NZ789638A - Pharmaceutical compositions - Google Patents

Pharmaceutical compositions

Info

Publication number
NZ789638A
NZ789638A NZ789638A NZ78963817A NZ789638A NZ 789638 A NZ789638 A NZ 789638A NZ 789638 A NZ789638 A NZ 789638A NZ 78963817 A NZ78963817 A NZ 78963817A NZ 789638 A NZ789638 A NZ 789638A
Authority
NZ
New Zealand
Prior art keywords
type
degrees
sulfate
crystal form
salt
Prior art date
Application number
NZ789638A
Inventor
Lixin Li
Original Assignee
Birdie Biopharmaceuticals Inc
Filing date
Publication date
Application filed by Birdie Biopharmaceuticals Inc filed Critical Birdie Biopharmaceuticals Inc
Publication of NZ789638A publication Critical patent/NZ789638A/en

Links

Abstract

The present disclosure relates to new crystal forms of chemical compounds, formulations including, methods of forming, and methods of using same.

Description

Pharmaceutical Compositions TECHNICAL FIELD The present disclosure relates to crystal forms of pharmaceutical compounds, formulations including, s of forming, and methods of using these crystal forms.
SUMMARY Described herein are new crystal forms of pharmaceutical nds. In one embodiment, the pharmaceutical nds are included in a ceutical composition useful for treating a disease or condition. In one embodiment, the disease or condition is cancer.
In some embodiments, pharmaceutical compositions are described including resiquimod in the form of a sulfate salt in crystal form A. The sulfate salt can be a monosulfate salt and/or an anhydrate. This l form can be prepared in an appropriate dosage from.
In one embodiment, the sulfate salt in crystal form A is characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 s 26, about 13.5 to about 14.5 degrees 26, about 18 to about 19 degrees 26, and/or about 15 to about 16 degrees 26.
The e salt in crystal form A can be stable at room temperature for at least about 2 days or at least about 1 week.
Other embodiments describe pharmaceutical compositions comprising a crystal form of a compound of Formula I: l N\: :X \ R3 N R2 Formula I can be a compound having a formula 4-amino-d-butyl(2- methylpropyl)-1H-imidazo-[4,5-c]-quinolinemethanol hemihydrate, 4-amino-d,d-dimethyl- xymethyl-1H-imidazo-[4,5-c]-quinolineethanol, 2-ethoxymethyl(2-methylpropyl)- 1H-imidazo-[4,5-c]-quinolinamine, or 4-aminophenylmethyl-1H-imidazo-[4,5-c]- quinoline—2—methanol. In one embodiment, Formula | is resiquimod.
In some embodiments, the crystal form of a compound of Formula I can be in form A and/or be a e salt. In one embodiment, a sulfate salt is a monosulfate salt and/or an anhyd rate.
Other embodiments provide s of treating diseases or conditions. In one embodiment, methods of treating cancer are described. The methods can comprise: administering a pharmaceutical composition ing a crystal form of a compound having a Formula I: N/ N X l \> ; \ 3 \ R N R2 BRIEF DESCRIPTION OF THE DRAWINGS illustrates XRPD patterns of mono-HCI salt crystal form Type A s. illustrates a PLM image of mono-HCI salt crystal form Type A (807919 illustrates TGA/DSC curves of mono-HCI salt l form Type A (807919- 16-A). illustrates an XRPD pattern of di-HCI salt crystal form Type A (807919 illustrates a PLM image of di-HCI salt crystal form Type A (807919-14—A). illustrates TGA/DSC curves of di-HCI salt l form Type A (807919—14- illustrates XRPD ns of sulfate crystal form Type A batches. illustrates a PLM image of sulfate crystal form Type A (807919A). illustrates TGA/DSC curves of sulfate crystal form Type A (807919A). illustrates XRPD patterns of phosphate crystal form Type A batches. illustrates a PLM image of phosphate crystal form Type A (807919—11-0). illustrates TGA/DSC curves of phosphate crystal form Type A (807919- 11-0). illustrates XRPD patterns of maleate crystal form Type A batches. illustrates a PLM image of maleate crystal form Type A (807919—11-B). illustrates TGA/DSC curves of e crystal form Type A (807919B). illustrates XRPD patterns of malate crystal form Type A s. illustrates a PLM image of malate crystal form Type A 9E). illustrates TGA/DSC curves of malate crystal form Type A 9—11-E). rates XRPD patterns of adipate crystal form Type A batches. illustrates a PLM image of adipate crystal form Type A (807919A). illustrates TGA/DSC curves of adipate crystal form Type A (807919A). illustrates a DVS plot of sulfate crystal form Type A (807919—11-A). illustrates a XRPD y of sulfate crystal form Type A (807919—11-A) pre and post DVS test. illustrates a DVS plot of phosphate crystal form Type A (807919—11-0). illustrates a XRPD overlay of phosphate crystal form Type A (807919—1 1- C) pre and post DVS test. illustrates a DVS plot of maleate crystal form Type A (807919—11-B). illustrates a XRPD overlay of maleate crystal form Type A (807919B) pre and post DVS test. illustrates a DVS plot of malate l form Type A (807919E). illustrates a XRPD overlay of malate crystal form Type A (807919E) pre and post DVS test. rates a DVS plot of adipate l form Type A (807919A). illustrates a XRPD overlay of adipate crystal form Type A (807919A) pre and post DVS test. illustrates a DVS plot of mono-HCI salt crystal form Type A (807919A). illustrates a XRPD overlay of mono-HCI salt crystal form Type A (807919- 16-A) pre and post DVS test. illustrates a DVS plot of di-HCI salt crystal form Type A (807919A). illustrates a XRPD overlay of di-HCI salt crystal form Type A (807919 A) pre and post DVS test. illustrates kinetic solubility of seven crystal forms described herein and freebase crystal forms (short : clear solutions were observed during the evaluation). illustrates a XRPD y of se crystal form Type A (807919A) after suspended for 24 hrs. illustrates a XRPD y of adipate crystal form Type A (807919A) after suspended for 24 hrs. illustrates a XRPD overlay of maleate crystal form Type A (807919B) after suspended for 24 hrs. illustrates a XRPD overlay of freebase crystal form Type A (807919A) pre and post ity test. illustrates a XRPD overlay of mono-HCI salt crystal form Type A (807919- 16-A) pre and post ity test. illustrates a XRPD overlay of di-HCI salt crystal form Type A (807919 A) pre and post stability test. illustrates a XRPD overlay of e crystal form Type A (807919A) pre and post stability test. illustrates a XRPD overlay of phosphate crystal form Type A (807919 C) pre and post stability test. illustrates a XRPD overlay of maleate crystal form Type A (807919B) pre and post stability test. illustrates a XRPD overlay of malate crystal form Type A (807919E) pre and post stability test. rates a XRPD overlay of adipate crystal form Type A (807919-12—A) pre and post ity test. rates a XRPD pattern of sulfate crystal form Type B (807919A13). illustrates TGA/DSC curves of sulfate crystal form Type B (807919 A13). illustrates a XRPD pattern of hemi-sulfate crystal form Type A (807919- 34-A). illustrates TGA/DSC curves of hemi-sulfate crystal form Type A (807919- 34-A). illustrates a XRPD overlay of slurry experiments at RT. illustrates a XRPD overlay of sulfate crystal form Type A (807919A) before and after storage. illustrates a XRPD n of freebase crystal form Type A 9A). illustrates a PLM image of freebase crystal form Type A (807919A). illustrates TGA/DSC curves of freebase crystal form Type A (807919 rates a DVS plot of freebase crystal form Type A (807919A). illustrates a XRPD overlay of freebase crystal form Type A (807919A) before and after DVS test. illustrates XRPD patterns of sulfate crystal form Type A batches. illustrates TGA/DSC curves of sulfate crystal form Type A (807919A). illustrates XRPD patterns of HCI salt crystal forms. illustrates TGA/DSC curves of HCI salt l form Type B (807919 illustrates a XRPD pattern of sulfate crystal form Type A (807919-07—A3). rates TGA/DSC curves of sulfate crystal form Type A (807919-07—A3). illustrates a XRPD pattern of phosphate crystal form Type A (807919 illustrates TGA/DSC curves of phosphate crystal form Type A (807919- 07-E5). illustrates a XRPD pattern of glycolate crystal form Type A 9 illustrates TGA/DSC curves of glycolate crystal form Type A (807919 illustrates a XRPD pattern of maleate crystal form Type A (807919D4). illustrates TGA/DSC curves of maleate crystal form Type A (807919 illustrates a XRPD pattern of malate l form Type A (807919B10). illustrates TGA/DSC curves of malate crystal form Type A (807919 B10). illustrates a XRPD pattern of adipate crystal form Type A (807919 B14). illustrates TGA/DSC curves of adipate crystal form Type A (807919 B14). illustrates a XRPD pattern of hippurate crystal form Type A (807919 B11). illustrates TGA/DSC curves of hippurate crystal form Type A 9 B11). rates XRPD patterns of tartrate crystal form Type A (807919-07—A6). illustrates XRPD patterns of tartrate l form Type B 9-07—E6). illustrates XRPD patterns of tartrate crystal Type C (807919-07—B6). illustrates TGA/DSC curves of te crystal form Type A (807919 illustrates TGA/DSC curves of tartrate crystal form Type B (807919 rates TGA/DSC curves of tartrate crystal form Type C (807919 illustrates XRPD patterns of fumarate l form Type A (807919-07—A7).
WO 32725 2017/089718 illustrates XRPD patterns of fumarate crystal form Type B (807919-07—E7). illustrates XRPD patterns of fumarate crystal form Type C (807919C7). illustrates TGA/DSC curves of fumarate crystal form Type A (807919 illustrates C curves of fumarate crystal form Type B (807919 illustrates TGA/DSC curves of fumarate crystal form Type C 9 illustrates XRPD patterns of citrate crystal form Type A (807919A8). illustrates XRPD patterns of citrate crystal form Type B (807919B8). illustrates TGA/DSC curves of citrate crystal form Type A (807919A8). illustrates TGA/DSC curves of citrate crystal form Type B 9B8). illustrates XRPD patterns of e crystal form Type A (807919012). illustrates XRPD patterns of lactate crystal Type B (807919A12). illustrates TGA/DSC curves of lactate crystal form Type A (807919 illustrates TGA/DSC curves of e crystal form Type B 9 illustrates XRPD patterns of succinate crystal Type A (807919013). illustrates XRPD patterns of succinate crystal Type B (807919-07—E13). illustrates TGA/DSC curves of ate crystal form Type A (807919 0 illustrates TGA/DSC curves of succinate crystal form Type B (807919- 07-E13) 1 illustrates XRPD patterns of te crystal Type A (807919-07—B15). 2 illustrates XRPD patterns of tosylate crystal Type B (807919-07—D15). 3 rates TGA/DSC curves of tosylate crystal form Type A (807919 4 illustrates TGA/DSC curves of tosylate crystal form Type B (807919 D15). 5 rates a XRPD pattern of mesylate crystal form Type A (807919 6 illustrates TGA/DSC curves of mesylate crystal form Type A (807919 7 illustrates XRPD patterns of oxalate crystal form Type A 9 8 illustrates XRPD patterns of e crystal form Type B (807919 9 illustrates TGA/DSC curves of oxalate crystal form Type A (807919 0 illustrates TGA/DSC curves of oxalate crystal form Type B 9 1 illustrates XRPD patterns of gentisate crystal form Type A (807919 2 illustrates XRPD patterns of gentisate crystal form Type B (807919 ] 3 illustrates TGA/DSC curves of gentisate crystal form Type A (807919 4 illustrates TGA/DSC curves of gentisate crystal form Type B (807919 5 illustrates XRPD patterns of benzoate crystal form Type A (807919 ] 6 illustrates XRPD patterns of benzoate crystal Type B (807919E19). 7 illustrates TGA/DSC curves of benzoate crystal form Type A (807919- 8 illustrates TGA/DSC curves of benzoate crystal form Type B (807919- 07-E19). 9 illustrates XRPD patterns of nitrate crystal form Type A (807919-07—D20). ] 0 rates XRPD patterns of e crystal form Type B (807919B20). 1 illustrates TGA/DSC curves of nitrate crystal form Type A (807919 020). 2 illustrates TGA/DSC curves of nitrate crystal form Type B (807919 3 illustrates an inter-conversion of freebase crystal forms. 4 illustrates a XRPD pattern of crystal form Type A (807920A). 5 illustrates C curves of crystal form Type A (807920A). 6 illustrates a XRPD pattern of crystal form Type C 0A11). 7 illustrates a XRPD pattern of crystal form Type F (807920-09—A4). 8 rates XRPD patterns of isomorphic Type B. 9 illustrates TGA/DSC curves of a first batch of crystal form Type B (807920-07—A13). 0 rates TGA/DSC curves of a second batch of crystal form Type B 0-07—A13). 1 illustrates TGA/DSC curves of a third batch of crystal form Type B (807920-07—A13). 2 illustrates a XRPD n of crystal form Type D (807920A9). ] 3 illustrates TGA/DSC curves of crystal form Type D (807920A9). 4 illustrates a XRPD pattern of crystal form Type E (807920A3). 5 illustrates TGA/DSC curves of crystal form Type E (807920-16—A3). 6 illustrates a XRPD pattern of crystal form Type G (807920-19—F). 7 illustrates TGA/DSC curves of crystal form Type G (807920F). 8 rates a XRPD pattern of sample H (807920A1). 9 illustrates TGA/DSC curves of sample H 0A1). 0 illustrates a DVS plot of crystal form Type A (807919A). 1 illustrates a XRPD overlay of crystal form Type A (807919A) before and after DVS test. 2 illustrates a XRPD overlay of l form Type A (807919A) before and after stability test.
DETAILED PTION Described herein are new crystal forms of chemical compounds, formulations including these crystal forms, methods of forming these crystal forms, and methods of using these l forms. In some embodiments, these new crystal forms can be referred to as polymorphs or isomorphs. Pharmaceutical polymorphism can have a direct effect on delivery of a given pharmaceutical active agent, ingredient, or drug. Polymorphic purity of samples can be d using techniques such as powder X—ray diffraction, lR/Raman spectroscopy, and utilizing the ences in their optical ties.
In general, active pharmaceutical ingredients (APls) in pharmaceutical compositions can be prepared in a variety of different forms including prodrugs, amorphous forms, solvates, hydrates, co-crystals, salts, and the like. The discovery of novel API forms may provide an opportunity to improve the performance characteristics of a pharmaceutical composition. Additionally, discovery of drug forms expands the array of resources available for designing pharmaceutical dosage forms with targeted release profiles or other desired characteristics.
A specific teristic that can be targeted includes the crystal form of an API.
