, 4-hydroxy benzoic acid, 3,4-dihydroxy benzoic acid, L-lysine, malic acid, salicyclic acid, succinic acid, tartaric acid, urea, vanillin, and vanillic acid. So far, only a Compound A/glutaric acid co-crystal has been observed.


In some embodiments, the co-crystal can be characterized by an XRPD pattern including peaks at about 9.74, 10.8, 11.0, 12.2, 16.1, 17.0, 19.2, 21.9, and 23.1 degrees 2θ. In some embodiments, the X-ray powder diffraction pattern of the co-crystal can further include one or more peaks from Table 30. In some embodiments, the co-crystal has an XRPD pattern substantially similar to that set forth in FIG. 28. In some embodiments, the co-crystal has purity of greater than 85% by weight, e.g., greater than 86% by weight, greater than 90% by weight, greater than 92.5% by weight, greater than 95% by weight, greater than 96% by weight, greater than 97% by weight, greater than 97.5% by weight, greater than 98% by weight, greater than 98.5% by weight, greater than 99% by weight, greater than 99.2% by weight, greater than 99.5% by weight, or greater than 99.8% by weight. For example, the content of impurities (i.e., any components of the composition other than Compound A, such as byproducts, starting material, solvent residues, heavy metal, etc.) is less than 15% by weight, less than 14% by weight, less than 10% by weight, less than 8% by weight, less than 5% by weight, less than 4% by weight, less than 3% by weight, less than 2% by weight, less than 1.5% by weight, less than 1% by weight, less than 0.8% by weight, less than 0.5% by weight, or less than 0.2% by weight.


In yet another aspect, the present disclosure provides a crystalline salt of Compound A. The crystalline salt comprises Compound A and one or more counter-ions. The molar ratio of Compound A over the counter-ion can be 1:1 to 2:1. In some embodiments, the molar ratio of Compound A over the counter-ion is 1:1.1. In some embodiments, the molar ratio of Compound A over the counter-ion is 1:1. In some embodiments, the counter-ion is L-lysine, L-arginine, 2-hydroxy-N,N,N-trimethylethan-1-aminium, diethylamine, ethanolamine, ethanol-2-diethylamine, Na+, Mg2+, K+, Ca2+, diethanolamine, triethanolamine, L-histidine, meglumine, or a combination thereof. In some embodiments, the crystalline salt has an XRPD pattern substantially similar to that set forth in any one of FIGS. 29-78.


In some embodiments, the counter-ion is L-lysine. In some embodiments, when the counter-ion is L-lysine, the crystalline salt can be characterized by an XRPD pattern including peaks at about 8.70, 9.22, 11.3, 17.0, and 24.8 degrees 2θ. The XRPD pattern can further include peaks at about 7.12, 18.4, 19.1, 20.4, and 25.7 degrees 2θ. In some embodiments, the L-lysine salt has an XRPD pattern substantially similar to that set forth in FIG. 76. The L-lysine salt can have a melting point of about 250° C.


In some embodiments, the counter-ion is L-arginine. The L-arginine salt can have an XRPD pattern substantially similar to that set forth in any one of FIGS. 67-70. The L-arginine salt can have a melting point of about 200° C.


In some embodiments, the counter-ion is 2-hydroxy-N,N,N-trimethylethan-1-aminium. In some embodiments, the 2-hydroxy-N,N,N-trimethylethan-1-aminium salt can have an XRPD pattern substantially similar to that set forth in FIG. 66. The 2-hydroxy-N,N,N-trimethylethan-1-aminium salt can have a melting point of about 229° C.


In yet another aspect, the present disclosure includes a salt of Compound A in the form of a solvate (referred to herein as “a salt solvate”). The salt solvate can contain one or more counter-ions. In some embodiments, the counter ion can be potassium, sodium, or magnesium. In some embodiments, the salt solvate is a salt of Compound A (e.g., potassium salt, sodium salt, or magnesium salt) in the form of an acetic acid solvate. In some embodiments, the salt solvate is a salt of Compound A in the form of a tetrahydrofuran solvate. In some embodiments, the salt solvate is a salt of Compound A that includes a solvent and water. Such morphic forms may include a solvent and water in a single chemical entity with Compound A and the counter-ion, or may comprise a physical mixture of Compound A as the salt in a hydrate and a solvate.


The potassium salt solvate of Compound A is useful for the removal of impurities, which may be removed during the isolation of the salt solvate.


In another aspect of the invention, a morphic form of one type may be converted to another. A morphic form comprising a solvate or hydrate may be converted to a form having a counter-ion.


In some embodiments, the crystalline salt has purity of greater than 85% by weight, e.g., greater than 86% by weight, greater than 90% by weight, greater than 92.5% by weight, greater than 95% by weight, greater than 96% by weight, greater than 97% by weight, greater than 97.5% by weight, greater than 98% by weight, greater than 98.5% by weight, greater than 99% by weight, greater than 99.2% by weight, greater than 99.5% by weight, or greater than 99.8% by weight. For example, the content of impurities (i.e., any components of the composition other than Compound A, such as byproducts, starting material, solvent residues, heavy metal, counter-ions, etc.) is less than 15% by weight, less than 14% by weight, less than 10% by weight, less than 8% by weight, less than 5% by weight, less than 4% by weight, less than 3% by weight, less than 2% by weight, less than 1.5% by weight, less than 1% by weight, less than 0.8% by weight, less than 0.5% by weight, or less than 0.2% by weight.


The present disclosure also provides a mixture of two or more of the forms disclosed herein. The forms can be present at any weight ratio in the mixture. For example, two or more of the morphic forms disclosed herein can be present in a mixture.


The present disclosure also provides a pharmaceutical composition comprising any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions of Compound A as disclosed herein. The pharmaceutical composition can further include at least one pharmaceutically acceptable excipient or carrier.


A “pharmaceutical composition” is a formulation containing a compound of the present invention in a form suitable for administration to a subject. In some embodiments, the pharmaceutical composition is in bulk or in unit dosage form. The unit dosage form can be in any of a variety of forms, including, for example, a capsule, an IV bag, a tablet, a single pump on an aerosol inhaler or a vial. The quantity of active ingredient (e.g., a formulation of the disclosed morphic forms, co-crystals, salts, and amorphous solid dispersions) in a unit dose of composition is an effective amount and is varied according to the particular treatment involved. One skilled in the art will appreciate that it is sometimes necessary to make routine variations to the dosage depending on the age and condition of the patient. The dosage will also depend on the route of administration. A variety of routes are contemplated, including oral, pulmonary, rectal, parenteral, transdermal, subcutaneous, intravenous, intramuscular, intraperitoneal, inhalational, buccal, sublingual, intrapleural, intrathecal, intranasal, and the like. Dosage forms for topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. In some embodiments, Compound A is mixed under sterile conditions with a pharmaceutically acceptable carrier, and with any preservatives, buffers or propellants that are required.


A pharmaceutical composition of the invention is formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), and transmucosal administration. Solutions or suspensions used for parenteral, intradermal, or subcutaneous application can include the following components: a sterile diluent such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerine, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl parabens; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates, and agents for the adjustment of tonicity such as sodium chloride or dextrose. The pH can be adjusted with acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.


In the practice of the method of the present invention, an effective amount of any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions of this invention is administered via any of the usual and acceptable methods known in the art, either singly or in combination. The compositions can thus be administered orally (e.g., buccal cavity), sublingually, parenterally (e.g., intramuscularly, intravenously, or subcutaneously), rectally (e.g., by suppositories or washings), transdermally (e.g., skin electroporation), or by inhalation (e.g., by aerosol), and in the form or solid, liquid or gaseous dosages, including tablets and suspensions. The administration can be conducted in a single unit dosage form with continuous therapy or in a single dose therapy ad libitum. The therapeutic composition can also be in the form of an oil emulsion or dispersion in conjunction with a lipophilic salt such as pamoic acid, or in the form of a biodegradable sustained-release composition for subcutaneous or intramuscular administration.


Useful pharmaceutical carriers for the preparation of the compositions hereof, can be solids, liquids or gases; thus, the compositions can take the form of tablets, pills, capsules, suppositories, powders, enterically coated or other protected formulations (e.g. binding on ion-exchange resins or packaging in lipid-protein vesicles), sustained release formulations, solutions, suspensions, elixirs, aerosols, and the like. The carrier can be selected from the various oils including those of petroleum, animal, vegetable or synthetic origin, e.g., peanut oil, soybean oil, mineral oil, sesame oil, and the like. Water, saline, aqueous dextrose, and glycols are preferred liquid carriers, particularly (when isotonic with the blood) for injectable solutions. For example, formulations for intravenous administration comprise sterile aqueous solutions of the active ingredient(s) which are prepared by dissolving solid active ingredient(s) in water to produce an aqueous solution, and rendering the solution sterile. Suitable pharmaceutical excipients include starch, cellulose, talc, glucose, lactose, talc, gelatin, malt, rice, flour, chalk, silica, magnesium stearate, sodium stearate, glycerol monostearate, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol, and the like. The compositions may be subjected to conventional pharmaceutical additives such as preservatives, stabilizing agents, wetting or emulsifying agents, salts for adjusting osmotic pressure, buffers and the like. Suitable pharmaceutical carriers and their formulation are described in Remington's Pharmaceutical Sciences by E. W. Martin. Such compositions will, in any event, contain an effective amount of the active compound together with a suitable carrier so as to prepare the proper dosage form for proper administration to the recipient.


The pharmaceutical preparations can also contain preserving agents, solubilizing agents, stabilizing agents, wetting agents, emulsifying agents, sweetening agents, coloring agents, flavoring agents, salts for varying the osmotic pressure, buffers, coating agents or antioxidants. They can also contain other therapeutically valuable substances, including additional active ingredients other than those of Compound A.


The compositions disclosed herein are useful as medicaments for the treatment of a resistance to thyroid hormone (RTH) syndrome in a subject who has at least one TRβ mutation. Accordingly, the present disclosure provides the compositions disclosed herein for use in treating a RTH syndrome in a subject having at least one TRβ mutation. The present disclosure also provides a method for treating a RTH syndrome in a subject having at least one TRβ mutation, the method comprising administering to the subject a therapeutically effective amount of any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions disclosed herein. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for the manufacture of a medicament for treating a RTH syndrome in a subject having at least one TRβ mutation in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for use in treating a RTH syndrome in a subject having at least one TRβ mutation in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount.


The subject may exhibit one or more symptoms of the RTH syndrome, such as obesity, hyperlipidemia, hypercholesterolemia, heterozygous familial hypercholesterolemia, diabetes, non-alcoholic steatohepatitis, fatty liver, fatty liver disease, bone disease, dyslipidemia, thyroid axis alteration, atherosclerosis, a cardiovascular disorder, tachycardia, hyperkinetic behavior, hypothyroidism, goiter, attention deficit hyperactivity disorder, learning disabilities, mental retardation, hearing loss, delayed bone age, neurologic or psychiatric disease or thyroid cancer. Details about the RTH syndrome can be found at Weiss and Refetoff, “Resistance to Thyroid Hormone (RTH) in the Absence of Abnormal Thyroid Hormone Receptor (TR) (nonTR-RTH),” Hot Thyroidology 09/09, 11 pages in total, the contents of which are incorporated by reference in their entireties.


Thyroid hormone receptor nucleic acids and polypeptides from various species (e.g., human, rat, chicken, etc.) have previously been described. See, e.g., R. L. Wagner et al. (2001), Molecular Endocrinology 15(3): 398-410; J. Sap et al. (1986), Nature 324:635-640; C. Weinberger et al. (1986), Nature 324:641-646; and C. C. Tompson et al. (1986), Science 237:1610-1614; each of which is incorporated herein by reference in its entirety. The amino acid sequence of human TRß is provided, e.g., by Genbank Accession No. P10828.2, incorporated herein by reference.









Amino acid sequence of the ligand binding domain


(residues 203-461) of human TRβ


(SEQ ID NO: 1)


ELQKSIGHKPEPTDEEWELIKTVTEAHVATNAQGSHWKQKRKFLPEDIGQA





PIVNAPEGGKVDLEAFSHFTKIITPAITRVVDFAKKLPMFCELPCEDQIIL





LKGCCMEIMSLRAAVRYDPESETLTLNGEMAVTRGQLKNGGLGVVSDAIFD





LGMSLSSFNLDDTEVALLQAVLLMSSDRPGLACVERIEKYQDSFLLAFEHY





INYRKHHVTHFWPKLLMKVTDLRMIGACHASRFLHMKVECPTELFPPLFLE





VFED






The residues at the 234, 243, 316, and 317 positions of human TRβ are underlined in SEQ ID NO: 1. The portion of the human TRβ nucleotide sequence that encodes the above amino acid sequence is SEQ ID NO: 2. The nucleotide sequence of human TRβ is provided, e.g., by Genbank Accession No. NM 000461.4, incorporated herein by reference.