The alteration of the crystal form of a given API can result in modification of target molecule physical ties. For example, s polymorphs of a given API can exhibit different aqueous solubility, while the thermodynamically stable polymorph would exhibit a lower lity than the table polymorph. In addition, pharmaceutical polymorphs can also differ in properties such as rate of dissolution, shelf life, bioavailability, morphology, vapor pressure, density, color, and compressibility. Accordingly, it may be desirable to enhance the properties of an API by forming lar complexes such as a crystal, a co-crystal, a salt, a solvate or a hydrate with respect to s solubility, rate of dissolution, bioavailability, Cmax, Tmax, physicochemical stability, down-stream processibility (e.g., flowability, compressibility, degree of brittleness, particle size manipulation), decrease in polymorphic form diversity, toxicity, taste, production costs, manufacturing methods, or a combination thereof.
New crystal forms of compounds and pharmaceutical compositions including the new l forms of these compounds are disclosed. The l forms can be of compounds having a structure of Formula I: WO 32725 l N:\ :X \ R3 N R2 or a pharmaceutically acceptable salt thereof, wherein R1 is hydrogen; C1-C1o straight chain or branched chain substituted or unsubstituted alkyl, wherein the substituent is Cg'Cs cycloalkyl or 03-06 cycloalkyl substituted by ht chain or branched chain C1-C4 alkyl; straight chain or branched chain 02-010 alkenyl; or substituted straight chain or branched chain 02-010 alkenyl, wherein the substituent is C3-C5 cycloalkyl or C3-C5 cycloalkyl tuted by straight chain or branched chain C1-C4 alkyl; C1- C6 hydroxyalkyl; alkoxyalkyl n the alkoxy moiety contains one to about four carbon atoms and the alkyl moiety contains one to about six carbon atoms; acyloxyalkyl n the acyloxy moiety is alkanoyloxy of two to about four carbon atoms or benzoyloxy, and the alkyl moiety contains one to about six carbon atoms; ; (phenyl)ethyl; or phenyl; the benzyl, (phenyl)ethyl, or phenyl substituent being optionally substituted on the benzene ring by one or two moieties independently ed from C1-C4 alkyl, C1-C4 alkoxy, or halogen, with the proviso that if the benzene ring is substituted by two moieties, then the moieties together contain no more than six carbon atoms; R2 and R3 are independently selected from hydrogen, C1-C4 alkyl, phenyl, or substituted phenyl, wherein the substituent is C1-C4 alkyl, C1-C4 alkoxy, or halogen; X is C1-C4 alkoxy, alkoxyalkyl wherein the alkoxy moiety contains one to about four carbon atoms and the alkyl moiety contains one to about four carbon atoms, C1-C4 hydroxyalkyl, C1- C4 haloalkyl, alkylamido wherein the alkyl group contains one to about four carbon atoms, amino, substituted amino wherein the substituent is C1-C4 alkyl or C1-C4 hydroxyalkyl, azide, chloro, hydroxy, 1-morpholino, 1-pyrrolidino, or C1-C4 alkylthio; and R is en, straight chain or branched chain C1-C4 alkoxy, halogen, or straight chain or ed chain C1-C4 alkyl.
In some embodiments, the crystal form of compounds having a ure of Formula | is a hloric acid salt, a sulfate salt, a phosphate salt, a maleate salt, a malate salt, an adipate salt, a ate salt, a hippurate salt, a tartrate salt, a fumarate salt, a citrate salt, a lactate salt, a succinate salt, a tosylate salt, mesylate salt, an oxalate salt, a gentisate salt, a benzoate salt, or a nitrate salt in crystalline form A, B, C, D, E, F or G.
In some embodiments, R1 may contain two to about ten carbon atoms. In other embodiments, R1 may contain two to about eight carbon atoms. In still other embodiments, R1 is 2-methylpropyl or benzyl.
In some embodiments, X can be azido, hydroxy, ethoxy, methoxy, 1-morpholino, or methylthio. In some embodiments, X can be azido, y, , methoxy, 1- morpholino, or methylthio when R1 is ylpropyl, 2-hydroxymethylpropyl, or benzyl.
Other substituents in compounds of Formula I that contain an alkyl radical (e.g., R when R is alkoxy or alkyl, or X when X is alkylamido) can contain two carbon atoms or, in some embodiments, one carbon atom in each alkyl radical.
In some embodiments, R is hydrogen.
Compounds of Formula I can include 4-amino-o-butyl(2-methylpropyl)-1H- imidazo-[4,5-c]-quinolinemethanol drate, 4-amino-d,o-dimethylethoxymethyl- 1H-imidazo-[4,5-c]-quinolineethanol, 2-ethoxymethyl(2-methylpropyl)-1H-imidazo-[4,5— c]-quinolin-4—amine, and ophenylmethyl-1H-imidazo-[4,5-c]-quinolinemethanol.
In one embodiment, a compound of Formula I can be resiquimod (1-[4—amino (ethoxymethyl)imidazo-[4,5-c]-quinolinyl]methylpropanol). Resiquimod can have a structure Halogen or halo groups in any of the compounds described herein can be F, Cl, Br, I, or At. In some embodiments, halogen or halo groups in any of the compounds described herein can be F, Cl, Br, or |.
These new crystal forms of a | compounds can be a HCI salt, a sulfate salt, a phosphate salt, a maleate salt, a malate salt, an adipate salt, a glycolate salt, a hippurate salt, a tartrate salt, a fumarate salt, a e salt, a lactate salt, a succinate salt, a tosylate salt, mesylate salt, an oxalate salt, a gentisate salt, a benzoate salt, or a nitrate salt in crystalline form A, B, C, D, E, F or G.
One embodiment includes resiquimod in the form of a monosulfate salt in crystal form A. The crystal form A can be characterized by an x-ray powder diffraction um that comprises peaks at about 7 to about 8 degrees 26, about 13.5 to about 14.5 degrees 26, about 19 to about 20 degrees 26, and/or about 19.5 to about 20.5 degrees 26.
Another ment es resiquimod in the form of a sulfate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction um that ses peaks at about 7 to about 8 degrees 26, about 9 to about 10 degrees 26, about 11 to about 12 degrees 26, about 14 to about 14.5 degrees 26, about 15 to about 16 degrees 26, about 17 to about 20 degrees 26, and/or about 24 to about 26 degrees 26.
Resiquimod can also be formed as a sulfate salt in crystal form A characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 11.5 to about 12 degrees 26, about 14 to about 14.5 degrees 26, about 16 to about 16 degrees 26, about 17 to about 18.5 degrees 26, about 19.5 to about 20.5 degrees 26, and/or about 24 to about 25 degrees 26.
Resiquimod can be formed as a sulfate salt in crystal form B. Such a crystal form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 9 to about 10 degrees 26, and/or about 19 to about 20.5 degrees 26.
Resiquimod can also be formed as a hemi-sulfate salt in crystal form A. Such a l form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 6.5 degrees 26, about 7 to about 8 degrees 26, about 8 to about 9 degrees 26, about 11 to about 12 s 26, about 12.5 to about 13 degrees 26, about 15 to about 15.5 degrees 26, about 16 to about 17 degrees 26, about 19 to about 19.5 degrees 26, about 21 to about 21.5 degrees 26, and/or about 23 to about 24 degrees 26.
Other compounds of imod can be formed in an acetate/acetic acid co- crystal of the resiquimod freebase form. Such a crystal form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 9 to about 10.5 degrees 26, about 11 to about 12 degrees 26, about 18 to about 19 degrees 26, about 19 to about 20 s 26, about 20.5 to about 21 degrees 26, about 22 to about 23 degrees 26, and/or about 25 to about 26 degrees 26.
Sulfate salts can also be provided in crystal form C, D, E, F, G, and H.
Conversion of sulfate salt form C can be interconverted to form A by storage at ambient WO 32725 ature, for example, overnight. Conversion of sulfate salt form D can be onverted to form A by heating, for example, to 100 °C. Conversion of sulfate salt form E can be interconverted to form A by heating, for example, to 120 °C. Conversion of e salt form F can be interconverted to form A by storage at ambient temperature, for example, for two days. Conversion of sulfate salt form G can be interconverted to form A by heating, for example, to 80 °C. Because interconversion of each metastable forms and solvates converted to form A, in some embodiments, form A is the thermodynamically stable form at room temperature.
Resiquimod can be formed as an anhydrate (Type A) sulfate salt. Such a crystal form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 8.5 to about 9 degrees 26, about 12 to about 13 degrees 26, about 16 to about 17 degrees 26, about 17.5 to about 18 degrees 26, about 19 to about 20.5 s 26, about 21 to about 22 degrees 26, about 23 to about 24 degrees 26, and/or about 29 to about 30 degrees 26.
Resiquimod can be formed as a solvate (Type B) sulfate salt. Such a crystal form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 6.5 degrees 26, about 12 to about 12.5 degrees 26, about 16 to about 16.5 degrees 26, about 21 to about 22.5 s 26, and/or about 24.5 to about 25 degrees 26.
Resiquimod can be formed as a sulfate salt Type C. Such a crystal form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 9 to about degrees 26, about 12 to about 12.5 degrees 26, about 14 to about 15 degrees 26, about 18 to about 19 degrees 26, about 19 to about 21.5 degrees 26, and/or about 28 to about 29 degrees 26.
Resiquimod can be formed as a DMAc solvate (Type D) sulfate salt. Such a crystal form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 8 to about 9 s 26, about 11 to about 11.5 degrees 26, about 16.5 to about 17 s 26, about 17.5 to about 18 degrees 26, about 21 to about 21.5 degrees 26, about 22.5 to about 23 degrees 26, and/or about 23.5 to about 24.5 degrees 26.
Resiquimod can be formed as a NMP solvate (Type E) sulfate salt. Such a crystal form can be characterized by x-ray powder ction spectrum that comprises peaks at about 8 to about 9 degrees 26, about 9 to about 9.5 degrees 26, about 11 to about 11.5 s 26, about 12 to about 13 degrees 26, about 16.5 to about 18 degrees 26, about 21 to about 21.5 s 26, about 22.5 to about 23 degrees 26, and/or about 23.5 to about 24 degrees 26.
Resiquimod can be formed as a sulfate salt Type F. Such a crystal form can be characterized by x-ray powder diffraction um that comprises peaks at about 8 to about 8.5 degrees 26, about 10 to about 11 degrees 26, about 12 to about 13 degrees 26, about 16 to about 17 degrees 26, about 17 to about 18 s 26, about 20.5 to about 21.5 degrees 26, about 24.5 to about 25 degrees 26, and/or about 28.5 to about 29 degrees 26.
Resiquimod can be formed as a anisole solvate (Type G) sulfate salt. Such a crystal form can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 8.5 to about 9 degrees 26, about 9.5 to about 10 degrees 26, about 13 to about 14 degrees 26, about 19 to about 19.5 degrees 26, and/or about 27.5 to about 28.5 s 26.
Other Formula | compounds described herein can be formed in similar salt configurations.
Crystal forms of Formula | compounds can be in Type A, Type B, Type C, Type D, Type E, Type F, Type G, and/or Type H. In some embodiments, the forms can be described as Type drate, Type B:solvate, Type C:mestable, Type D:dimethylacetamide (DMAc) solvate, Type E:N-methylpyrolidone (NMP) solvate, Type F:mestable, Type G:anisole solvate, and Type H:acetate/acetic acid co-crystal.
Still other compounds of Formula I bed herein can be formed as a sulfate salt in crystal form B. Sulfate salt in crystal form B can be a dimethyl sulfoxide (DMSO) e. Still other compounds of Formula I described herein can be formed as a hemi- sulfate salt in crystal form A.
] In some embodiments, form A can be stable at room temperature for at least about 1 day, at least about 2 days, at least about 3 days, at least about 1 week, at least about 2 weeks, at least about 1 month, at least about 6 months, or at least about 1 year.
Still other compounds of a I bed herein can be formed as a HCI salts, sulfate salts, phosphate salts, maleate salts, malate salts, adipate salts or combinations thereof. In some ments, the salts can be formed in form or type A.
In one embodiment, compounds of Formula I described herein can be formed as a mono-HCI salt in l form A. In one embodiment, compounds of Formula I described herein can be formed as a di-HCI salt in crystal form A. Either form of HCI salt can be formed as an anhydrate.
One embodiment includes resiquimod in the form of a HCI salt in crystal form A.
The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 9 to about 10 degrees 26, about 12 to about 13 degrees 26, about 14 to about 16 degrees 26, about 18 to about 23 degrees 26, about 23 to about 25 degrees 26, about 26 to about 27.5 degrees 26, and/or about 26 to about 27.5 degrees 26.
Another embodiment includes resiquimod in the form of a HCI salt in crystal form B. The crystal form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 8 degrees 26, about 19 to about 21 degrees 26, about 23 to about 24.5 degrees 26, about 26 to about 27 s 26, and/or about 28 to about 29 degrees 26.
Another embodiment includes resiquimod in the form of a CI salt in l form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 9 to about 10 degrees 26, about 13 to about 14 degrees 26, about 17 to about 18 degrees 26, about 20 to about 21 degrees 26, about 27 to about 28 degrees 26, and/or about 34 to about 35 degrees 26.
Another embodiment includes resiquimod in the form of a di-HCI salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 8 to about 9 degrees 26, about 14 to about 15 degrees 26, about 15 to about 16 degrees 26, about 19 to about 20 degrees 26, about 25 to about 26 degrees 26, and/or about 26.5 to about 27.5 s 26.
In one ment, compounds of Formula I described herein can be formed as an anhydrate phosphate salt in crystal form A.
Another embodiment es resiquimod in the form of a phosphate salt in l form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 10 to about 14.5 degrees 26, about 15 to about 16 degrees 26, about 20 to about 21 degrees 26, and/or about 25 to about 26 degrees 26. Other embodiments include resiquimod in the form of a phosphate salt in crystal form A characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8.5 degrees 26, about 10 to about 15.5 degrees 26, about 16 to about 18.5 degrees 26, about 19 to about 21 s 26, about 22 to about 23 s 26, about 23 to about 27 degrees 26, and/or about 28 to about 29 degrees 26.
In one embodiment, compounds of Formula I described herein can be formed as an anhydrate maleate salt in crystal form A. In another embodiment, compounds of Formula I described herein can be formed as an anhydrate mono-maleate salt in crystal form A.
Another ment includes resiquimod in the form of a maleate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 9 to about 10 degrees 26, about to about 11 degrees 26, about 15 to about 17 degrees 26, about 20 to about 21 degrees 26, about 21 to about 22 degrees 26, about 27 to about 28 degrees 26, and/or about 30 to about 31 degrees 26. Other ments include resiquimod in the form of a e salt in crystal form A characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 9 to about 10 degrees 26, about 10 to about 11 degrees 26, about 11 to about 12 degrees 26, about 15 to about 16.5 s 26, about 17 to about 19 degrees 26, about 20 to about 21 degrees 26, about 21 to about 22 degrees 26, about 24 to about 25 degrees 26, about 27 to about 28 degrees 26, and/or about 30 to about 31 degrees 26.
In one embodiment, nds of Formula I described herein can be formed as a anhydrate malate salt in crystal form A.
] Another embodiment includes resiquimod in the form of a malate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 8 to about 9 degrees 26, about 13 to about 14 degrees 26, about 17 to about 18 s 26, and/or about 24 to about 25.5 degrees 26. Other embodiments include resiquimod in the form of a malate salt in crystal form A characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 8 to about 9 degrees 26, about 17 to about 18 degrees 26, about 21.5 to about 23.5 degrees 26, and/or about 25 to about 26 degrees 26.
In one embodiment, compounds of Formula I described herein can be formed as an anhydrate e salt in crystal form A.
Another embodiment includes resiquimod in the form of an adipate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction um that comprises peaks at about 5.5 to about 6 degrees 26, about 11 to about 12 degrees 26, about 12 to about 13 degrees 26, about 13 to about 14 degrees 26, about 14 to about 15 degrees 26, about 18 to about 19 degrees 26, about 19 to about 20 s 26, about 21 to about 22 degrees 26, about 22 to about 23 degrees 26, and/or about 25 to about 28 degrees 26. Other embodiments include resiquimod in the form of an adipate salt in crystal form A characterized by x-ray powder diffraction spectrum that comprises peaks at about 5 to about 6.5 degrees 26, about 9 to about 11 degrees 26, about 12 to about 13.5 degrees 26, about 14 to about 15.5 degrees 26, about 17 to about 18 s 26, about 18 to about 19 degrees 26, about 19.5 to about 22 s 26, about 22 to about 25 degrees 26, and/or about 26 to about 27.5 degrees 26.
Formula | compounds can also be formed as ate salts. In one embodiment, compounds of Formula I described herein can be formed as a glycolate salt in crystal form A.
WO 32725 One embodiment es resiquimod in the form of a glycolate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 9 to about 10 degrees 26, about 11.5 to about 12.5 degrees 26, about 18 to about 19 degrees 26, about 19.5 to about 23 degrees 26, about 25 to about 26.5 degrees 26, and/or about 32 to about 33 degrees 26.
Formula | compounds can also be formed as hippurate salts. In one embodiment, compounds of Formula I described herein can be formed as a hippurate salt in crystal form A.
One ment includes resiquimod in the form of a hippurate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 5.5 to about 6.5 degrees 26, about 9 to about 10 degrees 26, about 11.5 to about 12.5 degrees 26, about 18.5 to about 19.5 degrees 26, about 21 to about 22 degrees 26, and/or about 25 to about 26 s 26. a | compounds can also be formed as tartrate salts. In another embodiment, compounds of Formula I described herein can be formed as a tartrate salt in l forms A, B, and/or C.
One embodiment includes resiquimod in the form of a tartrate salt in l form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 9 to about 10 degrees 26, about 18 to about 19 degrees 26, about 20 to about 22 degrees 26, and/or about 25 to about 26 degrees 26.
One embodiment includes resiquimod in the form of a tartrate salt in crystal form B. The crystal form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 8.5 to about 9 degrees 26, about 11 to about 12 degrees 26, about 13 to about 14 s 26, about 14 to about 15 degrees 26, about 16 to about 17 degrees 26, and/or about 23 to about 24.5 s 26.
One embodiment includes resiquimod in the form of a tartrate salt in l form C. The crystal form C can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 s 26, about 10 to about 11.5 degrees 26, and/or about 20 to about 21 degrees 26.
Formula | compounds can also be formed as fumarate salts. In another embodiment, compounds of Formula I described herein can be formed as a fumarate salt in crystal forms A, B, and/or C.
One embodiment includes resiquimod in the form of a te salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 7 to about 8 s 26, about 9 to about 10.5 degrees 26, about 12 to about 14 degrees 26, about 18 to about 19 degrees 26, about 19 to about 20 degrees 26, about 23 to about 24.5 degrees 26, and/or about 25 to about 26 degrees 26.
] One embodiment es imod in the form of a fumarate salt in crystal form B. The l form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 9.5 to about 10.5 degrees 26, about 12 to about 13 degrees 26, about 15 to about 16 s 26, about 17 to about 18 degrees 26, about 19 to about 21 degrees 26, and/or about 25 to about 26 degrees 26.
One embodiment includes resiquimod in the form of a fumarate salt in crystal form C. The crystal form C can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7 degrees 26, about 9 to about 10 degrees 26, about 11 to about 12 degrees 26, about 15 to about 16 degrees 26, about 21 to about 22 degrees 26, about 26 to about 27 degrees 26, and/or about 27.5 to about 28.5 degrees 26.
Formula | compounds can also be formed as citrate salts. In another embodiment, compounds of Formula I described herein can be formed as a citrate salt in crystal forms A and/or B.
One embodiment includes resiquimod in the form of a e salt in crystal form A.
The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 5 to about 6.5 degrees 26, about 11 to about 12 degrees 26, about 14.5 to about 15.5 degrees 26, about 17 to about 18.5 degrees 26, about 19 to about 20.5 degrees 26, about 21 to about 22 degrees 26, about 26 to about 27 degrees 26, and/or about 27.5 to about 28.5 degrees 26.
One embodiment includes resiquimod in the form of a citrate salt in crystal form B.
The crystal form B can be characterized by x-ray powder diffraction spectrum that ses peaks at about 5.5 to about 6.5 degrees 26, about 8 to about 9 degrees 26, about 9.5 to about 10.5 degrees 26, about 11 to about 12.5 degrees 26, about 18 to about 19.5 degrees 26, and/or about 21 to about 24.5 degrees 26. a | compounds can also be formed as lactate salts. In another embodiment, compounds of Formula I described herein can be formed as a lactate salt in crystal forms A and/or B.
One embodiment includes resiquimod in the form of a lactate salt in l form A.
The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 5 to about 8 degrees 26, about 8 to about 9 degrees 26, about 10 to about 11 degrees 26, about 12.5 to about 13.5 degrees 26, about 18.5 to about 19.5 degrees 26, and/or about 22 to about 23 degrees 26.
One embodiment includes imod in the form of a lactate salt in crystal form B.
The crystal form B can be terized by x-ray powder ction spectrum that comprises peaks at about 5 to about 6 degrees 26, about 6.5 to about 8 degrees 26, about 9 to about .5 degrees 26, and/or about 13.5 to about 14.5 s 26. a | compounds can also be formed as ate salts. In another ment, compounds of Formula I described herein can be formed as a succinate salt in crystal forms A and/or B.
One embodiment includes resiquimod in the form of a succinate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 5 to about 8 degrees 26, about 9 to about 11.5 s 26, about 18 to about 19 degrees 26, about 23 to about 24 s 26, and/or about 24.5 to about .5 degrees 26.
One embodiment es resiquimod in the form of a succinate salt in crystal form B. The crystal form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 8 to about 9 degrees 26, about 10 to about 11 degrees 26, about 12 to about 13 degrees 26, about 14 to about 15 degrees 26, about 16 to about 17 degrees 26, about 17 to about 18 s 26, and/or about 23.5 to about 24.5 degrees 26.
Formula | compounds can also be formed as tosylate salts. In another embodiment, compounds of Formula I described herein can be formed as a tosylate salt in crystal forms A and/or B.
One embodiment includes resiquimod in the form of a tosylate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 4 to about 5 degrees 26, about 9 to about 10 degrees 26, about 16 to about 17 degrees 26, about 19 to about 21 degrees 26, and/or about 24 to about 28 degrees 26.
One embodiment includes resiquimod in the form of a tosylate salt in crystal form B. The crystal form B can be terized by x-ray powder diffraction spectrum that comprises peaks at about 5 to about 6 degrees 26, about 7 to about 9 degrees 26, about 9.5 to about 11.5 degrees 26, about 12 to about 14 degrees 26, about 15 to about 19 degrees 26, about 19 to about 20.5 degrees 26, and/or about 23 to about 24 degrees 26.
Formula | compounds can also be formed as mesylate salts. In one embodiment, compounds of Formula I described herein can be formed as a mesylate salt in crystal form A.
One embodiment includes resiquimod in the form of a mesylate salt in l form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that ses peaks at about 8 to about 9 degrees 26, about 12 to about 13 degrees 26, about 14 to about 15 degrees 26, about 16 to about 17 degrees 26, about 18 to about 19.5 degrees 26, about 21 to about 22 degrees 26, and/or about 25.5 to about 26.5 degrees 26.
Formula | compounds can also be formed as oxalate salts. In another embodiment, compounds of Formula I described herein can be formed as an oxalate salt in crystal forms A and/or B.
One embodiment includes resiquimod in the form of an oxalate salt in crystal form A. The crystal form A can be terized by x-ray powder diffraction spectrum that comprises peaks at about 9 to about 10 degrees 26, about 14 to about 15 s 26, about 17 to about 18 degrees 26, about 18.5 to about 20 degrees 26, about 21 to about 22 degrees 26, about 23 to about 25.5 degrees 26, and/or about 30 to about 31 degrees 26.
One embodiment includes resiquimod in the form of an oxalate salt in crystal form B. The crystal form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 5 to about 6 degrees 26, about 9.5 to about 10 degrees 26, about .5 to about 11.5 degrees 26, about 13 to about 13.5 s 26, about 14.5 to about 15.5 degrees 26, about 16.5 to about 18 degrees 26, about 22 to about 24.5 degrees 26, and/or about 27 to about 28 degrees 26.
] Formula | compounds can also be formed as genisate salts. In another embodiment, compounds of Formula I bed herein can be formed as a gentisate salt in l forms A and/or B.
One embodiment includes resiquimod in the form of a gentisate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 6 to about 7.5 degrees 26, about 8 to about 9 degrees 26, about to about 11 degrees 26, about 14 to about 15 degrees 26, about 16 to about 17 degrees 26, about 18 to about 19 degrees 26, about 20 to about 21.5 degrees 26, and/or about 22.5 to about 23.5 degrees 26.
One embodiment includes resiquimod in the form of a gentisate salt in crystal form B. The crystal form B can be characterized by x-ray powder diffraction um that comprises peaks at about 6 to about 7 degrees 26, about 10 to about 10.5 degrees 26, about 12 to about 13 degrees 26, about 20 to about 21 s 26, about 24 to about 24.5 degrees 26, and/or about 26 to about 26.5 degrees 26.
Formula | compounds can also be formed as benzoate salts. In another embodiment, compounds of a I described herein can be formed as a benzoate salt in crystal forms A and/or B.
One ment includes resiquimod in the form of a benzoate salt in crystal form A. The crystal form A can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 9 degrees 26, about 10 to about 11.5 degrees 26, about 12 to about 12.5 degrees 26, about 14 to about 16.5 degrees 26, about 19 to about 22 degrees 26, about 23.5 to about 24.5 degrees 26, about 28.5 to about 29 degrees 26, and/or about 29 to about 30 degrees 26.
] One ment includes resiquimod in the form of a benzoate salt in crystal form B. The crystal form B can be characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8.5 s 26, about 12 to about 14 degrees 26, about 18 to about 19 degrees 26, about 19.5 to about 20.5 degrees 26, about 21 to about 23 degrees 26, about 24 to about 25 degrees 26, and/or about 26 to about 27 degrees 26.
Formula | compounds can also be formed as nitrate salts. In another embodiment, nds of Formula I described herein can be formed as a nitrate salt in crystal forms A and/or B.
One embodiment includes resiquimod in the form of a nitrate salt in crystal form A.
The crystal form A can be characterized by x-ray powder diffraction spectrum that ses peaks at about 9 to about 9.5 degrees 26, about 10 to about 10.5 degrees 26, about 11.5 to about 12.5 degrees 26, about 14 to about 15 degrees 26, about 16 to about 17.5 degrees 26, about 20 to about 22.5 degrees 26, about 25 to about 26 degrees 26, and/or about 28.5 to about 29.5 degrees 26.
One embodiment includes resiquimod in the form of a nitrate salt in crystal form B.
The crystal form B can be characterized by x-ray powder diffraction spectrum that ses peaks at about 9 to about 10 degrees 26, about 12.5 to about 13 degrees 26, about 14 to about 16 degrees 26, about 19.5 to about 21 s 26, about 25 to about 26 degrees 26, and/or about 26 to about 27 degrees 26.
] In some embodiments, a monosulfate salt in crystal form A can t to a hemi- sulfate at water activity of 0.8. Further, a monosulfate salt in crystal form A can demonstrate good physicochemical stability at 80 °C for 24 hours or more.
In some embodiments, crystal forms of compounds described herein have an XRPD pattern of Fig. 1, Fig. 4, Fig. 7, Fig. 10, Fig. 13, Fig. 16, Fig. 19, Fig. 48, Fig. 50, Fig. 54, Fig. 59, Fig. 61, Fig. 63, Fig. 65, Fig. 67, Fig. 69, Fig. 71, Fig. 73, Fig. 75, Fig. 77, Fig. 78, Fig. 79, Fig. 88, Fig. 84, Fig. 85, Fig. 89, Fig. 90, Fig. 98, Fig. 94, Fig. 97, Fig. 98, Fig. 101, Fig. 102, Fig. 105, Fig. 107, Fig. 108, Fig. 111, Fig. 112, Fig. 115, Fig. 118, Fig. 119, Fig. 120, Fig. 124, Fig. 128, Fig. 127, Fig. 128, Fig. 182, Fig. 184, Fig. 188, or Fig. 188.
In some ments, crystal forms of Formula | compounds may be slightly hygroscopic. In other embodiments, crystal forms of Formula | compounds may be non- copic. These crystal forms may also t no form change after a Dynamic Vapor Sorption (DVS) test.
In other ments, when compared to a freebase form of a Formula | compound, crystal forms show improved or comparable solubility in water and bio-relevant media at room ature.
Further, the crystal forms can have physical and chemical stability when ed to a Formula | compound freebase. In some embodiments, no form change and/or purity decrease may be exhibited when compared to freebase form, at 25 °C/60%RH and 40 °C/75%RH. In some embodiments, this absence of form change and/or purity decrease may remain for at least about 1 week, at least about 2 weeks, at least about 1 month, at least about 6 months, or at least about 1 year.
Another aspect provides improved aqueous lity of Formula | compound crystal forms compared with the parent compound or a freebase form thereof. r aspect provides improved aqueous solubility of resiquimod crystal forms compared with the parent compound or a freebase form f.
] Another aspect provides modified oral bioavailability values of Formula | compound crystal forms compared with the orally delivered parent compound or a freebase form thereof. Another aspect es modified oral bioavailability values of resiquimod crystal forms compared with the orally delivered parent compound or a se form thereof.
The techniques and approaches set forth in the present disclosure can further be used by the person of ry skill in the art to prepare variants thereof; the variants are considered to be part of the present disclosure.