Nucleic acid sequence encoding the ligand binding


domain of human TRβ


(SEQ ID NO: 2)


GAGCTGCAGAAGTCCATCGGGCACAAGCCAGAGCCCACAGACGAGGAATGG





GAGCTCATCAAAACTGTCACCGAAGCCCATGTGGCGACCAACGCCCAAGGC





AGCCACTGGAAGCAAAAACGGAAATTCCTGCCAGAAGACATTGGACAAGCA





CCAATAGTCAATGCCCCAGAAGGTGGAAAGGTTGACTTGGAAGCCTTCAGC





CATTTTACAAAAATCATCACACCAGCAATTACCAGAGTGGTGGATTTTGCC





AAAAAGTTGCCTATGTTTTGTGAGCTGCCATGTGAAGACCAGATCATCCTC





CTCAAAGGCTGCTGCATGGAGATCATGTCCCTTCGCGCTGCTGTGCGCTAT





GACCCAGAAAGTGAGACTTTAACCTTGAATGGGGAAATGGCAGTGACACGG





GGCCAGCTGAAAAATGGGGGTCTTGGGGTGGTGTCAGACGCCATCTTTGAC





CTGGGCATGTCTCTGTCTTCTTTCAACCTGGATGACACTGAAGTAGCCCTC





CTTCAGGCCGTCCTGCTGATGTCTTCAGATCGCCCGGGGCTTGCCTGTGTT





GAGAGAATAGAAAAGTACCAAGATAGTTTCCTGCTGGCCTTTGAACACTAT





ATCAATTACCGAAAACACCACGTGACACACTTTTGGCCAAAACTCCTGATG





AAGGTGACAGATCTGCGGATGATAGGAGCCTGCCATGCCAGCCGCTTCCTG





CACATGAAGGTGGAATGCCCCACAGAACTCTTCCCCCCTTTGTTCTTGGAA





GTGTTCGAGGATTAG






The TRβ mutation is selected from the group consisting of a substitution of threonine (T) for the wild type residue alanine (A) at amino acid position 234 of SEQ ID NO: 1 (A234T); a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 243 of SEQ ID NO: 1 (R243Q); a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 316 of SEQ ID NO: 1 (R316H); and a substitution of threonine (T) for the wild type residue alanine (A) at amino acid position 317 of SEQ ID NO: 1 (A317T).


The compositions disclosed herein are also useful as medicaments for the treatment of non-alcoholic steatohepatitis (NASH). NASH is liver inflammation and damage caused by a buildup of fat in the liver. Accordingly, the present disclosure provides the compositions disclosed herein for use in treating NASH in a subject in need thereof. The present disclosure also provides a method for treating NASH in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions disclosed herein. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for the manufacture of a medicament for treating non-alcoholic steatohepatitis in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for use in treating non-alcoholic steatohepatitis in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount.


The compositions disclosed herein are also useful as medicaments for the treatment of familial hypercholesterolemia (FH). FH is an inherited genetic disorder that causes dangerously high cholesterol levels, which can lead to heart disease, heart attack, or stroke at an early age if left untreated. There are two types of FH: homozygous FH (HoFH) and heterozygous FH (HeFH). Accordingly, the present disclosure provides the compositions disclosed herein for use in treating HoFH or HeFH in a subject in need thereof. The present disclosure also provides a method for treating HoFH or HeFH in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions disclosed herein. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for the manufacture of a medicament for treating familial hypercholesterolemia in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for use in treating familial hypercholesterolemia in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount.


The compositions disclosed herein are also useful as medicaments for the treatment of fatty liver disease. Fatty liver disease is a condition wherein large vacuoles of triglyceride fat accumulate in liver cells via the process of steatosis. Accordingly, the present disclosure provides the compositions disclosed herein for use in treating fatty liver disease in a subject in need thereof. The present disclosure also provides a method for treating fatty liver disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions disclosed herein. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for the manufacture of a medicament for treating fatty liver disease in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for use in treating fatty liver disease in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount.


The compositions disclosed herein are also useful as medicaments for the treatment of dyslipidemia. Dyslipidemia is a condition characterized by an abnormal amount of lipids (e.g. triglycerides, cholesterol and/or fat phospholipids) in the blood. Accordingly, the present disclosure provides the compositions disclosed herein for use in treating dyslipidemia in a subject in need thereof. The present disclosure also provides a method for treating dyslipidemia in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of any one of the morphic forms, co-crystals, salts, and amorphous solid dispersions disclosed herein. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for the manufacture of a medicament for treating dyslipidemia in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount. The present disclosure also provides a crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein for use in treating dyslipidemia in a subject in need thereof, wherein the crystalline salt, morphic form, co-crystal, or amorphous solid dispersion disclosed herein is for administration to the subject in at least one therapeutically effective amount.


The therapeutically effective amount or dosage according to this invention can vary within wide limits and may be determined in a manner known in the art. For example, the drug can be dosed according to body weight. Such dosage will be adjusted to the individual requirements in each particular case including the specific compound(s) being administered, the route of administration, the condition being treated, as well as the patient being treated. In another embodiment, the drug can be administered by fixed does, e.g., dose not adjusted according to body weight. In general, in the case of oral or parenteral administration to adult humans, a daily dosage of from about 0.5 mg to about 1000 mg should be appropriate, although the upper limit may be exceeded when indicated. The dosage is preferably from about 5 mg to about 400 mg per day. For example, the dosage is about 40 mg, about 50 mg, about 80 mg, about 100 mg, about 120 mg, about 140 mg, about 160 mg, about 180 mg, or about 200 mg. A preferred dosage may be from about 20 mg to about 200 mg per day. The daily dosage can be administered as a single dose or in divided doses, or for parenteral administration it may be given as continuous infusion.


An effective amount of a pharmaceutical agent is that which provides an objectively identifiable improvement as noted by the clinician or other qualified observer. As used herein, the term “dosage effective manner” refers to an amount of an active compound to produce the desired biological effect in a subject or cell.


The details of the invention are set forth in the accompanying description below. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, illustrative methods and materials are now described. Other features, objects, and advantages of the invention will be apparent from the description and from the claims. In the specification and the appended claims, the singular forms also include the plural unless the context clearly dictates otherwise. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. All patents and publications cited in this specification are incorporated herein by reference in their entireties.


Definitions

The articles “a” and “an” are used in this disclosure to refer to one or more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.


The term “and/or” is used in this disclosure to mean either “and” or “or” unless indicated otherwise.


The term “solvate” is used herein to describe a morphic form that includes an organic solvent chemically incorporated with the parent molecule in various fractional or integral molar ratios.


The term “hydrate” is used herein to describe a morphic form that includes water chemically incorporated with the parent molecule in various fractional or integral molar ratios.


The term “desolvate” is used herein to describe a morphic form resulting from a solvent being substantially removed from a solvate, typically by heat, vacuum, or both. In some embodiments, at least 75% by weight of the solvent is removed from the solvate to form a desolvate. In some embodiments, at least 80% by weight of the solvent is removed from the solvate to form a desolvate. In some embodiments, at least 85% by weight of the solvent is removed from the solvate to form a desolvate. In some embodiments, at least 90% by weight of the solvent is removed from the solvate to form a desolvate. In some embodiments, at least 95% by weight of the solvent is removed from the solvate to form a desolvate. In some embodiments, at least 99% by weight of the solvent is removed from the solvate to form a desolvate.


As used herein, the phrase “pharmaceutically acceptable” refers to those compounds, materials, compositions, carriers, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


“Pharmaceutically acceptable excipient or carrier” means an excipient or carrier that is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and includes excipient that is acceptable for veterinary use as well as human pharmaceutical use. A “pharmaceutically acceptable excipient” as used in the specification and claims includes both one and more than one such excipient.


The term “therapeutically effective amount”, as used herein, refers to an amount of a pharmaceutical agent to treat, ameliorate, or prevent an identified disease or condition, or to exhibit a detectable therapeutic or inhibitory effect. The effect can be detected by any assay method known in the art. The precise effective amount for a subject will depend upon the subject's body weight, size, and health; the nature and extent of the condition; and the therapeutic or combination of therapeutics selected for administration. Therapeutically effective amounts for a given situation can be determined by routine experimentation that is within the skill and judgment of the clinician. In a preferred aspect, the disease or condition to be treated is a metabolic disorder.


The term “subject” as used herein refers to a mammal, preferably a human.


“Treating” or “treatment” as used herein with regard to a condition may refer to preventing the condition, slowing the onset or rate of development of the condition, reducing the risk of developing the condition, preventing or delaying the development of symptoms associated with the condition, reducing or ending symptoms associated with the condition, generating a complete or partial regression of the condition, or some combination thereof.


As used herein, the term “about” when used in conjunction with numerical values and/or ranges generally refers to those numerical values and/or ranges near to a recited numerical value and/or range. In some instances, the term “about” can mean within ±10% of the recited value. For example, in some instances, “about 100 [units]” can mean within ±10% of 100 (e.g., from 90 to 110).


The term “substantially similar” used in reference to XRPD patterns means that the XRPD pattern of a polymorph may display “batch to batch” variations due to differences in the types of equipment used for the measurements, and fluctuations in both experimental conditions (e.g. purity and grain size of the sample) and instrumental settings (e.g. X-ray wavelengths; accuracy and sensitivity of the diffractometer; and “instrumental drift”) normally associated with the X-ray diffraction measurement. Due to these variations, the same polymorph may not contain XRPD peaks at exactly the same positions or intensities shown in the figures disclosed herein. Accordingly, the term “about” used in reference to the peaks in an XRPD pattern takes into account these variations and a skilled artisan would readily appreciate the scope.


EXAMPLES

The disclosure is further illustrated by the following examples and synthesis examples, which are not to be construed as limiting this disclosure in scope or spirit to the specific procedures herein described. It is to be understood that the examples are provided to illustrate certain embodiments and that no limitation to the scope of the disclosure is intended thereby. It is to be further understood that resort may be had to various other embodiments, modifications, and equivalents thereof which may suggest themselves to those skilled in the art without departing from the spirit of the present disclosure and/or scope of the appended claims.


Example 1. Solvates and Desolvates of Compound A

A suspension of Compound A and methanol was stirred for 2 days then filtered. XRPD analysis indicated the formation of the methanol solvate (Form B, FIG. 2). In a similar manner, the ethanol solvate (Form C, FIG. 3), acetone solvate (Form D, FIG. 4), THF solvate (Form E, FIG. 5), MIBK solvate (Form G, FIG. 7), isopropyl acetate solvate (Form H, FIG. 8), acetic acid solvate (Form I, FIG. 9), dimethyl acetamide solvate (Form K, FIG. 10), and acetonitrile solvate (Form L, FIG. 11).


A suspension of Compound A and ethyl acetate was stirred for 2 days then filtered. Heating to 100° C. for 1 hour generated the desolvate (Form F, FIG. 6). In a similar manner, a different desolvate, Form λ, is formed by heating the isopropyl acetate solvate (Form H, FIG. 27).


Form S+T was generated from the MIBK solvate of Compound A (Form G) by drying overnight at 30° C. under vacuum (FIG. 12). In a similar manner, Form S was generated from Form H. Form U was generated from Form I. Form V was generated from Form L. Form W was generated from an ethyl acetate solvate, formed by slurrying Compound A in ethyl acetate at 50° C.


Form Y (FIG. 18), a desolvate, was generated by evaporating a saturated solution of Compound A in ethanol at 50° C. and atmospheric pressure to dryness. In a similar manner, the following forms were isolated from saturated solutions of Compound A: Form Z (FIG. 19) from acetic acid solution, and Form α (FIG. 20) from acetone solution.


About 50-70 mg of Compound A was accurately weighed in a 4 mL vial and heated to 60° C. Then ethanol and a few drops of N-methyl pyrrolidone (NMP) were added. The contents were stirred with a magnetic stir bar until the solid was dissolved. The vial was placed in an ice/water mixture and kept cold until a precipitate formed. The solid was filtered to give Compound A Form β (FIG. 21), an NMP solvate. Ina similar manner, Form χ (FIG. 22), a DMSO solvate, was obtained by crystallization from IPA:DMSO (96.5:3.5).


Form δ (FIG. 23), a possible THF solvate, was obtained when a solution of Compound A in THF at 60° C. was added to either heptane or cyclohexane at room temperature. In a similar manner, Form C+ε (FIG. 24), a possible acetone solvate, was obtained by adding a solution of Compound A in ethanol:acetone (1:1) to heptane. Form ϕ (FIG. 25), and acetone solvate, was obtained by addition an acetone solution to heptane or cyclohexane.


The isopropyl alcohol (IPA) solvate Form η (FIG. 26) was generated by slurrying the MIBK solvate Form G of Compound A in IPA at room temperature.


A mixture of Form B and Form M (FIG. 85) can be generated by slurring Form A in methanol at 50° C.


A mixture of Form F and Form N (FIG. 86) can be generated by slurring Form A in ethyl acetate at 50° C.


A mixture of Form A and Form O (FIG. 87) can be generated by slurring Form A in acetonitrile at 50° C.


A mixture of Form B and Form P (FIG. 88) can be generated by drying methanol solvate at 30° C. under vacuum overnight.


A mixture of Form C and Form Q (FIG. 89) can be generated by drying ethanol solvate at 30° C. under vacuum overnight.


A mixture of Form F and Form R (FIG. 90) can be generated by drying ethyl acetate solvate at 30° C. under vacuum overnight.


Example 2. Amorphous Solid Dispersions of Compound A

The procedure used to prepare amorphous solid dispersions of Compound A is as follows.