] The presently described crystal forms of Formula | compounds can be used to treat a disease or condition. In some embodiments, the disease or condition is a cancer.
Cancers can include breast cancer, head and neck cancer, ovarian cancer, uterine cancer, skin cancer, brain cancer, bladder cancer, thyroid , liver cancer, pancreatic cancer, lung , ocular cancer, throat cancer, esophageal cancer, stomach cancer, intestinal cancer, rectal cancer, testicular cancer, ovarian cancer, vaginal cancer, bone cancer, blood cancer, prostate cancer, and the like.
The presently described crystal forms of Formula | compounds can be used to treat a tumor in a subject in need thereof. In some ments, the tumor is a carcinoma, a sarcoma, a b|astomas, or a combination thereof.
A carcinoma can include, without limitation, an adrenal gland tumor, a bone tumor, a brain tumor, a breast tumor, a bronchi tumor, a colon tumor, a gallbladder tumor, a kidney tumor, a larynx tumor, a liver tumor, a lung tumor, a neural tumor, a pancreatic tumor, a prostate tumor, a parathyroid tumor, a skin tumor, a stomach tumor, and a thyroid tumor. In other aspects of this embodiment, a carcinoma es, without limitation, an adenocarcinoma, an adenosquamous carcinoma, an anaplastic oma, a large cell carcinoma, a small cell carcinoma, and a squamous cell carcinoma. In other aspects of this embodiment, a carcinoma includes, without limitation, a small cell carcinoma, a combined small cell carcinoma, a verrucous carcinoma, a squamous cell carcinoma, a basal cell carcinoma, a transitional cell carcinoma, an ed papilloma, a linitis plastica, a familial adenomatous polyposis, an insulinoma, a glucagonoma, a gastrinoma, a VlPoma, a somatostatinoma, a cholangiocarcinoma, a Klatskin tumor, a hepatocellular adenoma, a hepatocellular carcinoma, a renal cell carcinoma, a endometrioid tumor, a renal oncocytoma, a prolactinoma, a multiple endocrine neoplasia, an adrenocortical adenoma, an adrenocortical carcinoma, a Hurthle cell, a neuroendocrine tumor, an adenoid cystic carcinoma, an oncocytoma, a clear cell arcinoma, an apudoma, a cylindroma, a papillary hidradenoma, a hidrocystoma, a syringoma, a syringocystadenoma papilliferum, a enoma, a cystadenocarcinoma, signet ring cell carcinoma, a mucinous cystadenoma, a mucinous cystadenocarcinoma, a mucoepidermoid carcinoma, an n serous cystadenoma, a atic serous enoma, a serous cystadenocarcinoma, a papillary serous cystadenocarcinoma, a mammary ductal carcinoma, a pancreatic ductal carcinoma, a comedocarcinoma, a Paget's disease of the , an extramammary Paget's disease, a lobular carcinoma in situ, an invasive lobular carcinoma, a medullary carcinoma of the , a medullary thyroid cancer, an acinic cell carcinoma, a Warthin's tumor, or a a.
Sarcomas can include, t tion, a soft tissue sarcoma, a connective tissue sarcoma, a lipomatous sarcoma, a myomatous sarcoma, a x mixed and stromal sarcoma, and a mesothelial. ln aspects of this embodiment, a non-hematologic sarcoma es, without tion, an adenomatoid tumor, an adenomyoma, an aggressive infantile fibromatosis, an alveolar rhabdomyosarcoma, an angiolipoleiomyoma, an angiomyolipoma, an angioleiomyoma, an angiomyxoma, an angiosarcoma, an aponeurotic fibroma, an Askin's tumor, an atypical fibroxanthoma, a benign lipoblastomatosis, a Brenner tumor, a carcinosarcoma, a chondroid lipoma, a chondrosarcoma, a clear-cell sarcoma, a clear-cell a of the , a collagenous fibroma, a cystosarcoma phyllodes, a dermatofibrosarcoma, a dermatofibrosarcoma protuberans (DFSP), a desmoplastic fibroma, a desmoplastic small round cell tumor, a diffuse infantile fibromatosis, an Ewing's/PNET sarcoma, a al scular fibroma, a fibroadenoma, a fibroma of tendon sheath, a fibromatosis colli, a fibrous histiocytoma, a fibrosarcoma, a gastrointestinal stromal tumor (GIST), a genital leiomyoma, a hemangioendothelioma, a hepatoblastoma, a hibernoma, a histiocytoma, an infantile digital atosis, an intradermal spindle cell lipoma, a juvenile hyaline fibromatosis, a Kaposi's sarcoma, a leiomyosarcoma, a liposarcoma, a mesoblastic nephroma, a mesothelioma, a mixed rian tumor, a multiple cutaneous leiomyoma, a multiple cutaneous and uterine leiomyomatosis syndrome, a myelolipoma, a coma, a myxoid liposarcoma, a myxosarcoma, a neural fibrolipoma, a neurofibrosarcoma, an oral submucous fibrosis, an ossifying fibromyxoid tumor, an arcoma, a pancreatoblastoma, a phyllodes tumor, a plantar fibromatosis, a pleomorphic adenoma, a pleomorphic fibroma, a pleomorphic lipoma, a rhabdomyosarcoma, a sarcoma botryoides, a schwannoma sarcoma, a solitary cutaneous leiomyoma, a solitary fibrous tumor, a spindle cell , a stromal tumor of undetermined malignant potential ), a al sarcoma, a vascular sarcoma, or a Vlfilms' tumor.
Blastomas can include, t limitation, a chondroblastoma, a hepatoblastoma, a medulloblastoma, a nephroblastoma, a neuroblastoma, a pancreatoblastoma, a pleuropulmonary blastoma, a retinoblastoma, or a lioblastoma multiforme.
Resiquimod and related Formula | compounds described herein can be agonists of TLR7/TLR8. Studies have found that many solid , such as breast cancer, head and neck cancer, or ovarian cancer, have pDC's invasion and factors ed by tumor cells that inhibit DC maturation. These immature DC cells did not play a role in promoting anti-tumor immunity. By contrast, DCs within the tumor microenvironment promote tumor growth by inhibiting antitumor ty and by promoting angiogenesis. There is evidence that Toll-like receptor 7 agonist imiquimod, and Toll-like receptor 9 agonist CpG drugs can ate pDC within the tumor microenvironment to inhibit tumor development.
Natural killer (NK) cells are a type of cytotoxic lymphocyte that constitutes a major component of the immune . NK cells are a subset of peripheral blood lymphocytes defined by the expression of CD56 or CD 16 and the e of the T cell receptor (CD3). They recognize and kill transformed cell lines without priming in an MHC-unrestricted fashion. NK cells play a major role in the rejection of tumors and cells infected by viruses. The process by which an NK cell recognizes a target cell and rs a sufficient signal to trigger target lysis is determined by an array of inhibitory and activating receptors on the cell e. NK mination of self from altered self involves inhibitory receptor recognition of MHC-l molecules and non-MHC ligands like CD48 and Clr-1b. NK recognition of infected or damaged cells (altered self) is coordinated through stress d ligands (e.g., MICA, MICB, Rae1, H60, Mult1) or virally d s (e.g., m157, hemagluttinin) recognized by various activating receptors, including NKGZD, Ly49H and NKp46/Ncr1.
] NK cells represent the predominant lymphoid cell in the peripheral blood for many months after allogeneic or autologous stem cell transplant and they have a primary role in immunity to pathogens during this period.
Human NK cells mediate the lysis of tumor cells and virus-infected cells via natural cytotoxicity and antibody-dependent cellular cytotoxicity (ADCC).
Human NK cells are lled by positive and negative cytolytic s. Negative (inhibitory) signals are uced by in domain containing receptors CD94/NKGZA and by some Killer lmmunoglobulin-like Receptors (Kle). The regulation of NK lysis by inhibitory signals is known as the “missing self” hypothesis in which specific HLA-class l alleles expressed on the target cell e ligate inhibitory receptors on NK cells. The down-regulation of HLA molecules on tumor cells and some virally infected cells (e.g. CMV) lowers this inhibition below a target threshold and the target cells may become susceptible to NK cell- mediated lysis if the target cells also carry NK—priming and activating molecules. TLR7, TLR8 or TLR9 agonists can activate both mDC and pDCs to produce type | lFNs and express costimulatory molecules such as GlTR-ligand, which subsequently activate NK cells to produce lFN-g and potently promote NK cell killing on.
Inhibitory receptors fall into two groups, those of the lg-superfamily called Killer globulin-like Receptors (Kle) and those of the lectin , the NKGZ, which form dimers with CD94 at the cell surface. Kle have a 2- or 3-domain extracellular structure and bind to HLA-A, -B or -C. The NKGZ/CD94 xes ligate HLA-E.
Inhibitory Kle have up to 4 intracellular domains which contain lTlMs and the best characterized are KlR2DL1, KIR2DL2 and KIR2DL3 which are known to bind HLA-C molecules. K|R2DL2 and KIR2DL3 bind the group 1 HLA-C alleles while K|R2DL1 binds to group 2 alleles. Certain leukemia/lymphoma cells express both group 1 and 2 HLA-C alleles and are known to be resistant to NK—mediated cell lysis.
Vlfith regards to positive activating signals, ADCC is thought to be mediated via CD 16, and a number of triggering receptors responsible for natural cytotoxicity have been identified, including CD2, CD38, CD69, NKRP-l, CD40, B7-2, NK—TR, NKp46, NKp30 and NKp44. In addition, several KIR molecules with short ytoplasmic tails are also stimulatory. These Kle (KIR2D81, K|R2D82 and KIR2DS4) are known to bind to HLA-C; their ellular domains being identical to their related inhibitory Kle. The activatory Kle lack the lTlMs and instead associate with DAP 12 leading to NK cell activation. The mechanism of control of expression of inhibitory versus tory Kle s unknown.
Several reports have described the expression of TLRs in mouse or human cancer or cancer cell lines. For example, TLR1 to TLR6 are expressed by colon, lung, prostate, and melanoma mouse tumor cell lines, TLR3 is expressed in human breast cancer cells, hepatocarcinoma and gastric carcinoma cells express TLR2 and TLR4, and TLR9 and TLR4 are expressed by human lung cancer cells. TLR7 and TLR8 are found in tumor cells of human lung cancer.
TLR are a family of proteins that sense a microbial product and/or initiates an adaptive immune response. TLRs te a dendritic cell (DC). TLRs are conserved membrane spanning molecules containing an ectodomain of leucine-rich repeats, a transmembrane domain and an intracellular TIR (Toll/interleukin receptor) domain. TLRs recognize distinct structures in microbes, often referred to as “PAMPs” (pathogen ated molecular patterns). Ligand binding to TLRs invokes a cascade of intra-cellular signaling pathways that induce the production of factors involved in inflammation and immunity.
TLR7 and TLR8 are phylogenetically and structurally related. TLR7 is selectively expressed by human pDCs and B cells. TLR8 is predominantly sed mDCs, monocytes, macrophages and myeloid suppressor cells. TLR7-specific agonists te plasmacytoid DCs (pDCs) to produce large amounts of type 1 lFNs and expressing high levels of costimulatory molecules that promote activation of T cells, NK cells, B cells and mDCs. TLR8-specific agonists activate d DCs, monocytes, macrophages or myeloid- derived suppressor cells to produce large amounts of type 1 lFN, lL-12 and lL-23, and express high levels of MHC class I, MHC class II and costimulatory molecules that promote the activation of antigen specific CD4 and CD8+ T cells.
Pharmaceutical compositions ing the l forms of the herein described compounds can be administered to an animal, such as a mammal. In some embodiments, the mammal can be a human, a cat, a dog, a horse, a pig, a cow, a whale, or the like.
Pharmaceutical itions may be prepared by combining a therapeutically effective amount of at least one crystal form of a herein described compound with conventional acceptable pharmaceutical excipients, and by preparation of unit dosage forms le for therapeutic use. As used herein, the term “pharmaceutical composition” and refers to a therapeutically effective concentration of an active compound, such as, e.g., any of the l forms of herein described nds. Preferably, the pharmaceutical composition does not produce an adverse, allergic, or other untoward or ed on when administered. A pharmaceutical composition disclosed herein can be useful for medical and veterinary applications. A ceutical composition may be administered alone, or in combination with other supplementary active compounds, agents, drugs or hormones. The pharmaceutical compositions may be ctured using any of a variety of processes, including, without limitation, conventional mixing, dissolving, granulating, - making, ting, emulsifying, encapsulating, entrapping, and lyophilizing. The pharmaceutical composition can take any of a variety of forms including, without limitation, a sterile solution, suspension, emulsion, lyophilizate, tablet, pill, pellet, capsule, powder, syrup, elixir, or any other dosage form suitable for administration.
A pharmaceutical ition can be a liquid formulation, olid formulation, or a solid formulation. A formulation disclosed herein can be produced in a manner to form one phase, such as, e.g., an oil or a solid. Alternatively, a formulation disclosed herein can be produced in a manner to form two phase, such as, e.g., an emulsion. A pharmaceutical composition disclosed herein intended for such administration may be prepared ing to any method known to the art for the manufacture of pharmaceutical compositions.
] Liquid formulations suitable for injection or topical (e.g., ocular) delivery may comprise physiologically able sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions and sterile powders for reconstitution into sterile injectable ons or sions. Examples of suitable aqueous and non-aqueous carriers, diluents, solvents or vehicles include water, ethanol, s (propylene glycol, polyethyleneglycol (PEG), glycerol, and the like), suitable mixtures thereof, vegetable oils (such as olive oil) and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of a coating such as in, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants.
Semi-solid formulations suitable for topical administration include, without limitation, ointments, creams, salves, and gels. In such solid formulations, the active nd may be admixed with at least one inert customary excipient (or carrier) such as, a lipid and/or polyethylene glycol.
Solid formulations suitable for oral administration include capsules, tablets, pills, powders and granules. In such solid formulations, the active compound may be admixed with at least one inert customary excipient (or carrier) such as sodium e or dicalcium ate or (a) fillers or extenders, as for example, starches, lactose, sucrose, glucose, mannitol and silicic acid, (b) binders, as for example, carboxymethylcellulose, alignates, n, polyvinylpyrrolidone, sucrose and acacia, (c) humectants, as for example, glycerol, (d) disintegrating agents, as for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, n complex silicates and sodium carbonate, (e) solution retarders, WO 32725 as for example, in, (f) absorption accelerators, as for example, quaternary um compounds, (g) wetting agents, as for example, cetyl alcohol and glycerol monostearate, (h) adsorbents, as for example, kaolin and ite, and (i) lubricants, as for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate or mixtures thereof. In the case of capsules, tablets and pills, the dosage forms may also comprise ing agents.
In liquid and olid formulations, a concentration of crystal forms of the herein described compounds may be between about 50 mg/mL to about 1,000 mg/mL. ln aspects of this embodiment, a therapeutically effective amount of a crystal form of the herein described compounds may be from, e.g., about 50 mg/mL to about 100 mg/mL, about 50 mg/mL to about 200 mg/mL, about 50 mg/mL to about 300 mg/mL, about 50 mg/mL to about 400 mg/mL, about 50 mg/mL to about 500 mg/mL, about 50 mg/mL to about 600 mg/mL, about 50 mg/mL to about 700 mg/mL, about 50 mg/mL to about 800 mg/mL, about 50 mg/mL to about 900 mg/mL, about 50 mg/mL to about 1,000 mg/mL, about 100 mg/mL to about 200 mg/mL, about 100 mg/mL to about 300 mg/mL, about 100 mg/mL to about 400 mg/mL, about 100 mg/mL to about 500 mg/mL, about 100 mg/mL to about 600 mg/mL, about 100 mg/mL to about 700 mg/mL, about 100 mg/mL to about 800 mg/mL, about 100 mg/mL to about 900 mg/mL, about 100 mg/mL to about 1,000 mg/mL, about 200 mg/mL to about 300 mg/mL, about 200 mg/mL to about 400 mg/mL, about 200 mg/mL to about 500 mg/mL, about 200 mg/mL to about 600 mg/mL, about 200 mg/mL to about 700 mg/mL, about 200 mg/mL to about 800 mg/mL, about 200 mg/mL to about 900 mg/mL, about 200 mg/mL to about 1,000 mg/mL, about 300 mg/mL to about 400 mg/mL, about 300 mg/mL to about 500 mg/mL, about 300 mg/mL to about 600 mg/mL, about 300 mg/mL to about 700 mg/mL, about 300 mg/mL to about 800 mg/mL, about 300 mg/mL to about 900 mg/mL, about 300 mg/mL to about 1,000 mg/mL, about 400 mg/mL to about 500 mg/mL, about 400 mg/mL to about 600 mg/mL, about 400 mg/mL to about 700 mg/mL, about 400 mg/mL to about 800 mg/mL, about 400 mg/mL to about 900 mg/mL, about 400 mg/mL to about 1,000 mg/mL, about 500 mg/mL to about 600 mg/mL, about 500 mg/mL to about 700 mg/mL, about 500 mg/mL to about 800 mg/mL, about 500 mg/mL to about 900 mg/mL, about 500 mg/mL to about 1,000 mg/mL, about 600 mg/mL to about 700 mg/mL, about 600 mg/mL to about 800 mg/mL, about 600 mg/mL to about 900 mg/mL, or about 600 mg/mL to about 1,000 mg/mL.
In semi-solid and solid formulations, an amount of a crystal form of the herein described compounds may be between about 0. 01% to about 45% by weight. In aspects of this embodiment, an amount of a crystal form of the herein described compounds may be from, e.g., about 0.1% to about 45% by weight, about 0.1% to about 40% by weight, about 0.1% to about 35% by weight, about 0.1 % to about 30% by weight, about 0.1% to about 25% by weight, about 0.1% to about 20% by weight, about 0.1% to about 15% by , about 0.1% to about 10% by weight, about 0.1% to about 5% by weight, about 1% to about 45% by weight, about 1% to about 40% by , about 1% to about 35% by weight, about 1% to about 30% by weight, about 1% to about 25% by , about 1% to about 20% by weight, about 1% to about 15% by weight, about 1% to about 10% by weight, about 1% to about 5% by weight, about 5% to about 45% by weight, about 5% to about 40% by weight, about 5% to about 35% by weight, about 5% to about 30% by weight, about 5% to about 25% by weight, about 5% to about 20% by weight, about 5% to about 15% by weight, about 5% to about 10% by weight, about 10% to about 45% by , about 10% to about 40% by weight, about 10% to about 35% by weight, about 10% to about 30% by weight, about 10% to about 25% by weight, about 10% to about 20% by weight, about 10% to about 15% by weight, about 15% to about 45% by weight, about 15% to about 40% by weight, about 15% to about 35% by weight, about 15% to about 30% by weight, about 15% to about 25% by weight, about 15% to about 20% by weight, about 20% to about 45% by weight, about 20% to about 40% by weight, about 20% to about 35% by weight, about 20% to about 30% by weight, about 20% to about 25% by weight, about 25% to about 45% by , about 25% to about 40% by weight, about 25% to about 35% by weight, or about 25% to about 30% by weight.
A ceutical ition disclosed herein can optionally include a pharmaceutically acceptable carrier that facilitates processing of an active compound into pharmaceutically acceptable compositions. Such a carrier generally is mixed with an active compound or permitted to dilute or enclose the active nd and can be a solid, semi- solid, or liquid agent. Any of a variety of pharmaceutically acceptable carriers can be used including, without tion, aqueous media such as, e.g., water, saline, glycine, onic acid and the like; solid carriers such as, e.g., starch, magnesium stearate, mannitol, sodium saccharin, talcum, cellulose, glucose, sucrose, lactose, trehalose, magnesium carbonate, and the like; solvents; dispersion media; coatings; antibacterial and ngal agents; ic and tion delaying agents; or any other inactive ingredient.
A pharmaceutical composition disclosed herein can optionally include, without limitation, other pharmaceutically able components (or pharmaceutical components), including, without limitation, buffers, preservatives, tonicity adjusters, salts, antioxidants, osmolality adjusting , physiological substances, pharmacological substances, bulking agents, emulsifying agents, wetting agents, sweetening or flavoring agents, and the like.
Various buffers and means for adjusting pH can be used to prepare a pharmaceutical composition disclosed herein, provided that the resulting preparation is pharmaceutically acceptable. Such buffers include, without limitation, acetate buffers, borate buffers, citrate s, phosphate buffers, neutral buffered saline, and phosphate buffered . It is understood that acids or bases can be used to adjust the pH of a composition as needed. ceutically able antioxidants include, without limitation, sodium metabisulfite, sodium thiosulfate, acetylcysteine, butylated hydroxyanisole, and butylated hydroxytoluene.
Useful preservatives include, without limitation, benzalkonium chloride, chlorobutanol, thimerosal, phenylmercuric e, phenylmercuric nitrate, a stabilized oxy chloro ition, such as, e.g., sodium chlorite and chelants, such as, e.g., DTPA or DTPA- bisamide, calcium DTPA, and CaNaDTPA-bisamide. Tonicity adjustors useful in a pharmaceutical composition include, t limitation, salts such as, e.g., sodium chloride, potassium chloride, mannitol or glycerin and other pharmaceutically acceptable tonicity adjustor.
Method of treating a disease or condition, such as a cancer, include administering a a | new crystal form such as in a pharmaceutical form. Administration can be once daily, twice daily, three times daily four times daily or more. In other embodiments, administration can be one every other day, once every three days, every four day, every five days, every six days, one a week, once every other week, once every three weeks, once a month, once every other month, every six months, once a year, or the like.
Examples Example 1 1. Summary Salt screening for resiquimod freebase was conducted to identify salt candidates with suitable physicochemical properties. Additionally, polymorph screening was carried out to identify g crystal forms of the salt candidate. l salt screening was performed under 100 conditions using 19 acids (two molar ratios of HCI) and five solvent systems. A total of 32 crystalline hits were isolated and characterized by X—ray powder diffraction (XRPD), thermogravimetric analysis (TGA), differential ng calorimetry (DSC), with the stoichiometry determined using proton nuclear magnetic resonance (1H NMR) or HPLC/IC. Considering the safety class of acids used, number of polymorphs observed, and physicochemical properties, seven lline salts namely mono-HCI salt, di-HCI salt, sulfate, phosphate, maleate, malate, and adipate were ed as leading salts for further evaluation.
All the seven salt leads were re-prepared to hundreds of milligrams (except di-HCI salt was provided) and further evaluated on hygroscopicity, kinetic solubility, and solid-state stability. As results show, 1) all salt leads were slightly hygroscopic with no form change WO 32725 after DVS test except HCI salts, 2) compared with freebase Type A, all salt leads show improved or able solubility in water and bio-relevant media (SGF, FaSSlF, and FeSSlF) at room temperature (RT, 20 1r 3 °C) except maleate, 3) no form change and decrease of HPLC purity were ed for salt leads and freebase Type A under 25 °C/60%RH and 40 RH for one week except di-HCI salt, suggesting good physical and chemical stability.
Based on the characterization and evaluation results, sulfate was selected as a salt ate and re-prepared on 6-g scale for polymorphism investigation. Using sulfate Type A as starting material, a polymorph screening was performed under 100 conditions. A total of three crystal forms were obtained, ing one anhydrate (Type A), one DMSO solvate (Type B), and one hemi-sulfate. Thus, sulfate Type A was selected as the leading form of mono-sulfate. Disproportionation risk and thermo-stability were investigated on sulfate Type A, and the result shows, 1) sulfate Type A converted to hemi-sulfate at water activity of 0.8, indicating the disproportionation risk at high relative humidity, 2) sulfate Type A shows good physicochemical stability at 80 °C for 24 hours. 2. Salt ing 2.1 Experiment Summary According to the pKa value of 7.2 determined on Sirius T3 and approximate solubility of freebase (807919A) at RT, nineteen salt formers and five solvent systems were used for the screening. se was dispersed in selected solvent with a glass vial and corresponding salt former was added with a molar charge ratio of 1:1 (for HCI/freebase, two ratios of both 1:1 and 2:1 were used). The mixtures of freebase and acid were stirred at RT for 2.5 days. Clear solutions obtained were slurried at 5 °C overnight to induce precipitation. If the s were still clear, they would be subjected to evaporation to dryness at RT, in order to maximize the chance of identifying as many crystalline hits as possible. Resulted solids were isolated and analyzed by XRPD.
As summarized in Table 2-1, a total of 32 crystalline hits were obtained and terized by TGA and DSC, with the stoichiometry determined by 1H NMR or HPLC/IC.
The characterization data were summarized in Table 2-2, and ed information was provided in Section 5.4.
Table 2-1 Summary of salt ing experiments seAType freebaseAType freebaseAType freebaseAType freebaseAType HCI salt Type HCI salt Type HCI salt Type A HCI salt Type A HCI salt Type A A+FB Type A* A+FB Type A* HCI saltAType HCI salt Type A HCI salt Type A HCI salt Type B HCI salt Type A sulfate Type A sulfate Type A sulfate Type A sulfate Type A* _ypep_hosphatemaleate A e Type A e_ypeA maleate Type A maleate Type A Type ate Typep_hosphateType phosphate Type phosphate Type _ypef_umaratetartrate A tartrate Type C _ypetartrate A tartrate Type A tartrate Type B AType fumarateAType f_umarateCType fumarateAType teEaType citrate Type A citrate Type B citrate Type A citrate Type A ate Type glycolate Type glycolate Type glycolate Type glycolate Type _ypeh_ippurateTypemalate A malate Type A malate Type A malate Type A malate Type A hippurate Type hippurate Type hippurate Type hippurate Type _ype_TAypelactate B lactate Type A* lactate Type A lactate Type A** e Type A* succinate succinateAType succinateAType succinateAType succinateEaType _aipateType A adipate Type A adipate Type A adipate Type A adipate Type A tosylate Type tosylate Type A te Type A tosylate Type A tosylate Type B te Type mesylate Type mesylate Type mesylate Type mesylate Type A+FB Type A* _ype_Typexoalate A oxalate Type A _ypeoxalate A _ypeoxalate B oxalate Type A* gentisate gentisate Type _Typegentisate _Typegentisate gentisate Type benzoateAType benzoateAType benzoateAType benzoateAType benzoateEBType nitrate Type nitrate Type A nitrate Type B nitrate Type A nitrate Type A B+FB Type A* **: Solids were generated via slurry at 5 °C overnight. FB: freebase *: Samples were obtained via evaporation to s at RT.
Blank experiments were set up to detect any possible change of freebase.
Table 2-2 Characterization summary of crystalline hits 807919 07 D1 250.0, 266.6 1.00 103.5, 110.2, 36.46 807919C2 181.1, 249.8, 266.0 98.08 807919 07-A3 229.9, 238.0, 807919-07 A7 0.81 252.9 109.6, 226.8, 116.08 807919-07 E7 0.61 237.9, 255.9 156.3, 237.8, 807919C7 1.03 248.8 807919-07 A8 165.4 1.02 192.13 807919B8 197.7 0.53 85.4, 159.5, 807919C12 1.07 90.08 169.4 807919-07 A12 142.9, 160.2 807919C13 175.8 1.00 118.09 84, 807919 07-E13 0.52 209.6 172.21 807919-07 B15 202.5 0.89 61.1, 185.4, 807919D15 . 0.92 189.9, 201.9 90.04 807919D17 190.5, 218.2 154.12 122.12 807919E19 102.5, 200.4m 63.02 Samples were dried at 50 °C overnight before characterization. 2.2 Re-preparation and Characterization of Salt Leads Based on the characterization results, seven salt leads were selected and re- prepared to hundreds of rams (except di-HCI salt Type A). The selection criteria include but not limited to: 1) low safety concern of acid (safety class I), 2) sharp x-ray powder diffraction (XRPD) peaks without apparent amorphous halo, 3) negligible weight loss in gravimetric analysis (TGA), and 4) neat thermal event with a sharp melting in differential scanning calorimetry (DSC). Preparation procedures for salt leads as well as other salts bed herein are described in Table 2-3.
Table 2-3 Preparation procedures of salts Crystal Form Preparation Procedures . Weigh 200 mg of freebase (807919A) into 5 mL tetrahydrofuran (THF) and stir at 50 °C to dissolve the sample.
. Add 59 uL HCI (charge ratio of 1:1) to the freebase solution, and then stir Mono-HCI salt magnetically with a speed of 750 rpm at room temperature (RT).
Type A (807919A) - Add ”2 m9 seed 9D1) Into the . uently, W the suspensnon at RT for 5.5 hrs.
Method 1 . Characterize the wet sample by XRPD and the crystal form conforms to mono-HCI salt Type A.
. Centrifuge and dry the wet cake at 50 °C for 2 hrs followed by drying under Crystal Form Preparation Procedures vacuum at RT for1 hr.
Collect the solids, 205.4 mg with a yield of 92.0%.
:PWN.‘ Weigh 328.5 mg freebase Type A to 10.5 mL THF to get a clear solution.
Mono-HCI salt Pipette 0.5 mL se solution to a 1.5-mL glass vial.
Type A Pipette 4.0 pL HCI to the freebase solution and stir at RT for 2.5 days. (807919A) Isolate the solids by fuging and then dry at 50 °C overnight.
Method 2 Weigh 14.8 mg se Type A to 0.5 mL EtOAc.
HCI Salt Type B Add 8.0 pL HCI to the freebase suspension and stir at RT for 2.5 days. (807919-07—C2) Isolate the solids by centrifuging and then dry at 50 °C overnight.