The procedure for the experiments with 1:4 ratio of Compound A:Polymer: (1) Approximately 10 mg of Compound A was weighed in 4 mL vials and −40 mg of the polymer is added to each of the respective vials; (2) Solvent was added to each of the respective vials for saturation; (3) Content of vials were stirred at room temperature, if dissolution was achieved at room temperature, the clear solution was transferred to 2 mL centrifuge tubes; (4) If complete dissolution was not achieved, vials were heated to 60° C. to achieve dissolution. If still dissolution was not achieved at 60° C., the vials were subjected to centrifugation and the supernatant was collected; (5) The solution/supernatant was subjected to evaporation in SpeedVac at about 50° C. and under vacuum; (6) The resulting solid/gel was dried further in a vacuum oven for at least 3 hours which resulted in a solid; and (7) X-ray diffraction was performed on the resulting solid.


The polymers used were polyvinylpyrrolidone MW 10000 (PVP-10), polyvinylpyrrolidone MW 40000 (PVP-40), hypromellose or hydroxy propyl methyl cellulose (HPR), poly(1-vinylpyrrolidone-co-vinyl acetate) (PVP-Co-VA). The solvent systems used were THF:water 9:1, Ethanol:acetone 1:1, acetone, and acetic acid. In addition, amorphous solid dispersions were prepared using a 2:1 ratio of polymer to compound A and the ethanol:acetone 1:1 and acetic acid solvent systems.


Example 3. Preparation of Co-Crystals of Compound A

The generation of co-crystals was attempted through different modes such as solvent drop milling, evaporation of the Compound A/co-former solution, and co-melting beyond the melting point of the co-former. Form A of Compound A was used as the starting material in all the experiments.


The procedure for making a Compound A/glutaric acid co-crystal is described below.


Compound A (154.7 mg) was weighed in a 4 mL vial, and 56.7 mg of glutaric acid was added to the vial (˜1.2 molar equivalents). The contents of the vial were well mixed with a spatula and heated up to 118° C. (20° C. higher than the melting point of co-former). The vial was kept for ˜5 minutes and cooled to 88° C., kept for 15-20 minutes and cooled down to RT. Partial conversion of Compound A to the co-crystal was observed by XRPD.


An additional 0.8 equivalents of glutaric acid were added to this solid. The vial was heated to 118° C. The internal temperature of the system was also checked with a thermocouple and was found to be 108° C. The system was kept at this temperature for 15-20 minutes with periodic mixing with a spatula, cooled down to 88° C. XRPD analysis confirmed conversion to the cocrystal of Compound A and glutaric acid.


The solid that was obtained after adding extra glutaric acid followed by thermal treatment was washed with distilled water (4×500 μL˜13.5 vol. with respect to Compound A) and was subjected to XRPD, no traces of excess glutaric acid was seen. This solid was further dried in a vacuum oven at 50° C. for 3-4 hours.


The co-crystal was not hygroscopic, with only 1.12% mass gain between 2% and 95% relative humidity (RH) environments and it was stable when subjected to 75% RH at 40° C. for 1 week. The isotherm is shown in FIG. 83. No difference in the XRPD pattern was seen after the DVS analysis, the XRPD patterns before and after DVS analysis are shown in FIG. 84.


The kinetic and thermodynamic solubility of the glutaric acid co-crystal was assessed in simulated fasted state gastric and intestinal fluids (FaSSGF and FaSSIF) and also in water. The pH of the system was also measured. The solubility assessment was also performed on Compound A for comparison. A small amount of Compound A and also the co-crystal were slurried in the respective fluid and a sample is collected after 1 hour for kinetic solubility assessment. Samples were collected after 24 hours for equilibrium solubility measurement. The solubility determination is done by HPLC through a short 20-minute generic method that was used during the salt screening project 100177FF. The solubility data is presented in Table 1.









TABLE 1







Solubilities of the Compound A free acid (FA) and the glutaric acid co-


crystal in the simulated fluids FaSSGF, FaSSIF and in water at 37° C.



















Solubility

XRPD


Experiment ID
Solvent
Compound
Kinetic/Equib
Area
(mg/mL)
pH
after slurry

















L100129-29-4
FaSSGF
Form A
Kinetic - 1
6.148
0.0011







hour


L100129-29-5
FaSSIF
Form A
Kinetic - 1
2654.464
0.4895





hour


L100129-29-6
Water
Form A
Kinetic - 1
140.511
0.0259





hour


L100129-29-7
FaSSGF
Glutaric acid
Kinetic - 1
0.551
0.0001




co-crystal
hour


L100129-29-8
FaSSIF
Glutaric acid
Kinetic - 1
1301.012
0.2399




co-crystal
hour


L100129-29-9
Water
Glutaric acid
Kinetic - 1
1.583
0.0003




co-crystal
hour


L100129-29-4
FaSSGF
Form A
Equilibrium -
6.160
0.0011
1.67
Form-A





24 hours


L100129-29-5
FaSSIF
Form A
Equilibrium -
2994.47
0.5522
6.44
Form-A





24 hours


L100129-29-6
Water
Form A
Equilibrium -
1046.449
0.1930
6.57
Dihydrate





24 hours


L100129-29-7
FaSSGF
Glutaric acid
Equilibrium -
5.214
0.0010
1.66
Dihydrate




co-crystal
24 hours


L100129-29-8
FaSSIF
Glutaric acid
Equilibrium -
2592.566
0.4781
6.31
Dihydrate




co-crystal
24 hours


L100129-29-9
Water
Glutaric acid
Equilibrium -
8.556
0.0016
4.27
Dihydrate




co-crystal
24 hours









Example 4. Salt Screening of Compound A

Anhydrous Form A of Compound A was used as the starting material in all the experiments.


Several different counter-ions (CIs) were used in an attempt to make salts with Compound A. Counter-ions Ca and Mg were used in two different ways, as their hydroxides, and also to produce hydroxide in-situ (in the form of CaO+Water or MgCl2+NaOH) to improve the chances of salt formation since the solubility of Ca and Mg hydroxides is poor in most of solvents/solvent systems. Several counter-ion systems used were: calcium hydroxide, calcium oxide (reacted with water in-situ), magnesium hydroxide, magnesium chloride (reacted with sodium hydroxide in-situ), sodium hydroxide, potassium hydroxide, ethanolamine, diethanolamine, triethanolamine, diethylamine, ethylenediamine, ethanol, 2-(diethylamine), choline hydroxide, L-arginine, L-histidine, L-lysine, meglumine. The IUPAC name for choline is 2-hydroxy-N,N,N-trimethylethan-1-aminium.


The initial salt screening was performed starting with 30 mg of Compound A and 1.1 equivalent of CI, both added as solutions in all cases (except in the case of the CIs Ca and Mg), to improve the salt formation. The vials were evaporated to dryness and slurries were made in five different process solvents to further improve the chances of salt formation. The slurries were filtered and the solids were analyzed by XRPD. Solids with unique patterns were dried and analyzed by XRPD again to see the effect of drying on solid form, all unique patterns were then exposed to saturated humidity environment at room temperature overnight and were re-subjected to XRPD. TGA/DSC was performed on all unique dry solids to identify their melting points and also to see the weight loss upon heating (to identify whether the solid is a solvate). H-NMR and HPLC analyses were performed on at least one version of the salt, to look for Compound A:CI stoichiometry, residual solvent content and also to look for signs of degradation of Compound A upon forming the salt.


All the counter-ions except ethylenediamine formed salts with Compound A. Use of ethylenediamine resulted in the degradation of Compound A, observed through HPLC. Scale-ups have been performed on all salts starting with ˜200 mg of Compound A. Two experiments were also conducted with 0.5 Eq Ca and Mg hydroxides to test the likelihood of forming a hemi-salt. The experiment with 0.5 Eq Calcium Hydroxide resulted in a new pattern that is different from the patterns obtained with 1 Eq. CI, indicating the formation of a calcium hemi-salt. However, magnesium hydroxide resulted in a solid that had the same pattern as its counter-part with 1 Eq CI.


The solubility of one form of each salt in Fasted State Simulated Gastric Fluid (FaSSGF), Fasted State Simulated Intestinal Fluid (FaSSIF) and Water was measured at 37° C. Normally, for each salt the solid form that showed relatively better stability in drying or humidity was scaled up and used in these experiments. In general, the effect of salt itself is more profound than individual solid forms of the salt when compared with the free molecule. However, the salts resulted in disproportionation in FaSSGF. The resulting solid was free molecule di-hydrate form by XRPD. However, the L-Histidine salt stayed intact. Several salts exhibited higher solubility in water. Notably, the sodium and potassium salts showed a solubility higher than 20 mg/mL in water. All the salts of Compound A obtained from the scale-up experiments were exposed to 75% relative humidity environment for 1 week for physical form stability assessment and changes were observed primarily in the case of the Hemi Ca and meglumine salts of Compound A when compared with their starting solids.


Preparation of Potassium Salt Acetic Acid Solvate of Compound A: To a 20 L Stainless Steel pressure vessel was charged Int. G (950 g, 1 equiv., 1.97 moles), KOAc (213 g, 1.1 equiv., 2.17 moles) and THF (14.25 L, 15 Vol.). The vessel was closed and pressurized with 10 psig of nitrogen, agitated with an overhead stirrer at 1000 rpm and heated to 90° C. During this time the pressure of the vessel rose to 41 psig, the reaction was continued at this pressure and at a temperature of 92° C. for approximately 12 h. The batch was then cooled to ambient temperature. The fine solids were filtered through an 18″ neutsche filter set up with a tight weave polypropylene cloth under nitrogen. The batch was filtered for a period of two hours. Following the initial filtration, the cake was slurry washed with 5 volumes of THF (4.75 L) four times, then was conditioned under nitrogen for 12 h, transferred to trays and dried in a vacuum oven (45° C.) for 2 days. The final isolated Compound A K-salt (18AK0164H) weighed 732 g (79%) yield with an overall purity of 99.60% AUC by UPLC with N/D MGl-100171(UPLC method) and <20 ppm of the dimeric impurity. NMR of the batch showed a 1:1.1 ratio of Compound A K-Salt to AcOH. The material showed 5.7 wt % of THF and a potency of 117%, uncorrected.


Conversion of Potassium-salt Acetic Acid Solvate of Compound A to THF solvate-hydrate: A 9 L carboy was charged with Compound A K-salt (620 g, 1 equiv., Lot #18AK0164H), K2CO3 (72 g, 0.4 equiv.), THF (1.24 L, 2 vol., bulk quality) and water (3.72 L, 6 vol.). The batch was agitated with an air motor stirrer to dissolve all the solids. After 15 minutes of stirring, the orange solution was transferred into a 10 L jacketed reactor via a 10 micron polypropylene filter using a transfer pump over 5 minutes. The lines were rinsed with DI water (465 mL)/THF (175 mL) mixture followed by a DI water rinse. The batch temperature was adjusted to 20° C. and acetic acid (0.165 L, 2.20 equiv.) charged to the batch over 1 h. Slurry formation started after about a quarter of the acid charged into the batch. After 2 h hold time, DI water (1.86 L, 3.00 vol.) was charged to the batch over 2 h at 20° C. and the slurry aged at 20° C. overnight. The batch was filtered and the filter cake washed with 1:5 THF/water (2×2 vol.) then dried in a vacuum oven at 45° C. for 12 h to give 570 g (86% yield) of Compound A THF Solvate (Lot #18AK0193C) of 99.81% purity by UPLC and with THF: H2O molar ratio of 0.56:0.67.


Preparation of Potassium Salt Tetrahydrofuran Solvate of Compound A from Compound A DMAc solvate (ASV-BO-194): To a 500-mL jacketed reactor equipped with mechanical stirrer, temperature probe, condenser and N2 inlet was charged Compound A DMAc solvate (24.5 g, 46.90 mmol, 1.00 equiv.), potassium carbonate (7.13 g, 51.59 mmol, 1.10 equiv.), MEK (245 mL, 10.0 vol.) and DI water (1.27 mL, 70.36 mmol, 1.50 equiv.). The slurry was heated to 45° C. over 1 hour and held at that temperature for 6 h. The batch was cooled to 20° C. over 3 h then held overnight at 20° C. The batch was then cooled to 5-10° C. then aged at that temperature for 1 h 45 min before being filtered to collect the solid product, which was manually smoothened on the filter and conditioned to deliquor the cake. The wet cake was slurried in THF (49 mL, 2.0 vol.) and aged for 30 min. The slurry was then filtered under vacuum. This THF-slurry procedure was repeated four more times, giving a total of 5×2 vol. THF washes. After the fifth wash, the batch was conditioned until no further THF emerged for the cake, then dried under vacuum at 40° C. (on the filter) to afford Compound A K salt (22.47 g, 85% uncorrected, Lot #ASV-BO-194-12). 1H NMR showed a Compound A K-salt/THF/water mole ratio: 1.00:0.22:0.51.