. Weigh 65 mg of sulfuric acid into a 20-mL glass vial with 5 mL of THF.
Weigh 200 mg of freebase (807919A, charge ratio of 1:1) to the acid solution, and stir magnetically with a speed of 750 rpm at RT.
Sulfate 3. Add ~2 mg seed (807919A3) into the system and still stir at RT ght.
Type A Characterize the wet sample by XRPD and the crystal form conforms to (807919A) sulfate Type A.
Method 1 Cool the suspension to 5 °C at a rate of 0.1 °C/min, and age at 5 °C overnight. fuge and dry the wet cake at 50 °C for 2 hrs followed by drying under vacuum at RT overnight.
Collect the solids, 247.1 mg with a yield of 94.2%.
Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution.
Sulfate Pipette 1.0 mL freebase solution to a 1.5 mL glass vial.
Type A Pipette 2.7 pL sulfuric acid to the freebase solution and stir at RT for 2.5 (807919A) days.
Method 2 4. Isolate the solids by fuging and then dry at 50 °C overnight.
Crystal Form Preparation Procedures Weigh 9.9 mg sulfate Type A to a 3-mL glass vial.
Add 2 mL DMSO to a 20-mL glass vial.
Sulfate Type B Seal the 3-mL vial into the 20-mL vial and keep the system at RT for 7 days. (807919A13) :PWN.‘ Isolate the solids for analysis.
Hemi-sulfate Weigh 14.9 mg sulfate Type A to a 1.5-mL glass vial.
Add 0.5 mL acetone/H20 96, v/v) and stir at RT for 5 days.
Type A e the solids by fuging. (807919A) Method 1 Weigh 60.7 mg freebase Type A to 0.3 mL acetone/H20 (604:396, v/v).
Hemi-sulfate Pipette 5.5 pL ic acid to freebase suspension.
Type A Add ~1 mg seed and stir at RT overnight. (807919A) Isolate the solids by vacuum filter.
Method 2 . Weigh 200 mg of freebase (807919A) into 5 mL THF and stir at 50 °C to dissolve the sample.
. Add 45 pL phosphoric acid (charge ratio of 1:1) to the freebase on, and then stir magnetically with a speed of 750 rpm at RT.
Phosphate . Add ~2 mg seed (807919E5) into the system and still stir at RT overnight.
Type A . Characterize the wet sample by XRPD and the crystal form conforms to (807919C) phosphate Type A.
Method 1 Cool the suspension to 5 °C at a rate of 0.1 °C/min, and age at 5 °C ght.
Centrifuge and dry the wet cake at 50 °C for 2 hrs followed by drying under vacuum at RT overnight.
Collect the solids, 234.0 mg with a yield of 89.2%.
Weigh 330.7 mg freebase Type A to 10.5 mL MeOH/HZO (9:1, v/v) to get a clear solution.
Pipette 0.5 mL freebase solution to a 1.5 mL glass vial.
Type A Pipette 3.2 pL phosphoric acid to the freebase solution and stir at RT for 2.5 (807919C) days.
Method 2 Isolate the solids by centrifuging and then dry at 50 °C overnight.
Maleate . Weigh 200 mg of freebase (807919A) into 5 mL THF and stir at 50 °C to Type A dissolve the sample. (807919B) . Add 82 mg maleic acid (charge ratio of 1 :1) to the freebase solution, and then Method 1 stir magnetically with a speed of 750 rpm at RT.
Crystal Form Preparation Procedures . Add ~2 mg seed (807919D4) into the system and still stir at RT overnight. . terize the wet sample by XRPD and the crystal form conforms to maleate Type A.
Cool the sion to 5 °C at a rate of 0.1 °C/min, and age at 5 °C overnight.
Centrifuge and dry the wet cake at 50 °C for 2 hrs followed by drying under vacuum at RT overnight.
Collect the , 266.5 mg with a yield of 97.3%.
Maleate Weigh 328.5 mg freebase Type A to 10.5 mL THF to get a clear solution.
Add 5.9 mg maleic acid to a 1.5-mL glass vial.
Type A (807919B) PWN.‘ Pipette 0.5 mL freebase solution to the vial and stir at RT for 2.5 days.
Isolate the solids by centrifuging and then dry at 50 °C overnight.
Method 2 . Weigh 200 mg of freebase (807919A) into 5 mL ethanol (EtOH) and stir at 50 °C to dissolve the sample.
. Add 91 mg L-malic acid (charge ratio of 1:1) to the freebase solution, and then stir magnetically with a speed of 750 rpm at RT.
Malate . Add additional 5.0 mL EtOH and ~2 mg seed 9B10) into the Type A system. Subsequently, stirthe sion at RT overnight. (807919E) Characterize the wet sample by XRPD and the crystal form conforms to Method 1 malate Type A.
Cool the suspension to 5 °C at a rate of 0.1 , and age at 5 °C overnight.
Centrifuge and dry the wet cake at 50 °C for 4 hrs followed by drying under vacuum at RT overnight.
Collect the solids, 238.4 mg with a yield of 83.6%.
Malate Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Add 6.8 mg L-malic acid to a 1.5-mL glass vial.
Type A Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days. (807919E) :PWN.‘ Isolate the solids by fuging and then dry at 50 °C overnight.
Method 2 Weigh 150 mg of freebase (807919A) into 5 mL EtOH and stir at 50 °C to dissolve the sample.
Adipate Add 45 mg adipic acid (charge ratio of 1:2, acid/base) to the freebase Type A solution, and then stir magnetically with a speed of 750 rpm at RT. (807919A) Add ~2 mg seed 9B14) into the system. Subsequently, stir the Method 1 suspension at RT overnight.
Characterize the wet sample by XRPD and the crystal form conforms to adipate Type A.
Crystal Form Preparation Procedures . Centrifuge and dry the wet cake under vacuum at RT overnight. 6. Collect the solids, 159.7 mg with a yield of 86.8%. 1. Weigh 329.5 mg se Type A to 16.5 mL EtOH to get a clear solution.
Ad'Ipa et 2. Add 7.1 mg adipic acid to a 1.5-mL glass vial.
Type A 3. Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days. (807919A) 4. Isolate the solids by fuging and then dry at 50 °C overnight._ _ _ _ Method 2 1. Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution. 2. Add 7.5 mg L-tartaric acid to a 1.5-mL glass vial.
Tartrate Type A 3. Pipette 1.0 mL freebase solution to the val and stir at RT for 2.5 days._ _ _ _ (807919A6) 4. Isolate the solids by centrifuging and then dry at 50 °C overnight._ _ _ _ 1. Weigh 330.7 mg freebase Type A to 10.5 mL MeOH/HZO (9:1, v/v) to get a clear on.
Tartrate Type B 2. Add 7.5 mg L-tartaric acid to a 1.5-mL glass vial. (807919E6) 3. Pipette 0.5 mL freebase on to the vial and stir at RT for 2.5 days. 4. Isolate the solids by centrifuging and then dry at 50 °C overnight. 1. Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Tartrate Type C 2. Add 7.6 mg L-tartaric acid to a 1.5-mL glass vial. 9B6) 3. Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days. 4. Isolate the solids by centrifuging and then dry at 50 °C overnight. 1. Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution. 2. Add 5.5 mg c acid to a 1.5-mL glass vial.
Fumarate Type A 3. Pipette 1.0 mL freebase solution to the val and stir at RT for 2.5 days._ _ _ _ (807919A7) 4. Isolate the solids by centrifuging and then dry at 50 °C overnight._ _ _ _ 1. Weigh 330.7 mg freebase Type A to 10.5 mL MeOH/HZO (9:1, v/v) to get a clear solution.
Fumarate Type B 2. Add 5.8 mg fumaric acid to a 1.5-mL glass vial. (807919E7) 3. e 0.5 mL freebase solution to the vial and stir at RT for 2.5 days. 4. Isolate the solids by centrifuging and then dry at 50 °C overnight. 1. Weigh 15.2 mg freebase Type A to 0.5 mL EtOAc. 2. Weigh 5.9 mg fumaric acid to the freebase suspension and stir at RT for 2.5 Fumarate Type C days. (807919C7) 3. e the solids by centrifuging and then dry at 50 °C overnight.
Crystal Form ation Procedures Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution.
Add 9.3 mg citric acid to a 1.5-mL glass vial.
Citrate Type A Pipette 1.0 mL se solution to the vial and stir at RT for 2.5 days. (807919A8) PWN.‘ Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Add 9.6 mg citric acid to a 1.5-mL glass vial. e Type B Pipette 0.8 mL se on to the vial and stir at RT for 2.5 days. (807919B8) PWN.‘ Isolate the solids by centrifuging and then dry at 50 °C overnight.
:PWN.‘ Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Add 4.0 mg glycolic acid to a 1.5-mL glass vial.
Glycolate Type A Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days. (807919B9) Isolate the solids by fuging and then dry at 50 °C overnight.
Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Add 8.4 mg hippuric acid to a 1.5-mL glass vial.
Hippurate Type A (807919B11) :PWN.‘ Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days.
Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 14.8 mg freebase Type A to 0.5 mL EtOAc.
Weigh 5.1 mg L-Iactic acid to the freebase suspension and stir at RT for 2.5 e Type A days. (807919C12) Isolate the solids by centrifuging and then dry at 50 °C overnight.
:PWN.‘ Weigh 330.3 mg freebase Type A to 21 mL e to get a clear solution.
Add 5.2 mg L-Iactic acid to a 1.5-mL glass vial.
Lactate Type B Pipette 1.0 mL freebase solution to the vial and stir at RT for 2.5 days. (807919A12) Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 15.5 mg freebase Type A to 0.5 mL EtOAc.
Weigh 5.6 mg succinic acid to the freebase suspension and stir at RT for 2.5 Succinate Type A days. (807919C13) Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 330.7 mg freebase Type A to 10.5 mL MeOH/HZO (9:1, v/v) to get a Succinate Type B clear solution. (807919E13) Add 5.6 mg succinic acid to a 1.5-mL glass vial. l Form Preparation Procedures e 0.5 mL freebase solution to the vial and stir at RT for 2.5 days.
Isolate the solids by centrifuging and then dry at 50 °C overnight.
:PWN.‘ Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Add 9.6 mg p-toluenesulfonic acid to a 1.5-mL glass vial.
Tosylate Type A Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days. (807919B15) Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 328.5 mg freebase Type A to 10.5 mL THF to get a clear solution.
Add 9.4 mg p-toluenesulfonic acid to a 1.5-mL glass vial.
Tosylate Type B (807919D15) :PWN.‘ e 0.5 mL freebase solution to the vial and stir at RT for 2.5 days.
Isolate the solids by centrifuging and then dry at 50 °C overnight.
:PWN.‘ Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution.
Add 4.6 mg methanesulfonic acid to a 1.5-mL glass vial.
Mesylate Type A Pipette 1.0 mL freebase solution to the vial and stir at RT for 2.5 days. (807919A16) Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution.
Add 6.1 mg oxalic acid to a 1.5-mL glass vial.
Oxalate Type A Pipette 0.8 mL freebase solution to the vial and stir at RT for 2.5 days. (807919B17) :PWN.‘ Isolate the solids by centrifuging and then dry at 50 °C overnight.
PWN.‘ Weigh 328.5 mg se Type A to 10.5 mL THF to get a clear solution.
Add 6.2 mg oxalic acid to a 1.5-mL glass vial.
Oxalate Type B Pipette 0.5 mL freebase solution to the vial and stir at RT for 2.5 days. (807919D17) Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution.
Add 7.3 mg gentisic acid to a 1.5-mL glass vial.
Gentisate Type A (807919A18) :PWN.‘ Pipette 1.0 mL freebase solution to the vial and stir at RT for 2.5 days.
Isolate the solids by centrifuging and then dry at 50 °C overnight.
Weigh 330.7 mg freebase Type A to 10.5 mL MeOH/HZO (9:1, v/v) to get a clear on. ate Type B Add 7.6 mg gensitic acid to a 1.5-mL glass vial. 9E18) Pipette 0.5 mL se solution to the vial and stir at RT for 2.5 days. 4. Isolate the solids by centrifuging and then dry at 50 °C overnight.
WO 32725 Crystal Form Preparation Procedures 1. Weigh 330.3 mg freebase Type A to 21 mL acetone to get a clear solution. 2. Add 6.1 mg benzoic acid to a 1.5-mL glass vial.
Benzoate Type A 3. Pipette 1.0 mL freebase solution to the Vial and stir at RT for 2.5 days._ _ _ _ (807919A19) 4. e the solids by centrifuging and then dry at 50 °C overnight._ _ _ _ 1. Weigh 330.7 mg freebase Type A to 10.5 mL MeOH/HZO (9:1, v/v) to get a clear solution.
Benzoate Type B 2. Add 6.0 mg benzoic acid to a 1.5-mL glass vial. 9E19) 3. Pipette 0.5 mL freebase solution to the vial and stir at RT for 2.5 days. 4. Isolate the solids by centrifuging and then dry at 50 °C overnight. 1. Weigh 328.5 mg freebase Type A to 10.5 mL THF to get a clear solution. 2. e 0.5 mL freebase solution to a 1.5-mL vial.
Nitrate Type A_ 3. Pipette 3.0 uL nitric aCId to the Vial and stir at RT for 2.5 days._ _ _ _ _ _ (807919D20) 4. Isolate the solids by centrifuging and then dry at 50 °C overnight._ _ _ _ 1. Weigh 329.5 mg freebase Type A to 16.5 mL EtOH to get a clear solution. 2. Pipette 0.8 mL freebase solution to a 1.5-mL vial.
Nitrate Type B_ 3. Pipette 3.0 uL nitric aCId to the Vial and stir at RT for 2.5 days._ _ _ _ _ _ (807919B20) 4. Isolate the solids by centrifuging and then dry at 50 °C overnight._ _ _ _ 1 Weigh 15.0 mg se Type A to 0.3 mL ethyl lactate.
Acetate/acetic 2 Weigh 1.1 mg acetic acid to 1.5 mL n-heptane. acid co-crystal 3. Pipette the acid solution to the freebase suspension and stir at RT overnight. (807920A1) 4 Isolate the solids and then dry at t conditions overnight. 2.2.1 Mono-HCI Salt Type A Mono-HCI salt Type A was successfully re-prepared as ced by XRPD results in XRPD data for mono-HCI salt Type A e (peak shift within i0.2°) primary peaks at 20.5, 6.9, and 27.3; secondary peaks at 9.8, 13.7, and 34.3; and tertiary peaks at 17.4, 21.3, and 24.8.
PLM image displayed in illustrated aggregation of small particles (< 10 pm).
As per TGA and DSC data in sample 9-16—A) shows a weight loss of 1.1% up to 130 °C and two endothermic peaks at 250.4 °C and 266.2 °C (peak temperature) before decomposition, indicating an anhydrate for mono-HCI salt Type A. A purity of 99.5 area% was detected by high performance liquid chromatography (HPLC) in Table 2-4. Also, the stoichiometric ratio was ined as 1.01 (acid/base) by HPLC/IC for the re-prepared sample.