Conversion of Compound A Potassium salt to Compound A Tetrahydrofuran Solvate: Compound A potassium salt (16.50 g, 34.86 mmol, Lot #ASV-BP-6-8): Analysis: Compound A Potassium salt/DMAc/THF/water mole ratio: 1.00:0.02:0.16:0.65) on the filter frit was suspended in 60 mL of a THF/water solution prepared by mixing 99 mL (6 vol.) DI water and 33 mL (2 vol.) THF and the resultant slurry was stirred at room temperature, affording partial dissolution. The mixture was filtered, then the solid residue was suspended in ˜30 mL of the THF/water solution. Further dissolution occurred after agitation. This mixture was then filtered. Again the solid residue was suspended in ˜30 mL of the THF/water solution. The majority of the residual solid dissolved and this was then filtered. With the 500-mL jacketed reactor as the receiver under vacuum, the filtrate was transferred to the reactor through in-line filter (Whatman, 0.3 micron glass microfiber filter) into the reactor. During the transfer process, residual solid was observed in the filter line. This wash rinsed into the reactor with the aid of an additional 3:1 water/THF solution (1 vol.), followed by a DI water rinse (16 mL, 1.0 vol.). The batch temperature was adjusted to 20° C. and acetic acid (4.4 mL, 2.20 molar equiv.) charged to the batch over 30 min. After a 1.25 hour hold, DI water (50 mL, 3.00 vol.) was charged to the tan slurry batch over 2 h at 20° C. and the slurry aged at 20° C. overnight. The batch was filtered and the filter cake washed with 1:5 THF/Water (2×49 mL, 2×2 vol.) and dried in a vacuum oven at 41° C. to afford Compound A THF solvate (13.08 g, Lot #ASV-BP-18-3). 1H NMR analysis showed a Compound A: THF mole ratio of 1.00:0.95.









TABLE 2







Solubility












Exp Id



Solubility,
Resulting


(L100-)
CI
System
Area
mg/mL
form















L100110-

FaSSGF

0.1
Free form


3-2




Anhydrous


L100110-

FaSSGF

0.0067
Free form


19-5




dihydrate


110-86-1
Ca
FASSGF
19.122
0.0010
Dihydrate


110-86-4
Hemi
FASSGF
46.495
0.0025
Dihydrate



Ca


110-86-7
Mg
FASSGF
16.168
0.0009
Dihydrate


110-86-10
Na
FASSGF
14.098
0.0008
Dihydrate


110-86-13
K
FASSGF
22.635
0.0012
Dihydrate


110-86-16
Hemi
FASSGF
14.186
0.0008
Dihydrate



Mg


L100110-

FaSSIF

4.5
Free form


19-3




Anhydrous


L100110-

FaSSIF

2.9
Free form


19-6




dihydrate


110-86-2
Ca
FASSIF
10714.18
0.58
Amorphous


110-86-5
Hemi
FASSIF
14766.83
0.80
Hydrate of



Ca



Calcium salt







primarily







(2-D)


110-86-8
Mg
FASSIF
29216.45
1.59
Weakly







hydrate of







Magnesium







salt (3-C)


110-86-11
Na
FASSIF
45994.03
2.50
Dihydrate +







Extra *


110-86-14
K
FASSIF
50008.67
2.72
Dihydrate +







Extra *


110-86-17
Hemi
FASSIF
25888.9
1.41
Dihydrate



Mg


L100110-

Water

0.2
Free form


34-1




Anhydrous


L100110-

Water

0.1
Free form


19-4




dihydrate


110-86-3
Ca
Water
6486.92
0.35
Dihydrate


110-86-6
Hemi
Water
5912.01
0.32
Dihydrate



Ca


110-86-9
Mg
Water
12032.2
0.65
Dihydrate


110-86-12
Na-
Water
4615.626
0.25
Clear solution



100X



(solubility >



dil



25 mg/mL)


110-86-15
K-
Water
3921.429
0.21
Clear solution



100X



(solubility >



dil



21.3 mg/mL)


110-86-18
Hemi
Water
12371.22
0.67
Dihydrate



Mg









Example 5. Method for Obtaining XRPD Patterns

X-ray powder diffraction was done using a Rigaku MiniFlex 600. Samples were prepared on Si zero-return wafers. A typical scan is from 2θ of 4 to 30 degrees, with step size 0.05 degrees over five minutes with 40 kV and 15 mA. A high-resolution scan is from 2θ of 4 to 40 degrees, with step size 0.05 degrees over thirty minutes with 40 kV and 15 mA. Typical parameters for XRPD are listed below.


X-ray wavelength: Cu Kα1, 1.540598 Å; X-ray tube setting: 40 kV, 15 mA; Slit condition: variable+fixed slit system; Scan mode: continuous; Scan range (° 2TH): 4-30; Step size (° 2TH): 0.05; Scan speed (°/min): 5.


Tables 3-80 provide the major 2θ peaks and d-spacings for each of the crystalline forms disclosed herein.