Table 2-4 HPLC purity profile of mono-HCI salt Type A (807919A) # RRT Area% # RRT Area% 1 0.75 0.04 3 1.00 99.54 2 0.83 0.38 4 1.43 0.04 2.2.2 Di-HCI Salt Type A Di-HCI salt Type A was characterized by XRPD, TGA, DSC, polarized light microscope (PLM) and HPLC/IC. The XRPD n was shown in and PLM image was displayed in XRPD data for di-HCI salt Type A e (peak shift within i0.2°) primary peaks at 7.1, 8.2, and 19.6; secondary peaks at 15.4, 16.4, and 25.6; and tertiary peaks at 7.3, 14.9, and 27.0.
TGA and DSC results shown in a weight loss of 1.2% up to 100 °C and four endothermic peaks at 99.5 °C, 191.7 °C, 250.7 °C and 261.9 °C (peak temperature) before decomposition. Also, a purity of 99.5 area% was detected by HPLC in Table 2-5 and the stoichiometry was calculated as 2.15 (acid/base) by HPLC/IC.
Table 2-5 HPLC purity e of di-HCI salt Type A (807919-14—A) # RRT Area% # RRT Area% 1 0.75 0.04 3 1.00 99.53 2 0.83 0.39 4 1.43 0.04 2.2.3 Sulfate Type A XRPD patterns comparison in shows that the re-produced sample (807919- 11-A) conformed to sulfate Type A. XRPD data for sulfate Type A provide (peak shift within i0.2°) primary peaks at 7.6, 11.7, and 18.2; secondary peaks at 15.5, 17.6, and 24.4; and tertiary peaks at 9.6, 13.4, and 23.5.
Small particles (< 10 um) and aggregates were rated in TGA and DSC results were displayed in A weight loss of 0.9% was observed up to 130 °C in TGA and the DSC curve shows a sharp melting peak at 214.4 °C (onset temperature) before decomposition. A purity of 99.2 area% was detected by HPLC in Table 26 Also, the stoichiometric ratio was determined as 1.03 (acid/base) for the re-prepared batch. Combined with 1H NMR and TGA/DSC results, sulfate Type A was identified as an anhydrate of mono- sulfate.
Table 2-6 HPLC purity profile of sulfate Type A (807919A) # RRT Area% # RRT Area% 1 0.75 0.05 3 1.00 99.19 2 0.83 0.68 4 1.43 0.09 2.2.4 Phosphate Type A Phosphate Type A was successfully re-prepared as evidenced by XRPD s in . XRPD data for phosphate Type A provide (peak shift within i0.2°) primary peaks at 7.7, 15.3, and 20.6; secondary peaks at 12.4, 18.5, and 25.2; and tertiary peaks at 14.3, 16.7, and 17.7.
] PLM image displayed in illustrated aggregation of small particles (< 10 pm). As per TGA and DSC data in , phosphate Type A (807919C) shows a weight loss of 1.2% up to 130 °C and an endothermic peak at 241.0 °C (onset temperature) before decomposition. A purity of 99.4 area% was detected by HPLC in Table 2-7. Also, the stoichiometry of pared sample was determined as 1.07 (acid/base) by HPLC/IC.
Table 2-7 HPLC purity profile of phosphate Type A (807919—11-C) # RRT Area% # RRT Area% 1 0.83 0.55 3 1.43 0.09 2 1.00 99.36 -- -- -- 2.2.5 Maleate Type A Maleate Type A (807919-07—D4) was generated via reactive llization (molar ratio of 1:1) in THF at RT. XRPD results in shows maleate Type A was successfully re-prepared. XRPD data for maleate Type A provide (peak shift within i0.2°) primary peaks at 7.5, 10.3, and 24.7; secondary peaks at 9.3, 16.5, and 18.0; and tertiary peaks at 15.7, .7, and 21.4.
Small particles (< 10 um) and aggregates were illustrated in . TGA and DSC data shows a weight loss of 1.3% up to 130 °C, and a possible melting erm at 224.1 °C (onset temperature) before decomposition was observed in DSC (). A purity of 99.2 area% was detected by HPLC in Table 2-8. 1H NMR results indicate a stoichiometry of 0.96 (acid/base) for the re-prepared e Type A (807919—11-B). ed with 1H NMR and TGA/DSC s, maleate Type A was identified to be an anhydrate of mono- maleate Table 2-8 HPLC purity profile of maleate Type A (807919B) # RRT Area% # RRT Area% 1 0.75 0.10 3 1.00 99.19 2 0.83 0.63 4 1.43 0.09 2.2.6 Malate Type A Malate Type A was successfully re-prepared as evidenced by XRPD results in . XRPD data for malate Type A provide (peak shift within i0.2°) primary peaks at 6.5, 8.5, and 23.2; secondary peaks at 12.0, 13.0, and 17.1; and tertiary peaks at 8.8, 20.5, and .3.
PLM image displayed in illustrated aggregation of irregular les. As per TGA and DSC data in , malate Type A (807919E) shows a weight loss of 1.0% up to 130 °C and a sharp endothermic peak at 192.9 °C (onset temperature) before decomposition. A purity of 99.9 area % was ed by HPLC in Table 2-9. Further, the stoichiometry of pared sample was determined as 1.02 (acid/base) by 1H NMR.
Table 2-9 HPLC purity profile of malate Type A (807919E) # RRT Area% # RRT Area% 1 0.75 0.55 3 1.43 0.07 2 1.00 99.88 -- -- -- 2.2.7 e Type A XRPD patterns comparison in shows that the re-produced sample 9A) conformed to adipate Type A. XRPD data for adipate Type A provide (peak shift within i0.2°) primary peaks at 5.9, 12.5, and 21.3; secondary peaks at 13.9, 18.8, and 26.7; and tertiary peaks at 14.4, 19.7, and 22.6.
Small particles (< 10 um) and severe aggregates were illustrated in . TGA and DSC results were displayed in . A weight loss of 0.9% was observed up to 130 °C in TGA and the DSC curve shows a sharp melting peak at 218.0 °C (onset temperature) before decomposition. A purity of 99.9 area% was detected by HPLC in Table 2-10. Further, the iometric ratio was determined as 0.52 (acid/base) for the re-prepared batch, suggesting the formation of hemi-adipate.
Table 2-10 HPLC purity profile of adipate Type A (807919A) # RRT Area% # RRT Area% 1 0.83 0.06 2 1.00 99.94 2.3 Evaluation of Salt Leads Further tion study of hygroscopicity, kinetic solubility, and solid-state stability was conducted to better understand the physicochemical properties of seven leads.
As results shows: 1) All salt leads are slightly hygroscopic with no form change after DVS tion except mono-HCI salt Type A and di-HCI salt Type A, 2) Compared with freebase Type A, all salt leads displayed improved or comparable lity in water and bio-relevant media except maleate Type A, 3) As evidenced by no substantial change in crystal form or HPLC purity, all salt leads shows good physical and chemical stability except di-HCI salt Type A. 2.3.1 Hygroscopicity DVS isotherm plot was collected at 25 °C to investigate the solid form stability as a function of humidity. For the six anhydrous salts (mono-HCI salt Type A, sulfate Type A, phosphate Type A, maleate Type A, malate Type A, and adipate Type A), solids were pre- dried at 0% RH to remove the unbounded solvent or water before started. For the possible hydrate/solvate di-HCI salt Type A, solids were brated at ambient ty (~30%RH) before testing.
As evidenced by the water uptake of 0.2~1.1% up to 80%RH, five salt forms (sulfate Type A, phosphate Type A, maleate Type A, malate Type A, and adipate Type A) were slightly hygroscopic. No solid form change was observed for all the five leads after DVS evaluation (-31).
DVS plots displayed in and illustrate that both HCI salt forms were hygroscopic. For mono-HCI salt Type A (807919A), a water uptake of 2.9% was observed up to 80%RH and no form change was detected after DVS test (). For di- HCI salt Type A (807919A), a water uptake of 12.2% was detected up to 80%RH and one plateau was observed at ~20%RH, suggesting the possible existence of a hydrate. In addition, di-HCI salt Type A converted to a new form which contains diffraction peaks of CI salt Type A after DVS evaluation (), ting the disproportionation risk of di-HCI salt Type A at high relative humidity. 2.3.2 Kinetic Solubility Kinetic solubility of seven salt leads was measured in water and three bio-relevant media (SGF, FeSSIF, and ) at RT, using freebase Type A 9A) as control.
All solubility samples (initial solid loading of ~5 mg/mL) were kept rolling on a rolling tor at a speed of 25 rpm, and sampled at 1, 2, 4 and 24 hours, respectively. After being centrifuged and separated using 0.45 pm Nylon , filtrates were collected for HPLC and pH test, and wet cakes for XRPD characterization. lf clear solutions were obtained after 24 hours, accurate concentration and purity were measured for the solutions.
The results were summarized in Table 2-11, and the kinetic solubility profiles are yed in Fle. 36A-D. Compared with freebase Type A, mono-HCl salt Type A, di-HCl salt Type A, e Type A, ate Type A, malate Type A, and adipate Type A shows ed or comparable solubility in water and bio-relevant buffers. Also, remaining solids after suspended 24 hours shows no form change (-38). Meanwhile, decreased solubility was observed in SGF, FaSSlF, and FeSSlF after the formation of mono-maleate (maleate Type A) while no form change was detected after kinetic solubility evaluation (). In addition, no degradation was observed for clear solutions after 24 hours as evidenced by the HPLC results in Table 2-12.
Table 2-11 Summary of kinetic lity results at RT Kinetic Solubility in Water 1 hr 2 hrs 4 hrs 24 hrs Solid Form— S pH FC S pH FC S pH FC S pH FC Mono-HCI C N/A N/A C N/A N/A C N/A N/A 4.5* 4.7 N/A saltTypeA Di-HClsaIt C N/A N/A C N/A N/A C N/A N/A 3.8* 2.0 N/A TypeA Sulfate C N/A N/A C N/A N/A C N/A N/A 4.1* 2.2 N/A TypeA Phosphate C N/A N/A C N/A N/A C N/A N/A 3.9* 4.2 N/A TypeA Malate C N/A N/A C N/A N/A C N/A N/A 3.7* 4.3 N/A TypeA Maleate 0.93 4.2 No 0.98 4.3 No 1.1 4.2 No 0.97 5.2 No TypeA Adipate 2.8 5.9 No 2.7 5.8 No 2.8 5.9 No 2.7 6.0 No TypeA Freebase 0.5 8.4 No 0.5 8.6 No 0.5 8.1 No 0.5 8.1 No TypeA Kinetic Solubility in SGF 1 hr 2 hrs 4 hrs 24 hrs Solid Form S pH FC pH FC FC 3 pH FC C N/A N/A N/A N/A N/A N/A 4.7* 1.8 N/A salt Type A Di-HCI salt C N/A N/A N/A N/A N/A N/A 3.9* 1.6 N/A Type A Sulfate C N/A N/A N/A N/A N/A N/A 3.9* 1.8 N/A Type A Phosphate C N/A N/A N/A N/A N/A N/A 4.0* 2.2 N/A Type A Malate C N/A N/A N/A N/A N/A N/A 4.3* 2.4 N/A Type A Maleate 1.4 2.0 No 1.5 2.0 No 1.6 2.1 No 1 .5 1 .9 No Type A Adipate 4.7 3.4 N/A" N/A N/A N/A N/A 4.7* 3.4 N/A Type A Freebase 2.7 3.6 N/A" 3.5 5.0 N/A" 5.0 5.0 N/A 5.0 4.9 N/A Type A Kinetic Solubility in FaSSlF 1 hr 2 hrs 4 hrs 24 hrs Solid Form S pH FC pH FC FC 3 pH FC Mono-HCI C N/A N/A N/A N/A N/A N/A 4.5* 6.3 N/A salt Type A Di-HCI salt C N/A N/A N/A N/A N/A N/A 3.9* 3.5 N/A Type A Sulfate C N/A N/A N/A N/A N/A N/A 4.0* 3.2 N/A Type A Phosphate C N/A N/A N/A N/A N/A N/A 4.3* 6.4 N/A Type A Malate C N/A N/A N/A N/A N/A N/A 37* 5.5 N/A Type A Maleate 2.2 6.1 No 2.3 6.3 No 2.4 6.1 No 2.3 6.1 No Type A Adipate 4.3 6.4 No 4.5 6.2 N/A" 4.6 6.4 N/A" 4.5* 6.4 N/A Type A Freebase 2.7 7.0 No 2.8 7.0 No 2.8 7.1 No 2.9 7.0 No Type A Kinetic lity in FeSSIF 1hr 2 hrs 4 hrs 24 hrs Solid Form FC pH FC FC S pH FC Mono-HCI N/A N/A N/A N/A N/A N/A 4.6* 5.0 N/A salt Type A Di-HCI salt N/A N/A N/A N/A N/A N/A 3.9* 4.9 N/A Type A Sulfate N/A N/A N/A N/A N/A N/A 4.2* 4.8 N/A Type A N/A N/A N/A N/A N/A N/A 3.8* 5.0 N/A Type A Malate N/A N/A N/A N/A N/A N/A 3.5* 4.8 N/A Type A Maleate 1.5 4.9 No 1.6 5.0 No 1.7 5.0 No 1 .6 5.2 No Type A Adipate N/A N/A N/A N/A N/A N/A 4.2* 5.0 N/A Type A Freebase 4.9 N/A N/A 4.9 N/A N/A 4.9 N/A N/A 4.9* 5.4 N/A Type A S: Solubility, pH: Final pH of supernatant, FC: Solid form change.
C: Clear, N/A: No data was available, N/A*: Limited solid for is.
*: The concentration and pH data of clear solutions were collected.
Table 2-12 HPLC purity results of clear samples after kinetic solubility test Crystal Form HPLC Purity Condition (Batch No.) Area% % of Initial Initial 99.54 Water 99.56 100.0 Mono-HCI salt Type A SGF 99.55 100.0 (807919A) FaSSIF 99.61 100.1 FeSSIF 99.56 100.0 Initial 99.56 Di-HCI salt Type A Water 99.51 99.9 (807919A) SGF 99.56 100.0 FaSSIF 99.57 100.0 WO 32725 Crystal Form HPLC Purity Condition_ _ (Batch N0-) Area% % of Initial FeSSIF 99.57 100.0 Initial 99.32 -- Water 99.31 100.0 Sulfate Type A SGF 99.30 100.0 (807919A) FaSSIF 99.36 100.0 FeSSIF 99.31 100.0 Initial 99.31 -- Water 99.34 100.0 Phosphate Type A SGF 99.33 100.0 (807919C) FaSSIF 99.34 100.0 FeSSIF 99.30 100.0 Initial 99.81 -- Water 99.80 100.0 Malate Type A SGF 99.73 99.9 (807919E) FaSSIF 99.78 100.0 FeSSIF 99.86 100.1 Initial 99.90 -- Adipate Type A SGF 99.87 100.0 (807919A) FeSSIF 99.94 100.0 Freebase Type A Initial 99.32 __ (807919A) FeSSIF 99.25 99.9 2.3.3 Physical and Chemical Stability Physicochemical stability of seven salt leads was evaluated under 25 °C/60%RH and 40 °C/75%RH for one week, using freebase Type A (807919-05—A) as l. Stability samples were terized by XRPD and HPLC, with the results summarized in Table 2-13.