TABLE 3







XRPD Peak positions of Form A














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.27
10.68878
1607
37



10.56
8.37014
4379
100



11.21
7.8855
1470
34



15.81
5.59963
660
15



16.43
5.38989
1085
25



17.72
5.00087
1342
31



18.45
4.80591
1166
27



18.75
4.72983
3414
78



22.30
3.98389
772
18



22.70
3.91432
1990
45



22.99
3.86513
1803
41



23.63
3.7622
1491
34



24.72
3.59862
2192
50



26.57
3.35183
274
6



27.49
3.24143
268
6



29.00
3.07664
323
7



29.52
3.02378
259
6



30.13
2.96403
922
21



32.04
2.79156
414
9



32.33
2.76688
423
10



35.25
2.54415
265
6

















TABLE 4







XRPD Peak positions of Form B














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.92
14.91066
5642
100



11.80
7.49683
4906
87



14.95
5.9199
298
5



17.46
5.07401
502
9

















TABLE 5







XRPD Peak positions of Form C














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.74
15.39153
2067
27



10.37
8.51986
480
6



11.45
7.72222
7751
100



15.42
5.74041
484
6



15.83
5.59273
350
5



16.70
5.30287
510
7



17.19
5.15513
654
8



17.69
5.01081
892
12



19.27
4.60254
1010
13



19.66
4.5128
787
10



21.41
4.14655
1028
13



22.95
3.87215
474
6



23.26
3.82149
550
7



24.28
3.66251
1400
18



24.55
3.62315
550
7



25.81
3.44968
545
7



26.25
3.39277
456
6



26.54
3.35528
304
4



28.82
3.09492
560
7



29.39
3.03701
441
6

















TABLE 6







XRPD Peak positions of Form D














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.52
16.00195
1507
17



8.52
10.37128
2171
24



11.01
8.03145
9097
100



16.49
5.3729
1420
16



17.19
5.15457
417
5



18.25
4.85666
1357
15



20.13
4.40764
781
9



21.03
4.22172
873
10



21.20
4.18738
1401
15



23.38
3.80249
412
5



24.03
3.70114
1717
19



25.61
3.47527
543
6



28.51
3.12802
425
5

















TABLE 7







XRPD Peak positions of Form E














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.13
12.39225
2144
100



8.77
10.08041
286
13



10.06
8.78695
362
17



10.81
8.17815
993
46



11.45
7.71869
207
10



12.05
7.34104
410
19



12.34
7.16766
853
40



14.14
6.26049
1196
56



14.73
6.01006
2013
94



15.01
5.89719
180
8



15.47
5.72179
431
20



16.09
5.50291
1244
58



16.67
5.31374
340
16



17.46
5.074
477
22



18.10
4.89713
730
34



18.64
4.7562
281
13



19.88
4.46331
689
32



20.19
4.39405
635
30



21.04
4.21909
431
20



21.22
4.18406
965
45



21.63
4.10543
290
14



22.65
3.92339
1035
48



22.90
3.88116
819
38



24.36
3.65154
620
29



24.66
3.60676
202
9



25.34
3.51261
1159
54



26.34
3.38042
453
21



27.10
3.28758
264
12



27.28
3.26673
187
9



27.77
3.20938
362
17



28.77
3.10029
167
8



29.32
3.04372
302
14



29.60
3.0152
263
12

















TABLE 8







XRPD Peak positions of Form F














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.28
12.13064
156
13



10.12
8.73306
925
78



10.41
8.4933
920
78



11.37
7.77672
292
25



13.91
6.36341
1179
100



16.20
5.46847
262
22



16.38
5.40798
331
28



17.05
5.19679
456
39



18.41
4.81512
162
14



18.90
4.69209
77
7



19.27
4.60216
129
11



20.94
4.23938
107
9



22.00
4.03681
396
34



22.51
3.94607
226
19



23.21
3.82963
73
6



23.81
3.73378
424
36



25.30
3.51739
74
6



25.85
3.44375
81
7



27.97
3.18771
227
19



29.53
3.02271
676
57



29.87
2.98899
83
7

















TABLE 9







XRPD Peak positions of Form G














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.71
11.45694
321
6



9.50
9.29954
3351
64



9.80
9.02038
264
5



10.70
8.26425
335
6



11.42
7.74427
573
11



12.91
6.85398
1574
30



13.06
6.77304
630
12



14.16
6.25129
331
6



15.32
5.78082
242
5



16.39
5.40376
342
7



16.72
5.29747
1503
29



16.93
5.23349
953
18



17.28
5.12802
1583
30



18.26
4.85418
367
7



18.95
4.67825
651
13



19.52
4.545
1324
25



20.20
4.39282
1156
22



20.75
4.27707
290
6



21.17
4.19266
499
10



22.94
3.87415
805
15



24.11
3.68838
255
5



24.39
3.64666
481
9



25.59
3.47856
5204
100



26.91
3.31057
389
7



28.26
3.15574
3363
65



29.10
3.06569
428
8

















TABLE 10







XRPD Peak positions of Form H














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















9.22
9.57947
7600
100



11.80
7.49312
869
11



12.08
7.32216
348
5



13.17
6.71738
1220
16



15.63
5.6641
712
9



16.77
5.28195
1040
14



18.40
4.81891
1013
13



19.34
4.58495
1245
16



19.78
4.48413
2744
36



21.18
4.19151
919
12



21.38
4.15202
481
6



22.78
3.90049
1111
15



22.98
3.8673
646
9



23.58
3.77027
2990
39



24.18
3.67827
830
11



25.11
3.54312
586
8



25.87
3.44169
1788
24



27.01
3.29814
349
5



28.00
3.18407
1677
22



28.55
3.12393
351
5



29.01
3.07499
439
6

















TABLE 11







XRPD Peak positions of Form I














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.15
17.14998
234
5



5.77
15.29146
796
18



9.30
9.50135
1010
23



9.61
9.1997
262
6



10.23
8.64045
4364
100



11.55
7.65485
1675
38



15.33
5.77637
313
7



21.87
4.06139
707
16



22.29
3.9856
337
8



23.80
3.73541
375
9

















TABLE 12







XRPD Peak positions of Form K














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.42
10.48827
4822
100



10.36
8.53374
365
8



11.37
7.77279
2754
57



12.96
6.8237
817
17



13.67
6.47249
872
18



14.45
6.12352
983
20



15.56
5.69196
922
19



16.42
5.39359
285
6



18.04
4.9142
354
7



18.90
4.69153
988
21



20.46
4.33778
392
8



21.14
4.19865
1327
28



21.56
4.11889
1755
36



23.65
3.75927
574
12



23.96
3.71172
688
14



24.94
3.5668
226
5



25.32
3.51471
366
8



25.57
3.48028
819
17



26.03
3.42028
694
14



26.20
3.39821
284
6



26.66
3.34086
646
13



27.20
3.27634
293
6



29.25
3.0512
417
9

















TABLE 13







XRPD Peak positions of Form L














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.11
10.88728
170
12



10.53
8.39548
319
23



11.49
7.6921
904
64



11.87
7.45234
1402
100



12.27
7.20569
141
10



15.21
5.82222
794
57



15.65
5.6584
470
34



16.04
5.52206
591
42



16.88
5.24756
455
32



17.10
5.18216
849
61



17.70
5.00658
143
10



18.43
4.81125
278
20



18.71
4.73921
332
24



21.32
4.16467
128
9



22.04
4.02938
413
29



22.81
3.89506
695
50



23.45
3.79117
422
30



24.08
3.69229
206
15



24.72
3.59921
266
19



26.37
3.37757
328
23



29.03
3.07381
91
6



29.49
3.02679
70
5

















TABLE 14







XRPD Peak positions of Form S + T














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.42
11.91007
448
36



8.80
10.03632
152
12



9.36
9.44359
213
17



9.50
9.30169
121
10



10.50
8.4148
1239
98



11.25
7.86127
336
27



12.39
7.14051
450
36



12.91
6.8537
74
6



14.30
6.18723
1260
100



14.74
6.00598
244
19



15.32
5.77749
93
7



15.81
5.60115
718
57



16.75
5.28996
268
21



17.13
5.17217
136
11



17.72
5.00027
285
23



18.09
4.90081
312
25



18.39
4.82152
979
78



19.61
4.52303
64
5



20.09
4.41564
594
47



20.54
4.32074
281
22



21.10
4.20647
315
25



21.89
4.05726
364
29



22.53
3.94393
78
6



23.23
3.8257
1048
83



24.40
3.64512
208
17



25.48
3.49332
514
41



26.86
3.31655
262
21



27.78
3.20839
237
19



28.33
3.14737
388
31

















TABLE 15







XRPD Peak positions of Form S














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.39
11.95027
327
24



8.73
10.11806
359
26



9.18
9.62337
51
4



10.54
8.38858
1266
91



11.27
7.84766
278
20



12.34
7.16511
693
50



14.40
6.14604
1384
100



14.74
6.00469
188
14



15.40
5.7507
132
10



15.79
5.60785
737
53



16.70
5.30353
445
32



17.11
5.17788
231
17



17.44
5.08208
328
24



17.72
5.00006
1026
74



18.14
4.887
755
55



18.42
4.81251
1157
84



19.69
4.50542
108
8



20.06
4.42329
788
57



20.57
4.31465
534
39



21.17
4.19318
482
35



21.93
4.04945
562
41



22.60
3.93034
270
20



23.27
3.81985
1220
88



24.40
3.64441
459
33



24.72
3.59863
94
7



25.46
3.49626
606
44



25.77
3.45466
303
22



26.23
3.39484
154
11



26.94
3.30691
258
19



27.83
3.20304
514
37



28.26
3.15557
390
28



28.52
3.1267
320
23



28.87
3.08971
307
22



29.85
2.99094
248
18

















TABLE 16







XRPD Peak positions of Form U














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.79
15.25652
539
34



8.43
10.4795
1496
94



9.62
9.18534
167
10



10.38
8.51392
205
13



11.36
7.78383
1264
79



11.62
7.61171
1590
100



12.93
6.8429
214
13



13.68
6.46712
219
14



14.47
6.1181
547
34



15.59
5.67796
299
19



16.46
5.38212
156
10



17.01
5.20821
152
10



17.40
5.09199
134
8



18.07
4.90473
107
7



18.45
4.80391
51
3



18.91
4.6903
562
35



19.33
4.58723
89
6



20.50
4.32989
83
5



21.08
4.21039
702
44



21.56
4.11765
757
48



22.49
3.94958
80
5



23.21
3.82911
96
6



23.60
3.76677
216
14



23.97
3.71009
381
24



25.02
3.55676
57
4



25.49
3.49201
408
26



26.01
3.42279
275
17



26.74
3.33167
112
7



27.22
3.27325
82
5



27.47
3.24404
102
6



29.21
3.05524
166
10

















TABLE 17







XRPD Peak positions of Form V














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.35
13.90226
263
32



7.33
12.05205
199
24



7.98
11.07194
69
8



8.21
10.76694
42
5



8.73
10.12354
67
8



10.56
8.37353
833
100



11.21
7.88582
166
20



11.83
7.47628
45
5



12.31
7.18552
214
26



13.44
6.58433
62
7



14.37
6.15866
214
26



14.68
6.02788
211
25



15.65
5.65959
683
82



16.51
5.36422
263
32



16.83
5.26498
271
33



17.68
5.0125
205
25



18.34
4.83358
291
35



18.74
4.73121
197
24



20.06
4.42375
160
19



21.10
4.20759
161
19



21.89
4.05761
167
20



22.27
3.98886
58
7



22.65
3.92216
96
12



23.18
3.83439
114
14



23.68
3.75429
189
23



24.70
3.60133
223
27



25.43
3.49941
148
18

















TABLE 18







XRPD Peak positions of Form W














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















10.06
8.78727
435
39



10.38
8.51351
871
78



10.67
8.28753
425
38



11.08
7.98164
129
11



11.35
7.78861
168
15



11.74
7.53486
360
32



11.89
7.4355
202
18



12.50
7.0739
177
16



13.90
6.36748
1002
89



14.69
6.02387
145
13



16.24
5.45326
195
17



16.34
5.42066
156
14



17.06
5.19266
278
25



18.39
4.82085
93
8



18.69
4.74351
114
10



18.91
4.68901
121
11



21.98
4.04154
163
15



22.51
3.94719
102
9



23.79
3.73785
154
14



24.43
3.64088
1123
100



25.90
3.43783
88
8



27.56
3.23416
157
14



27.95
3.18928
249
22



28.54
3.12464
183
16



29.48
3.02756
631
56

















TABLE 19







XRPD Peak positions of Form X














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.57
13.43846
567
25



8.20
10.77946
923
41



9.66
9.14872
1358
60



10.17
8.69028
2260
100



10.50
8.41944
1741
77



11.17
7.9177
694
31



11.66
7.58578
209
9



13.73
6.44445
126
6



15.72
5.63451
274
12



16.39
5.40323
572
25



17.65
5.02051
576
26



18.32
4.83943
416
18



18.67
4.74992
1124
50



18.98
4.67231
575
25



19.57
4.53358
277
12



21.92
4.05128
118
5



22.21
3.99855
259
11



22.60
3.93073
629
28



22.88
3.88323
383
17



23.36
3.80557
145
6



23.56
3.77329
346
15



24.65
3.60914
1075
48



26.79
3.32498
119
5



27.43
3.24868
116
5

















TABLE 20







XRPD Peak positions of Form Y














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.51
13.56982
688
100



8.16
10.82987
35
5



10.68
8.27907
77
11



12.96
6.82732
685
100



13.34
6.6317
164
24



14.25
6.21038
48
7



16.31
5.43026
87
13



17.79
4.98166
49
7



18.92
4.6866
78
11



19.47
4.55556
245
36



22.38
3.96985
61
9



24.19
3.6757
172
25



26.02
3.42199
51
7



28.73
3.1048
40
6

















TABLE 21







XRPD Peak positions of Form Z














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.83
15.15971
78
8



8.20
10.76811
132
13



8.42
10.49873
185
18



10.63
8.31858
1014
100



11.23
7.87332
261
26



11.55
7.65819
450
44



11.97
7.38713
231
23



14.34
6.1711
250
25



15.59
5.67832
363
36



16.17
5.47742
333
33



17.29
5.12414
18
2



17.64
5.02504
240
24



18.05
4.91146
480
47



18.67
4.74926
333
33



21.56
4.11901
82
8



22.32
3.979
159
16



22.85
3.88914
100
10



23.57
3.77129
79
8



24.11
3.68778
411
40



24.31
3.65902
588
58



24.62
3.61317
174
17



25.32
3.51459
47
5



26.72
3.33401
86
9



27.59
3.23007
56
6



28.73
3.1048
148
15



29.17
3.05883
80
8

















TABLE 22







XRPD Peak positions of Form α














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.26
12.16109
356
100



10.06
8.78569
326
92



10.36
8.53276
289
81



10.62
8.32671
236
66



11.23
7.87415
95
27



11.90
7.42931
149
42



13.85
6.3878
222
62



15.79
5.60772
37
10



16.46
5.38026
134
38



17.31
5.11738
80
22



21.91
4.05326
109
31



22.43
3.96018
121
34



24.12
3.68699
135
38



27.87
3.19903
36
10



29.39
3.03621
74
21

















TABLE 23







XRPD Peak positions of Form β














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.36
11.99797
1320
49



10.52
8.40273
2711
100



11.20
7.89298
415
15



12.35
7.15982
438
16



14.25
6.21004
2210
82



14.67
6.03184
636
23



15.26
5.80059
297
11



15.73
5.62778
1766
65



16.72
5.29918
179
7



17.11
5.17935
128
5



17.78
4.98572
288
11



18.07
4.90535
609
22



18.27
4.85076
2030
75



19.50
4.54843
164
6



20.11
4.41112
753
28



20.43
4.34446
967
36



21.00
4.2262
1300
48



21.75
4.08293
822
30



22.04
4.02993
393
15



22.49
3.95055
216
8



23.16
3.83754
1882
69



23.59
3.76772
159
6



24.49
3.63263
164
6



25.45
3.49699
449
17



26.35
3.37971
124
5



26.76
3.32885
670
25



27.78
3.209
280
10



28.02
3.18207
322
12



28.32
3.14845
534
20



28.61
3.1179
440
16



29.13
3.06289
126
5



29.50
3.02506
620
23

















TABLE 24







XRPD Peak positions of Form χ














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.53
10.36124
434
22



11.16
7.92161
1937
100



16.72
5.29732
132
7



17.11
5.17685
134
7



18.38
4.82347
408
21



19.16
4.62832
152
8



20.14
4.40478
306
16



21.19
4.18862
221
11



21.38
4.15259
360
19



21.52
4.12508
228
12



22.41
3.96478
242
12



23.15
3.83851
92
5



24.18
3.67732
254
13



25.91
3.4365
257
13



28.79
3.09877
114
6

















TABLE 25







XRPD Peak positions of Form δ














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















10.90
8.11024
290
100



12.14
7.28726
105
36



13.22
6.68946
41
14



14.44
6.12953
124
43



14.70
6.02007
35
12



17.39
5.09676
53
18



18.14
4.886
68
24



19.55
4.5377
63
22



20.21
4.38972
18
6



22.66
3.92145
42
14



24.48
3.63399
215
74



26.98
3.3026
87
30

















TABLE 26







XRPD Peak positions of a mixture of Form


C and a possible acetone solvate (Form ε)














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.73
15.41658
368
27



10.33
8.55984
128
10



11.41
7.75225
1348
100



11.67
7.57375
67
5



11.81
7.48709
87
6



15.39
5.75169
121
9



15.82
5.59883
75
6



16.60
5.33454
445
33



17.11
5.17708
115
9



17.64
5.02327
316
23



19.23
4.61134
109
8



19.62
4.52189
88
6



21.36
4.15655
161
12



21.84
4.06688
125
9



22.42
3.96186
87
6



22.88
3.88362
77
6



23.18
3.83343
263
19



23.60
3.76748
76
6



24.22
3.67188
249
18



25.74
3.45803
69
5



26.24
3.39337
111
8



28.74
3.10357
76
6

















TABLE 27







XRPD Peak positions of Form ϕ














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.95
12.69992
851
14



10.76
8.21266
220
4



11.06
7.99275
293
5



13.87
6.37839
1447
24



16.05
5.51793
346
6



16.48
5.3753
568
10



20.85
4.25639
5936
100



22.33
3.97807
944
16



22.71
3.91233
330
6



27.92
3.19264
1800
30



29.00
3.07696
627
11

















TABLE 28







XRPD Peak positions of Form η














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.87
12.85507
192
69



7.69
11.48644
209
75



12.06
7.32999
31
11



14.30
6.19077
65
23



16.32
5.42733
38
13



16.91
5.23944
76
27



18.17
4.87733
77
28



20.45
4.33961
220
79



21.08
4.21201
51
18



22.99
3.86525
279
100



23.85
3.72784
114
41



24.77
3.59205
49
18



25.46
3.49531
40
14



25.99
3.42553
34
12



26.56
3.35323
21
7



27.49
3.24178
49
18



28.17
3.16482
92
33



29.21
3.05472
31
11

















TABLE 29







XRPD Peak positions of Form λ














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.85
9.97936
57
6



10.64
8.30722
834
94



11.24
7.86653
227
26



12.01
7.36601
291
33



14.33
6.1747
271
30



15.59
5.67972
261
29



16.20
5.46571
704
79



17.31
5.11846
206
23



17.60
5.03393
333
37



18.01
4.92036
528
59



19.56
4.53532
82
9



20.10
4.41393
151
17



22.34
3.97569
262
29



22.81
3.89618
50
6



24.26
3.66562
889
100



26.78
3.32625
117
13



27.68
3.22066
76
9



28.75
3.10234
183
21

















TABLE 30







XRPD Peak positions of the glutaric acid cocrystal














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.15
10.84284
194
7



9.74
9.07047
1061
39



10.78
8.1976
1982
74



11.04
8.00901
1835
68



12.17
7.26756
1645
61



14.59
6.06438
312
12



16.06
5.51456
1314
49



17.02
5.20511
799
30



17.62
5.03029
492
18



18.71
4.73919
326
12



19.15
4.63132
934
35



19.61
4.52359
389
14



21.47
4.13454
468
17



21.86
4.06253
1453
54



23.06
3.85374
2689
100



24.51
3.62834
470
17



25.13
3.54038
267
10



26.63
3.34464
216
8



27.10
3.28755
407
15



27.43
3.24858
267
10



27.99
3.1849
151
6



29.18
3.05837
321
12



32.84
2.72493
360
13



33.50
2.67298
206
8



37.17
2.41685
125
5

















TABLE 31







XRPD Peak positions of the Calcium salt of Compound


A from the experiment L100110-68-1 (Form 1-A)














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.55
19.41373
1603
100



6.53
13.5336
987
62



7.25
12.18703
414
26



8.52
10.36821
324
20



23.37
3.80366
105
7



25.00
3.5593
121
8



29.51
3.02493
199
12

















TABLE 32







XRPD Peak positions of the Calcium salt of Compound


A from the experiment L100110-68-3-Wet (Form 1-B)














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.02
21.95006
2299
100



6.99
12.64126
96
4



7.88
11.21031
473
21



8.73
10.12473
92
4



15.86
5.58166
95
4

















TABLE 33







XRPD Peak positions of the Calcium salt of Compound


A from the experiment L100110-68-8 (Form 2-B)














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.94
14.87269
304
99



6.90
12.80769
162
53



7.79
11.3457
307
100



9.57
9.23139
19
6



11.98
7.38409
53
17



13.57
6.52017
24
8



14.19
6.2359
19
6



20.38
4.35414
37
12



25.25
3.52448
21
7

















TABLE 34







XRPD Peak positions of the Calcium salt of Compound


A from the experiment L100110-68-10 after exposing


to saturated humidity environment (Form 2-D)














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.51
19.57407
76
13



5.76
15.33611
118
21



7.44
11.87012
38
7



8.68
10.18068
496
88



10.79
8.1896
62
11



11.38
7.76999
241
43



14.12
6.26516
150
27



16.77
5.28335
563
100



18.57
4.7754
27
5



20.60
4.30778
38
7



21.61
4.1095
29
5



23.41
3.79764
325
58



26.29
3.38736
83
15



28.29
3.15205
136
24



29.50
3.02559
272
48

















TABLE 35







XRPD Peak positions of the Magnesium salt of Compound


A from the experiment L100110-68-11-Dry (3-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.06
21.73785
9654
100



8.04
10.98764
1168
12



8.80
10.03642
1012
10



9.27
9.52989
2027
21



11.00
8.03807
898
9



15.80
5.60268
1284
13



18.01
4.9209
740
8



18.64
4.75617
1288
13



19.37
4.5788
722
7



22.02
4.03289
1852
19



23.39
3.79958
899
9



38.06
2.36248
1271
13

















TABLE 36







XRPD Peak positions of the Magnesium salt of Compound


A from the experiment L100110-68-11 upon exposing it


to saturated humidity environment at room temperature














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.69
15.51348
470
31



8.87
9.96636
1541
100



10.87
8.132
244
16



11.46
7.71218
543
35



13.20
6.70116
245
16



14.24
6.21326
139
9



16.55
5.35354
291
19



18.60
4.76784
468
30



20.55
4.31771
74
5



21.85
4.06444
224
15



23.62
3.76347
299
19



24.41
3.64333
138
9



26.35
3.37926
301
20



27.76
3.21078
187
12



28.59
3.11989
112
7

















TABLE 37







XRPD Peak positions of the Magnesium salt of Compound A


from the experiment L100110-68-13 before drying (3-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.47
16.13545
131
25



6.93
12.73634
496
96



9.11
9.70297
30
6



10.72
8.24296
519
100



13.01
6.79699
32
6



14.92
5.93135
45
9



15.19
5.82791
62
12



16.08
5.50592
257
50



18.67
4.7492
302
58



20.63
4.30185
25
5



21.43
4.143
86
17

















TABLE 38







XRPD Peak positions of the Magnesium salt of Compound A from


the experiment L100110-68-13, dried after deliquescing (3-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















9.63
9.17326
682
100



10.75
8.22179
85
12



13.70
6.46016
118
17



15.70
5.63949
31
5



17.16
5.16179
54
8



18.63
4.75863
217
32



19.75
4.49151
73
11



21.70
4.09288
280
41



22.23
3.99595
32
5



26.27
3.38972
195
29

















TABLE 39







XRPD Peak positions of the Magnesium salt of Compound A from


the experiment L100110-68-17, before drying (Form 4-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.13
17.20435
705
100



6.04
14.6144
59
8



10.04
8.80367
213
30



15.42
5.74009
68
10



18.32
4.8385
43
6



27.44
3.24795
156
22

















TABLE 40







XRPD Peak positions of the Magnesium salt of Compound A from


the experiment L100110-68-20, before drying (Form 4-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















9.97
8.86232
20
10



11.08
7.97695
87
45



16.54
5.35375
76
39



18.57
4.7754
20
10



22.45
3.95625
67
34



27.48
3.2426
196
100

















TABLE 41







XRPD Peak positions of the Magnesium salt of Compound A from


the experiment L100110-68-20, after drying (Form 4-E).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.92
9.90545
140
54



10.59
8.35011
258
100



15.33
5.77434
101
39



15.88
5.57481
47
18



17.79
4.98152
116
45



20.45
4.33966
38
15



21.73
4.08729
170
66



27.39
3.25357
128
50



28.23
3.15834
29
11

















TABLE 42







XRPD Peak positions of the Sodium salt of Compound


A from the experiment L100110-68-21 (Form 5-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.04
21.82961
811
100



8.02
11.01682
125
15



10.11
8.74579
168
21

















TABLE 43







XRPD Peak positions of the Sodium salt of Compound


A from the experiment L100110-68-24 (Form 5-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.06
21.73785
474
65



7.76
11.38701
577
79



10.33
8.55811
733
100



20.83
4.26167
114
16



25.83
3.44676
56
8

















TABLE 44







XRPD Peak positions of the Sodium salt of Compound


A from the experiment L100110-68-25 (Form 5-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.72
15.42652
275
61



6.48
13.63258
448
100



8.49
10.40558
208
46

















TABLE 45







XRPD Peak positions of the Sodium salt of Compound A from the


experiment L100110-68-25 after humidity exposure that resulted


in substantial improvement in crystallinity (Form 5-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.15
17.15855
10385
53



7.30
12.09301
19470
100



10.22
8.64785
1577
8



14.57
6.07274
8966
46



17.02
5.20512
2386
12



20.50
4.32901
3157
16



21.90
4.05459
2197
11

















TABLE 46







XRPD Peak positions of the Potassium salt of Compound A from


the experiment L100110-68-26 after drying (Form 6-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.13
21.36074
1068
100



8.42
10.49468
47
4



11.97
7.38706
145
14

















TABLE 47







XRPD Peak positions of the Potassium salt of Compound


A from the experiment L100110-68-29 (Form 6-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.15
21.25539
1912
55



6.73
13.11434
3466
100



9.54
9.26631
133
4



11.90
7.43037
150
4



17.83
4.96945
257
7



21.48
4.13371
129
4



31.42
2.84461
229
7

















TABLE 48







XRPD Peak positions of the Potassium salt of Compound A from


the experiment L100110-68-30 after drying (Form 6-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.24
10.71704
441
56



10.91
8.10348
299
38



22.17
4.00659
788
100



30.78
2.9028
47
6



38.86
2.31546
65
8

















TABLE 49







XRPD Peak positions of the Potassium salt of Compound


A from the experiment L100110-68-30-H (Form 6-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.91
9.9141
789
60



11.81
7.4864
258
20



14.09
6.28163
103
8



15.75
5.62047
163
12



17.30
5.12046
117
9



19.96
4.44385
134
10



21.98
4.03979
1312
100



23.22
3.82704
92
7



24.18
3.67732
190
15



25.92
3.43477
66
5



27.09
3.28884
95
7



28.46
3.13404
154
12

















TABLE 50







XRPD Peak positions of the Ethanolamine salt of Compound


A from the experiment L100110-68-31 (Form 7-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.04
17.51318
380
10



8.17
10.80774
1193
32



10.09
8.75893
3735
100



11.04
8.00809
1024
27



13.82
6.40101
279
7



14.94
5.92583
202
5



16.40
5.39914
429
11



18.61
4.76291
1020
27



19.86
4.46687
200
5



22.69
3.91561
732
20



23.49
3.78349
2
0



25.29
3.51929
153
4



25.59
3.47759
264
7



26.88
3.31475
281
8



27.76
3.21134
629
17



28.05
3.17894
135
4

















TABLE 51







XRPD Peak positions of the Ethanolamine salt of Compound A


from the experiment L100110-68-32 after drying (Form 7-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















8.41
10.50243
8644
94



10.21
8.65795
9198
100



13.15
6.72755
1713
19



16.31
5.42927
671
7



16.83
5.26504
3247
35



18.93
4.68344
605
7



20.32
4.36721
1177
13



20.68
4.29139
679
7



21.16
4.19515
573
6



24.61
3.61417
1034
11



25.04
3.55349
3272
36



26.03
3.41984
1693
18



27.74
3.21305
1965
21



29.24
3.0519
535
6



29.89
2.98695
909
10



34.12
2.62598
1233
13



35.63
2.51757
389
4

















TABLE 52







XRPD Peak positions of the Diethanolamine salt of Compound


A from the experiment L100110-68-36 (Form 8-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.62
13.34209
1970
100



7.99
11.0497
418
21



11.86
7.45418
1593
81



12.52
7.06306
207
11



14.14
6.25715
579
29



15.17
5.83499
86
4



16.06
5.51371
134
7



18.72
4.73628
122
6



19.29
4.59738
72
4



19.94
4.44815
226
12



21.27
4.17306
253
13



21.69
4.09351
507
26



22.15
4.00999
371
19



22.85
3.88855
653
33



23.65
3.75859
177
9



24.57
3.6204
701
36



25.65
3.46998
353
18



26.12
3.4093
484
25



28.47
3.13287
306
16

















TABLE 53







XRPD Peak positions of the Diethanolamine salt of Compound


A from the experiment L100110-68-38 (Form 8-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.80
12.98593
10954
100



9.60
9.20224
8979
82



11.35
7.78675
1374
13



13.18
6.71147
4006
37



16.36
5.41348
2290
21



17.99
4.92574
1324
12



18.20
4.87048
400
4



18.81
4.71456
544
5



19.20
4.61866
4211
38



20.14
4.40634
2275
21



20.44
4.34166
4914
45



21.38
4.15282
9437
86



22.78
3.90121
2427
22



24.42
3.64182
5115
47



24.83
3.58325
853
8



25.69
3.46464
909
8



26.64
3.34358
1757
16



27.10
3.2873
2199
20



27.74
3.21354
3515
32



28.91
3.08603
1134
10



29.15
3.0608
1739
16



29.56
3.01962
411
4



30.38
2.93984
908
8



30.89
2.89233
1139
10



31.61
2.82802
396
4



33.03
2.70994
837
8



34.12
2.62559
437
4



34.96
2.56427
466
4



36.59
2.45401
462
4

















TABLE 54







XRPD Peak positions of the Diethanolamine salt of Compound


A from the experiment L100110-68-36 after subjecting


to saturated humidity environment at RT (Form 8-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.91
17.97329
564
30



5.09
17.35412
1877
100



6.51
13.5755
411
22



7.67
11.51328
144
8



7.91
11.17124
76
4



8.50
10.39291
193
10



8.95
9.87429
975
52



9.73
9.07873
1204
64



10.12
8.73284
1222
65



11.91
7.4269
236
13



15.30
5.78658
1091
58



17.75
4.99325
123
7



18.27
4.85216
112
6



19.25
4.60735
117
6



20.50
4.32805
171
9



21.17
4.19262
236
13



21.57
4.11737
190
10



23.00
3.86345
882
47



23.50
3.78192
330
18



25.66
3.46911
132
7



26.14
3.40577
149
8



29.60
3.01547
79
4

















TABLE 55







XRPD Peak positions of the Diethanolamine salt of Compound A


from the experiment L100110-68-40 before drying (Form 8-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.75
13.08575
1872
100



8.01
11.02396
143
8



12.11
7.30222
935
50



13.97
6.3334
341
18



14.46
6.12158
259
14



18.63
4.75847
81
4



20.24
4.38347
214
11



20.97
4.23276
143
8



21.57
4.11681
207
11



22.12
4.01613
209
11



22.64
3.92409
308
16



24.43
3.64133
167
9



25.12
3.54221
213
11



25.98
3.42708
321
17



28.69
3.10878
159
8




















Table 56. XRPD Peak positions of the Diethanolamine salt


of Compound A from the experiment L100110-68-40 after drying


followed by exposure to saturated humidity environment


(Form 8-E) (Essentially Form 8-B with extra peaks).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.88
18.1047
1278
6



6.84
12.91887
6782
31



9.62
9.1878
22046
100



11.38
7.76763
993
5



18.02
4.9176
841
4



19.23
4.61183
4971
23



20.45
4.3397
5404
25



25.75
3.45762
1125
5

















TABLE 57







XRPD Peak positions of the Triethanolamine salt of Compound


A from the experiment L100110-68-42 (Form 9-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.03
21.90272
937
15



7.75
11.39109
4762
76



10.35
8.53952
6300
100



10.86
8.14284
4254
68



11.87
7.44941
762
12



12.21
7.24113
1718
27



13.57
6.52209
321
5



14.53
6.08924
1901
30



15.47
5.72377
1291
20



15.81
5.60181
2918
46



16.68
5.3095
1210
19



17.11
5.17765
503
8



17.50
5.06374
677
11



17.94
4.93979
891
14



18.49
4.79524
2588
41



19.67
4.51008
925
15



20.59
4.30963
3642
58



21.36
4.15644
4267
68



21.73
4.08589
1844
29



22.46
3.95474
3714
59



22.76
3.90369
1071
17



23.85
3.72826
1461
23



24.05
3.69751
2086
33



24.45
3.63748
1062
17



26.06
3.41707
548
9



26.40
3.37284
1402
22



26.62
3.3462
1189
19



26.95
3.30598
915
15



27.35
3.2588
982
16



28.09
3.17414
853
14



28.60
3.11887
351
6



29.29
3.04674
1189
19



29.73
3.00264
1600
25



30.25
2.9524
356
6



30.90
2.89137
415
7



31.85
2.80749
552
9



32.21
2.77673
451
7



32.64
2.74161
284
5



33.90
2.64223
512
8



34.61
2.58931
514
8



36.06
2.48854
331
5



38.29
2.34861
277
4

















TABLE 58







XRPD Peak positions of the Triethanolamine salt of Compound


A from the experiment L100110-68-44 (Form 9-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.38
11.96556
5277
65



9.80
9.01543
390
5



10.92
8.09534
4218
52



12.23
7.23046
1276
16



12.88
6.86771
988
12



14.69
6.02464
1947
24



15.42
5.74026
2803
35



16.32
5.42746
3141
39



16.85
5.25854
990
12



17.76
4.99134
563
7



18.10
4.89692
874
11



18.75
4.72947
326
4



19.12
4.63815
1555
19



19.77
4.48789
1384
17



20.35
4.35999
1615
20



21.48
4.13368
8083
100



22.59
3.9329
4036
50



23.51
3.78062
794
10



24.05
3.69679
641
8



25.98
3.42691
1015
13



26.36
3.37815
1606
20



26.64
3.34315
1666
21



27.33
3.26019
830
10



29.61
3.01471
1116
14



30.13
2.96366
976
12



30.80
2.90049
437
5



31.62
2.82771
825
10



34.05
2.6308
453
6

















TABLE 59







XRPD Peak positions of the Triethanolamine salt of Compound


A from the experiment L100110-68-41 after drying and subjecting


it to saturated humidity environment at RT (Form 9-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.65
18.97718
27633
100



5.90
14.98024
2297
8



9.24
9.56562
16687
60



11.76
7.52087
1280
5



13.83
6.40025
8526
31



23.14
3.8407
1237
4

















TABLE 60







XRPD Peak positions of the Triethanolamine salt of Compound


A from the experiment LI00110-68-44 after drying, subjecting


it to saturated humidity environment at RT (Form 9-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.67
18.91096
1331
82



5.89
14.98856
1237
76



7.51
11.7633
346
21



8.31
10.62569
1619
100



8.89
9.94186
373
23



9.23
9.57156
740
46



11.39
7.76057
466
29



11.72
7.54559
656
41



12.46
7.1005
1137
70



12.81
6.90253
125
8



13.81
6.40901
327
20



15.24
5.80955
238
15



16.65
5.32042
390
24



17.72
5.00127
206
13



18.76
4.72621
189
12



20.30
4.37048
137
8



20.57
4.31356
173
11



21.04
4.21902
100
6



21.78
4.07639
87
5



22.53
3.94262
694
43



23.28
3.81761
263
16



24.42
3.64237
134
8



24.81
3.58583
87
5



26.11
3.41021
170
11



26.82
3.32193
189
12



28.09
3.1742
118
7



29.33
3.04314
248
15

















TABLE 61







XRPD Peak positions of the Triethanolamine salt of Compound


A from the experiment L100110-68-45 (Form 9-E).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.89
11.19788
3398
100



10.50
8.41706
972
29



11.23
7.87285
1747
51



11.78
7.50953
413
12



12.57
7.03403
862
25



13.86
6.38651
245
7



15.76
5.61774
356
10



16.51
5.36604
840
25



17.38
5.09913
1466
43



19.00
4.66637
1515
45



20.25
4.38149
405
12



20.89
4.24944
216
6



21.36
4.1573
494
15



22.21
3.99951
396
12



23.57
3.77205
680
20



24.16
3.68124
219
6



25.21
3.52941
301
9



26.39
3.37443
210
6



27.11
3.28616
512
15



28.88
3.08853
179
5



29.63
3.01231
124
4



30.16
2.96059
323
10



31.73
2.81796
170
5



36.31
2.47234
122
4

















TABLE 62







XRPD Peak positions of the Diethylamine salt of Compound


A from the scale-up experiment L100110-85-9 (Form 10-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.02
12.58251
84
8



8.40
10.52239
130
12



9.04
9.77431
441
40



9.81
9.00901
534
49



11.66
7.58328
45
4



12.10
7.3101
163
15



12.34
7.16522
406
37



13.70
6.45893
113
10



14.76
5.99498
1091
100



18.56
4.77732
57
5



19.87
4.46419
86
8



20.91
4.24543
55
5



21.72
4.08808
63
6



23.14
3.84024
138
13



24.25
3.66686
182
17



26.36
3.3785
73
7

















TABLE 63







XRPD Peak positions of the Diethylamine salt of Compound A from


the experiment L100110-68-46 followed by drying (Form 10-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.16
14.34705
912
34



8.99
9.83108
247
9



10.01
8.83204
1422
53



12.40
7.13058
379
14



14.31
6.18306
1150
43



15.09
5.86776
2671
100



17.07
5.1892
502
19



20.95
4.23783
590
22



21.75
4.08304
121
5



23.25
3.82268
800
30



24.11
3.68828
134
5



26.34
3.3803
169
6



27.86
3.20014
121
5



29.00
3.07656
178
7



30.40
2.93771
240
9



32.92
2.71843
133
5

















TABLE 64







XRPD Peak positions of the Diethylamine salt of Compound A from


the experiment L100110-68-49 (Form 10-B + extra minor peaks).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.28
14.0724
600
34



7.10
12.44613
841
47



8.32
10.62079
630
36



8.96
9.86277
1394
79



9.71
9.1011
872
49



12.00
7.36715
1020
57



12.31
7.18294
417
23



13.58
6.5176
347
20



14.74
6.0039
1774
100



18.43
4.81058
289
16



21.76
4.0812
423
24



22.93
3.87536
363
20



24.16
3.68081
251
14



25.12
3.54196
268
15



26.24
3.39324
161
9



28.54
3.12516
137
8



29.68
3.00722
159
9



30.68
2.91168
159
9



31.58
2.83112
68
4

















TABLE 65







XRPD Peak positions of the Ethanol-2-diethylamine salt of


Compound A from the experiment L100110-68-56 (Form 12-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.82
18.33542
3585
99



9.61
9.19525
2275
63



10.44
8.46428
695
19



11.23
7.87607
2935
81



11.52
7.67196
778
21



12.26
7.21549
737
20



13.40
6.60445
557
15



14.01
6.31512
3624
100



14.47
6.11723
1416
39



14.85
5.95909
2576
71



15.81
5.60104
568
16



16.47
5.37784
286
8



17.68
5.01192
1555
43



18.90
4.69128
1993
55



19.28
4.59981
385
11



19.72
4.49874
1176
32



19.95
4.44596
1335
37



20.31
4.36967
1503
41



20.93
4.24163
1275
35



21.59
4.11201
2345
65



22.65
3.92246
660
18



23.15
3.8388
1501
41



23.75
3.74355
655
18



24.55
3.62312
357
10



25.00
3.55876
1101
30



25.65
3.46996
934
26



26.10
3.41201
978
27



26.51
3.35989
685
19



27.27
3.26723
976
27



28.16
3.16683
996
27



28.62
3.11687
363
10



29.45
3.03042
352
10



30.70
2.90964
438
12



31.82
2.80995
355
10



32.78
2.73024
333
9



33.31
2.6875
161
4



34.38
2.60642
263
7



35.17
2.54987
231
6



38.46
2.33859
194
5

















TABLE 66







XRPD Peak positions of the Ethanol-2-diethylamine salt of


Compound A from the experiment L100110-68-60 (Form 12-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.79
18.44824
1410
100



9.10
9.70753
729
52



12.32
7.17607
92
7



15.61
5.67238
85
6



16.39
5.40362
298
21



22.87
3.8849
82
6



26.80
3.32352
57
4

















TABLE 67







XRPD Peak positions of the Ethanol-2-diethylamine salt of


Compound A from the experiment L100110-68-60 after subjecting


it to saturated humidity environment at RT (Form 12-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.40
20.07702
83
34



5.86
15.08102
246
100



8.63
10.23481
99
40



9.01
9.80211
27
11



11.78
7.50911
142
58



15.40
5.74964
12
5



17.67
5.01424
18
7



19.73
4.49648
11
4



22.53
3.94246
32
13



23.73
3.74702
20
8

















TABLE 68







XRPD Peak positions of the Choline hydroxide salt of Compound


A from the experiment L100110-68-64 (Form 13-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.36
16.48646
1214
27



8.62
10.25278
3256
74



10.53
8.39598
1222
28



12.28
7.19929
794
18



14.64
6.04675
1023
23



14.88
5.95004
2588
58



17.53
5.05496
4427
100



18.49
4.79444
1660
37



19.02
4.66273
343
8



20.04
4.42651
1484
34



20.83
4.26197
2404
54



21.60
4.11151
1316
30



24.02
3.70146
657
15



25.40
3.50333
1003
23



26.71
3.33426
529
12



27.57
3.23237
311
7



28.37
3.14285
583
13



28.96
3.08034
869
20



30.46
2.93267
238
5



31.55
2.8338
201
5



32.98
2.71412
227
5



33.94
2.63946
263
6



36.61
2.45263
159
4

















TABLE 69







XRPD Peak positions of the L-Arginine salt of Compound


A from the experiment L100110-68-66 (Form 14-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.99
12.62765
5901
100



7.93
11.13591
5751
97



9.74
9.07485
2946
50



13.38
6.6107
5532
94



13.98
6.32999
1790
30



14.91
5.93592
1086
18



16.13
5.49144
2027
34



18.25
4.85601
1740
29



18.83
4.70865
546
9



19.42
4.56677
1325
22



19.79
4.48263
2178
37



20.49
4.33104
571
10



20.86
4.25455
1613
27



22.21
3.99997
454
8



22.67
3.91846
1078
18



23.90
3.72061
2028
34



25.31
3.51592
502
9



25.84
3.44493
345
6



26.71
3.33517
238
4



28.20
3.16206
866
15



31.08
2.87541
334
6



32.61
2.74333
251
4



33.24
2.69275
214
4



36.01
2.49181
262
4

















TABLE 70







XRPD Peak positions of the L-Arginine salt of Compound


A from the experiment L100110-68-68 (Form 14-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.02
12.57436
2969
55



7.99
11.05088
5413
100



9.14
9.66904
2398
44



9.59
9.21739
2602
48



13.26
6.67243
2652
49



13.97
6.33294
1452
27



15.13
5.85036
466
9



15.79
5.60747
960
18



16.49
5.37226
1496
28



18.48
4.79717
3018
56



19.20
4.61895
518
10



20.20
4.39182
1717
32



21.00
4.22667
1198
22



21.30
4.16729
594
11



22.40
3.96495
458
8



22.99
3.86461
1490
28



23.65
3.75831
1403
26



24.00
3.70474
1702
31



25.29
3.51851
415
8



26.07
3.41562
586
11



26.45
3.36662
531
10



27.36
3.25689
331
6



28.08
3.17565
373
7



28.57
3.1222
880
16



30.35
2.94229
355
7



31.20
2.86416
636
12



34.23
2.6172
281
5

















TABLE 71







XRPD Peak positions of the L-Arginine salt of Compound


A from the experiment L100110-68-69 (Form 14-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















4.10
21.52352
143
21



5.50
16.05026
669
100



7.05
12.52711
358
53



7.50
11.78467
259
39



8.91
9.91629
198
30



9.48
9.32647
76
11



10.68
8.2747
106
16



12.42
7.12201
59
9



15.85
5.58547
451
67



16.32
5.42607
38
6



16.77
5.28335
34
5



18.09
4.90053
129
19



19.40
4.57114
65
10



20.30
4.37134
187
28



21.70
4.0916
132
20



24.26
3.66567
117
17



28.05
3.17894
91
14

















TABLE 72







XRPD Peak positions of the L-Arginine salt of Compound A from


the experiment L100110-68-70 after drying (Form 14-E).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.95
12.7084
765
100



9.03
9.78635
695
91



9.68
9.12658
84
11



10.90
8.11047
406
53



13.34
6.63191
139
18



14.20
6.23263
267
35



14.55
6.08369
411
54



19.45
4.55982
517
68



20.03
4.4292
71
9



21.07
4.21385
144
19



23.36
3.80567
188
25



24.09
3.69069
57
7



26.59
3.34973
78
10



28.41
3.13937
258
34

















TABLE 73







XRPD Peak positions of the L-Histidine salt of Compound A


from the experiment L100110-68-71 after drying (Form 15-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















6.91
12.79054
3644
100



7.72
11.4422
1797
49



7.91
11.17124
655
18



9.43
9.36766
287
8



10.36
8.53008
167
5



12.06
7.33127
186
5



13.80
6.41071
241
7



14.34
6.17179
724
20



15.30
5.78822
183
5



16.91
5.23987
241
7



18.19
4.87352
253
7



18.96
4.67646
2265
62



20.38
4.35314
1019
28



21.14
4.19991
1195
33



22.17
4.00622
212
6



22.47
3.95426
166
5



23.03
3.85829
1255
34



24.01
3.70303
825
23



25.53
3.48635
140
4



26.00
3.42485
151
4



27.51
3.2394
199
5



28.21
3.16054
344
9



29.25
3.05057
164
5



29.84
2.99196
149
4

















TABLE 74







XRPD Peak positions of the L-Histidine salt of Compound A from


the experiment L100110-68-71 after drying, followed by subjecting


it to saturated humidity environment at RT (Form 15-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.88
15.0202
2317
82



9.49
9.31552
244
9



10.16
8.69923
237
8



11.75
7.52658
1143
41



13.13
6.73636
126
4



14.57
6.07544
394
14



15.28
5.79368
137
5



18.96
4.67646
2818
100



21.16
4.19451
672
24



22.61
3.92956
349
12



23.25
3.82284
232
8



24.15
3.68265
477
17



25.25
3.52418
370
13



26.30
3.38575
455
16



26.68
3.33798
311
11



27.52
3.23858
218
8

















TABLE 75







XRPD Peak positions of the L-Histidine salt of Compound A


from the experiment L100110-68-72 after drying (Form 15-C).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















9.50
9.30504
142
12



15.32
5.77725
292
25



18.99
4.66879
1169
100



21.14
4.19944
1171
100



22.12
4.01482
91
8



22.54
3.94098
115
10



24.16
3.68131
700
60



29.88
2.98821
226
19

















TABLE 76







XRPD Peak positions of the L-Histidine salt of Compound A from


the experiment L100110-68-75 before drying (Form 15-D).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















9.40
9.40551
76
13



10.35
8.5374
111
19



11.47
7.7104
369
64



15.33
5.77342
347
60



15.85
5.5869
244
42



16.82
5.26627
294
51



18.97
4.67518
525
91



21.19
4.1889
578
100



21.85
4.06511
168
29



24.17
3.67863
399
69



29.87
2.98917
64
11

















TABLE 77







XRPD Peak positions of the L-Histidine salt of Compound


A from the experiment L100110-68-75 (Form 15-E).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















5.94
14.87186
633
24



6.36
13.89127
1184
45



8.05
10.97998
643
25



9.50
9.3007
339
13



10.50
8.41928
2219
85



11.16
7.91943
394
15



11.78
7.50728
267
10



14.59
6.06685
484
19



15.37
5.7587
969
37



17.67
5.01613
227
9



18.14
4.88751
481
18



18.66
4.75027
99
4



18.98
4.67148
1986
76



21.15
4.19792
2605
100



22.54
3.94077
262
10



24.13
3.68452
1676
64



24.63
3.61185
915
35



25.38
3.50607
820
31



29.87
2.98839
397
15



32.16
2.78137
215
8



33.82
2.648
121
5



37.76
2.38075
524
20

















TABLE 78







XRPD Peak positions of the L-Lysine salt of Compound


A from the experiment L100110-68-79 (Form 16-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.12
12.40428
932
18



8.70
10.15282
2290
43



9.22
9.58923
5315
100



11.33
7.80251
1586
30



14.21
6.22779
448
8



15.89
5.57162
223
4



17.04
5.19801
2428
46



17.42
5.08617
708
13



18.36
4.82873
967
18



19.06
4.65208
1000
19



20.42
4.34592
952
18



21.45
4.14016
310
6



21.89
4.0566
788
15



22.56
3.93721
263
5



23.60
3.76691
399
8



24.02
3.70199
454
9



24.80
3.58779
1433
27



25.73
3.45912
805
15



27.00
3.3
333
6



27.52
3.23908
232
4



28.26
3.15499
204
4



34.15
2.62379
258
5



35.10
2.55487
214
4

















TABLE 79







XRPD Peak positions of the Meglumine salt of Compound


A from the experiment L100110-68-82 (Form 17-A).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.34
12.03989
2788
100



9.83
8.99314
155
6



10.72
8.2425
504
18



12.98
6.81658
138
5



15.56
5.69055
1896
68



16.77
5.28219
432
15



18.44
4.80783
433
16



19.24
4.60992
416
15



21.59
4.11228
1376
49



22.25
3.99202
580
21



26.25
3.3926
114
4



29.06
3.06999
175
6



37.08
2.42261
104
4

















TABLE 80







XRPD Peak positions of the Meglumine salt of Compound


A from the experiment L100110-68-85 (Form 17-B).














Height
Relative Intensity,



2-θ
d (A°)
(counts)
%
















7.50
11.77025
2296
100



11.04
8.01083
465
20



11.46
7.71675
604
26



12.22
7.23718
224
10



13.32
6.64298
310
14



15.60
5.67428
254
11



17.19
5.15324
598
26



17.95
4.9379
275
12



19.29
4.59784
449
20



20.32
4.36618
153
7



22.19
4.00331
691
30



22.84
3.89095
212
9



24.15
3.68285
246
11



26.36
3.37794
350
15



28.38
3.14239
564
25



29.85
2.99034
91
4










EQUIVALENTS

While the present invention has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and other variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present invention.

Claims
  • 1. A crystalline salt of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile (“Compound A”).
  • 2. The crystalline salt of claim 1, characterized as having a counter-ion, wherein the counter-ion is selected from L-lysine, L-arginine, 2-hydroxy-N,N,N-trimethylethan-1-aminium, diethylamine, ethanolamine, ethanol-2-diethylamine, Na+, Mg2+, K+, Ca2+, diethanolamine, triethanolamine, L-histidine, and meglumine.
  • 3. The crystalline salt of claim 1, wherein the counter-ion is L-lysine.
  • 4. The crystalline salt of claim 1, wherein the counter-ion is L-arginine.
  • 5. The crystalline salt of claim 1, wherein the counter-ion is 2-hydroxy-N,N,N-trimethylethan-1-aminium.
  • 6. The crystalline salt of claim 3, characterized by an X-ray powder diffraction pattern including peaks at about 8.70, 9.22, 11.3, 17.0, and 24.8 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 7. The crystalline salt of claim 3, having an X-ray diffraction pattern substantially similar to that set forth in FIG. 76.
  • 8. (canceled)
  • 9. The crystalline salt of claim 4, having an X-ray diffraction pattern substantially similar to that set forth in any one of FIGS. 67-70.
  • 10. (canceled)
  • 11. The crystalline salt of claim 5, having an X-ray diffraction pattern substantially similar to that set forth in FIG. 66.
  • 12. (canceled)
  • 13. The crystalline salt of claim 1, having a purity of Compound A of greater than 90% by weight.
  • 14. (canceled)
  • 15. (canceled)
  • 16. A pharmaceutical composition comprising the crystalline salt of claim 1.
  • 17. A morphic form (Form B) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 5.92, 11.8, and 17.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 18. (canceled)
  • 19. A morphic form (Form C) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 5.74, 11.5, 17.7, 19.3, 19.7, 21.4, 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 20. (canceled)
  • 21. A morphic form (Form D) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 5.52, 8.52, 11.0, 16.5, 18.3, 21.0, 21.2, and 24.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 22. (canceled)
  • 23. A morphic form (Form E) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 7.13, 10.8, 12.3, 14.1, 14.7, 15.5, 16.1, 17.5, 18.1, 19.9, 20.2, 21.0, 21.2, 22.7, 22.9, 24.4, 25.3, and 26.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 24. (canceled)
  • 25. A morphic form (Form F) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.1, 10.4, 11.4, 13.9, 16.2, 16.4, 17.1, 22.0, 23.8, and 29.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 26. (canceled)
  • 27. A morphic form (Form G) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 9.50, 12.9, 16.7, 17.3, 19.5, 20.2, 25.6, and 28.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 28. (canceled)
  • 29. A morphic form (Form H) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 9.22, 19.8, 23.6, 25.9, and 28.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 30. (canceled)
  • 31. A morphic form (Form I) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 5.77, 9.30, 10.2, 11.6, and 21.9 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 32. (canceled)
  • 33. A morphic form (Form K) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 8.42, 11.4, 14.5, 18.9, 21.1, and 21.6 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 34. (canceled)
  • 35. A morphic form (Form L) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.5, 11.5, 11.9, 15.2, 15.7, 16.0, 16.9, 17.1, 18.4, 18.7, 22.0, 22.8, 23.5, and 26.4 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 36. (canceled)
  • 37. A morphic form (Form S+T) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 7.42, 10.5, 11.3, 12.4, 14.3, 15.8, 16.8, 17.7, 18.1, 18.4, 20.1, 20.5, 21.1, 21.9, 23.2, 25.5, 26.9, and 28.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 38. (canceled)
  • 39. A morphic form (Form S) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.5, 12.3, 14.4, 15.8, 16.7, 17.7, 18.1, 18.4, 20.1, 20.6, 21.2, 21.9, 23.3, 24.4, 25.5, and 27.8 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 40. (canceled)
  • 41. A morphic form (Form U) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 5.79, 8.43, 11.4, 11.6, 14.5, 18.9, 21.1, and 21.6 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 42. (canceled)
  • 43. A morphic form (Form V) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 6.35, 10.6, 15.6, 16.5, 16.8, and 18.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 44. (canceled)
  • 45. A morphic form (Form W) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.1, 10.4, 10.7, 11.7, 13.9, 24.4, and 29.5 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 46. (canceled)
  • 47. A morphic form (Form X) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 9.66, 10.2, 10.5, 11.2, 18.7, and 24.7 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 48. (canceled)
  • 49. A morphic form (Form Y) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 6.51, 13.0, 13.3, 19.5, and 24.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 50. (canceled)
  • 51. A morphic form (Form Z) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.6, 11.2, 11.6, 12.0, 14.3, 15.6, 16.2, 17.6, 18.1, 18.7, 24.1, and 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 52. (canceled)
  • 53. A morphic form (Form α) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 7.26, 10.1, 10.4, 10.6, 11.9, 13.9, 16.5, 21.9, 22.4, and 24.1 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 54. (canceled)
  • 55. A morphic form (Form β) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 7.36, 10.5, 14.3, 15.7, 18.3, 20.4, 21.0, 21.8, and 23.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 56. (canceled)
  • 57. A morphic form (Form χ) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 8.53, 11.2, 18.4, 20.1, and 21.4 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 58. (canceled)
  • 59. A morphic form (Form δ) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.9, 12.1, 14.4, 18.1, 19.6, 24.5, and 27.0 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 60. (canceled)
  • 61. A morphic form (Form ε) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 5.73, 11.4, 16.6, 17.6, 23.2, and 24.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 62. (canceled)
  • 63. A morphic form (Form ϕ) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 6.95, 13.9, 20.9, 22.3, and 27.9 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 64. (canceled)
  • 65. A morphic form (Form η) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 6.87, 7.69, 20.5, 23.0, 23.9, and 28.2 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 66. (canceled)
  • 67. A morphic form (Form λ) of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile, characterized by an X-ray powder diffraction pattern including peaks at about 10.6, 12.0, 14.3, 16.2, 17.6, 18.0, and 24.3 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 68. (canceled)
  • 69. A co-crystal of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile and glutaric acid, characterized by an X-ray powder diffraction pattern including peaks at about 9.74, 10.8, 11.0, 12.2, 16.1, 17.0, 19.2, 21.9, and 23.1 degrees 2θ, wherein the x-ray powder diffraction pattern is obtained using a Cu Kα radiation source (1.54 Å).
  • 70. The co-crystal of claim 69, having an X-ray diffraction pattern substantially similar to that set forth in FIG. 28.
  • 71. An amorphous solid dispersion of 2-(3,5-dichloro-4-((5-isopropyl-6-oxo-1,6-dihydropyridazin-3-yl)oxy)phenyl)-3,5-dioxo-2,3,4,5-tetrahydro-1,2,4-triazine-6-carbonitrile (“Compound A”), wherein the amorphous solid dispersion comprises a polymer.
  • 72. The amorphous solid dispersion of claim 71, wherein the polymer is polyvinylpyrrolidone.
  • 73. The amorphous solid dispersion of claim 71, wherein the weight ratio of Compound A over the polymer is about 1:2.
  • 74. The amorphous solid dispersion of claim 71, wherein the weight ratio of Compound A over the polymer is about 1:4.
  • 75. A method for treating a resistance to thyroid hormone (RTH) syndrome in a subject having at least one TRβ mutation, the method comprising administering to the subject a therapeutically effective amount of the crystalline salt of claim 1.
  • 76. (canceled)
  • 77. (canceled)
  • 78. The method of claim 75, wherein the subject has obesity, hyperlipidemia, hypercholesterolemia, heterozygous familial hypercholesterolemia, diabetes, non-alcoholic steatohepatitis, fatty liver, fatty liver disease, bone disease, thyroid axis alteration, atherosclerosis, a cardiovascular disorder, tachycardia, hyperkinetic behavior, hypothyroidism, goiter, attention deficit hyperactivity disorder, dyslipidemia, learning disabilities, mental retardation, hearing loss, delayed bone age, neurologic or psychiatric disease or thyroid cancer.
  • 79. The method claim 75, wherein the TRβ mutation is selected from the group consisting of a substitution of threonine (T) for the wild type residue alanine (A) at amino acid position 234 of SEQ ID NO: 1 (A234T); a substitution of glutamine (Q) for the wild type residue arginine (R) at amino acid position 243 of SEQ ID NO: 1 (R243Q); a substitution of histidine (H) for the wild type residue arginine (R) at amino acid position 316 of SEQ ID NO: 1 (R316H); and a substitution of threonine (T) for the wild type residue alanine (A) at amino acid position 317 of SEQ ID NO: 1 (A317T).
  • 80. (canceled)
  • 81. A method for treating non-alcoholic steatohepatitis in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the crystalline salt of claim 1.
  • 82-84. (canceled)
  • 85. A method for treating familial hypercholesterolemia in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the crystalline salt of claim 1.
  • 86-89. (canceled)
  • 90. A method for treating fatty liver disease in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the crystalline salt of claim 1.
  • 91-93. (canceled)
  • 94. A method for treating dyslipidemia in a subject in need thereof, the method comprising administering to the subject a therapeutically effective amount of the crystalline salt of claim 1.
  • 95-98. (canceled)
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of priority to U.S. Provisional Application No. 62/692,914, filed on Jul. 2, 2018, the contents of which are hereby incorporated by reference in their entireties.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2019/040276 7/2/2019 WO 00
Provisional Applications (1)
Number Date Country
62692914 Jul 2018 US