The XRPD patterns were shown from -47, ting no form change for investigated forms except di-HCI salt Type A. Also, no substantial purity change was observed for seven leads and freebase Type A. All the data indicated good physical and chemical ity for mono-HCI salt Type A, sulfate Type A, phosphate Type A, maleate Type A, malate Type A, adipate Type A, and freebase Type A under tested conditions at least one week.
Table 2-13 Stability tion summary of salt leads and freebase Type A Crystal Form HPLC Purity HPLC Purity Form Condition (Batch No.) (Initial, area%) Area% % of l change Freebase Type A 25 °C/60%RH 99.15 99.8 No 99.32 (807919A) 40 RH 99.28 100.0 No Mono-HCI salt Type A 25 °C/60%RH 99.46 99.9 No 99.54 (807919A) 40 °C/75%RH 99.47 99.9 No Di-HCI salt Type A 25 °C/60%RH 99.62 100.1 Yes 99.56 (807919A) 40 °C/75%RH 99.70 100.1 Yes Sulfate Type A 25 °C/60%RH 99.33 100.0 No 99.32 (807919A) 40 °C/75%RH 99.37 100.1 No ate Type A 25 °C/60%RH 99.31 100.0 No 99.31 (807919C) 40 °C/75%RH 99.37 100.1 No Maleate Type A 25 °C/60%RH 99.32 100.0 No 99.32 (807919B) 40 °C/75%RH 99.36 100.0 No Malate Type A 25 °C/60%RH 99.83 100.0 No 99.81 (807919E) 40 °C/75%RH 99.77 100.0 No e Type A 25 RH 99.88 100.0 No 99.90 (807919A) 40 °C/75%RH 99.89 100.0 No 2.4 Conclusions A total of 32 crystalline hits were ted via salt screening. Based on the characterization results, seven salt leads, namely mono-HCI salt Type A, di-HCI salt Type A, sulfate Type A, phosphate Type A, maleate Type A, malate Type A, and adipate Type A, were selected to re-prepared for further evaluation ing hygroscopicity, kinetic solubility, and solid-state stability. Considering the results summarized in Table 2-14 and Table 2-15, sulfate was recommended as a salt candidate for further polymorphism investigation.
Table 2-14 Characterization summary of salt leads and freebase Type A (Mi) Mono-HCI salt Di-HCI salt Sulfate Phosphate Crystal Form Type A Type A Type A Type A Batch No. 807919A 807919A 807919A 807919C Speculated Form Anhydrate Hydrate/Solvate Anhydrate Anhydrate Safety Class* I l l l Stoichiometry 1.01 2.15 1.03 1.07 WO 32725 Mono-HCI salt Di-HCI salt Sulfate Phosphate Crystal Form Type A Type A Type A Type A (acid/base) Crystallinity High High High High Weight Loss (%) 1.1 1.2 0.9 1.2 Endotherm (onset, (99.5,191.7, 246.7, 264.8 214.4 (250.9, 254.2)** °C) 250.7, 261 .9)** HPLC Purity (area%) 99.54 99.53 99.19 99.36 Morphology Small particles (<10 um) and aggregation Water Uptake at 2.9 12.2 0.4 1.1 °C/80% RH Form Change Post No Yes No No DVS Test Kinetic Solubility at >3.8 (water/bio-relevant media) RT (mg/mL) Good chemical ity under 25 °C/60%RH and 40 °C/75%RH at One-week Solid-state least one week for all salt forms except di-HCl salt Type A exhibited form change *: Safety class of acid used, according to Handbook of Pharmaceutical Salts: Properties, Selection and Uses, Wiley-VCH: Zurich, 2002. **: peak temperature Table 2-15 Characterization summary of salt leads and freebase Type A (ll/ll) Maleate Malate Adipate Freebase Crystal Form Type A Type A Type A Type A Batch No. 807919-1 1 -B 807919E 807919A 807919A Speculated Form An hydrate Anhydrate Anhydrate Anhydrate Safety Class* | | | -- Stoichiometry 0.96 1.02 0.52 -- base) Crystallinity High High High High Weight Loss (%) 1.3 1.0 0.9 0.3 Endotherm (onset, °C) 224.1 192.9 218.0 193.3 HPLC Purity (area%) 99.19 99.88 99.94 99.09 Small particles Irregular particles Morphology (<10 um) and Small particles (<10 um)_ and aggregation_ and ation_ aggregation Maleate Malate Adipate se Crystal Form Type A Type A Type A Type A Water Uptake at 0.4 0.2 0.7 0.1 °C/80% RH Form Change Post No No No No DVS Test ~1.0 (water) ~2.7 (water) ~0.5 (water) Kinetic Solubility at RT_ _ _ _ >3.5 (water/bio-_ ~1.5 (bio-relevant_ >4.2 (bio-relevant_ ~2.9 (FaSSIF) (mg/mL) relevant media)_ media)_ media)_ >4.9 eSSIF) One-week Solid-state Good physiochemical stability under 25 °C/60%RH and 40 °C/75%RH Sta b'l'tH y *: Safety class of acid used, according to Handbook of Pharmaceutical Salts: Properties, Selection and Uses, Wiley-VCH: Zurich, 2002. 3 Polymorphism igation on Sulfate 3.1 Polymorph Screening Summary Using re-prepared sulfate Type A 9A) as starting material, polymorph ing experiments were conducted under 100 ions with different crystallization or solid transition methods. The detailed procedures can be found in Section 5.5.
As results summarized in Table 3-1 and Table 3-2, three crystal forms were obtained, with starting sulfate Type A as an anhydrate, sulfate Type B as a DMSO solvate, and hemi-sulfate Type A as a hydrate.
Table 3-1 Summary of polymorph ing experiments Method No. of Experiments Crystal Form olvent Addition 18 Sulfate Type A Solid Vapor Diffusion 13 Sulfate Type A, B on Vapor Diffusion 10 Sulfate Type A Slow Evaporation 8 Sulfate Type A, hemi-sulfate Type A Polymer-induced Crystallization 6 Sulfate Type A Slurry at RT/50 °C 38 Sulfate Type A, hemi-sulfate Type A Slow Cooling 7 Sulfate Type A, B Total 100 Sulfate Type A/B, hemi-sulfate Type A Table 3-2 Characterization y of sulfate forms Crystal Form Crystallization Wt Endotherm Stoichiometry HPLC Comment (Sample ID) Condition Loss (onset, °C) (acid/base) purity (%) (area%) Reactive Sulfate Type A crystallization_ _ _ In 2.0 209.6 1.1 99.4 Anhydrate (807919A) Solid vapor Sulfate Type B DMSO diffusion_ _ _ In 11.7 111.2, 202.2 N/A N/A (807919A13) solvate Reactive Hemi-sulfate Type A crystallization .9 105.3*, 217.0 0.5 99.6 Hydrate (807919A) acetone/H20 (aW=0.8) *: peak temperature 3.1.1 Sulfate Type B Sulfate Type B sample 9A13) was obtained via solid vapor diffusion in DMSO at RT, with the XRPD pattern yed in . XRPD data for sulfate Type B provide (peak shift within i0.2°) primary peaks at 7.0, 9.6, and 20.0; secondary peaks at 18.2, 19.6, and 25.2; and tertiary peaks at 14.0, 24.6, and 28.3.
TGA and DSC s were shown in . A weight loss of 11.7% was observed up to 130 °C and DSC shows two endothermic peaks at 111.2 °C and 202.2 °C (onset temperature) before decomposition, with the first due to desolvation and second attributed to melting. Sulfate Type B converts to anhydrate e Type A after being heated to 120 °C. Also, DMSO content of 11.3% was detected by 1H NMR, which was consistent with weight loss in TGA. Considering all the characterization data, sulfate Type B was calculated as a DMSO solvate. 3.1.2 Hemi-sulfate Type A Hemi-sulfate Type A was obtained in acetone/H20 (aw=0.8) system. Hemi-sulfate Type A sample (807919-34—A) was generated via reactive crystallization in acetone/H20 (aw=0.8) at RT, with a molar charge ratio of 0.5:1 base). The XRPD pattern was shown in and TGA/DSC data were displayed in . XRPD data for ulfate Type A provide (peak shift within i0.2°) primary peaks at 8.5, 11.4, and 12.7; secondary peaks at 6.3, 16.6, and 19.2; and tertiary peaks at 7.6, 15.3, and 23.4.
A weight loss of 5.9% was observed up to 80 °C in TGA and DSC result shows two ermic peaks at 105.3 °C and 219.2 °C (peak temperature) before decomposition, with the first due to dehydration and the second uted to melting. A purity of 99.6 area% was detected via HPLC (Table 3-3). 1H NMR results shows limited acetone detected.
Combined with the stoichiometry of 0.50 base) detected by HPLC/IC, sample (807919- 34-A) was speculated as a hydrate of hemi-sulfate.
Table 3-3 HPLC purity profile of hemi-sulfate Type A (807919A) # RRT Area% # RRT Area% 1 0.75 0.05 3 1.00 99.62 2 0.83 0.33 -- -- -- 3.2 Stability Research for Sulfate Type A 3.2.1 Disproportionation Risk Study A series of slurry experiments were performed at s water activities (0~0.8) to te the disproportionation risk. For s, about 15 mg sulfate Type A sample were weighed to 0.5 mL acetone/H20 systems with aW range from 0 to 0.8. After the sions d at RT for 5 days, the remaining solids were characterized by XRPD. As results shown in Table 3—4 and , no form change was observed when aW lower than 0.6 while hemi- sulfate Type A was generated at aw=0.8, ting the disproportionation risk of sulfate Type A at high relative humidity.
Table 3—4 Summary of slurry experiments results at RT Experiment ID Starting Form Acetone/H20 (v:v) aw* Final Form 807919A17 1000:0 0 Sulfate Type A 807919A18 984:16 0.211 Sulfate Type A 807919A19 Sulfate Type A 948:52 0.406 Sulfate Type A 807919A20 857:143 0.600 e Type A 807919A21 604:396 0.801 Hemi-sulfate Type A *: calculated value 3.2.2 Thermo-stability Study To understand the thermo-stability under elevated temperature, sulfate Type A sample (807919A) was stored at 80 °C for 24 hours and then tested by XRPD and HPLC.
As displayed in Table 3—5 and , no solid form change or HPLC impurity increase was observed, suggesting good physical and chemical stability under the tested condition.
Table 3-5 HPLC purity profile of sulfate Type A (807919A) before and after storage Area (%) Initial 80 °C/24 hrs 0.75 0.11 0.07 0.83 0.64 0.63 1.00 99.14 99.21 1.42 0.11 0.09 3.3 Conclusions A total of three crystal forms were obtained via polymorph screening, including two mono-sulfate (anhydrate Type A/DMSO solvate Type B) and hemi-sulfate Type A.
In addition, the disproportionation risk and thermo-stability were evaluated for sulfate Type A. As results show: 1) sulfate Type A converted to hemi-sulfate Type A at aw=0.8, suggesting the disproportionation risk at high ve humidity, 2) sulfate Type A shows no substantial change in crystal form or HPLC purity, indicating the good thermo- stability after storage at 80 °C for 24 hours. Based on the polymorph screening and evaluation results, sulfate Type A was speculated as the thermodynamically stable form at RT of ulfate. 4. Conclusions Salt screening for resiquimod se was performed under 100 conditions and a total of 32 crystalline hits were isolated. Based on the characterization results, seven salt leads of mono-HCI salt, di-HCI salt, sulfate, ate, e, , and e were ed as leading salts for further evaluation including hygroscopicity, kinetic solubility, and solid-state stability. As evidenced by the results, sulfate with good physicochemical properties was recommended as salt candidate. Using sulfate Type A as starting material, a polymorph screening was performed under 100 conditions and three crystalline forms were observed, including one anhydrate (Type A), one DMSO solvate (Type B), and one hemi- sulfate, suggesting sulfate Type A as a leading form of mono-sulfate. In addition, sulfate Type A shows good physicochemical properties under 80 °C for 24 hours but could convert to hemi-sulfate at high relative humidity. .1 Characterization of Starting Materials .1.1 ng Freebase of Salt Screening The starting se (sample resiquimod, with a CP ID of 807919-05—A) was characterized by XRPD, PLM, TGA, DSC, HPLC, and DVS.
XRPD result in shows the sample (807919A) was crystalline and defined as freebase Type A. PLM image displayed in illustrated aggregation of small particles (< 10 pm). As per TGA and DSC results shown in , a weight loss of 0.3% was observed up to 150 °C in TGA and the DSC curve show a single endothermic peak at 193.3 °C (onset temperature). A purity of 99.1 area% was detected via HPLC (Table 5-1).
DVS plot in shows a water uptake of 0.1% up to 80%RH, suggesting freebase Type A was non-hygroscopic. Also, no form change was observed after DVS evaluation in .
The received freebase Type A (807919-05—A) was used as the starting material for salt screening. Solubility of Type A was estimated in nine solvents at RT. Approximately 2 mg of solids were weighed into each 3-mL glass vial, to which each of the solvents in Table -2 was added in ents of 100 uL until the solids dissolved completely or the total volume reached 1 mL. Solubility ranges of the starting material summarized in Table 5-2 were used to guide the solvent selection for salt screening.
Table 5-1 HPLC purity profile of freebase Type A (807919A) # RRT Area% # RRT Area% 1 0.75 0.05 4 1.00 99.09 2 0.83 0.72 5 1.44 0.10 3 0.87 0.05 -- -- -- Table 5-2 Solubility estimation of freebase Type A (807919A) at RT Solvent Solubility (mg/mL) Solvent Solubility (mg/mL) MeOH S>42.0 DCM 2.1 <S<7.0 EtOH 22.0<S<44.0 EtOAc 2.0<S<6.7 THF 20.0<S<40.0 ane S<2.1 Acetone 7.3<S<22.0 H20 S<2.0 ACN 2.2<S<7.3 -- -- .1.2 Starting Sulfate of Polymorph Screening XRPD comparison in ted sulfate Type A (807919A) was sfully re-prepared on 6-g scale. Detailed procedures were provided in Table 5-3. As per TGA and DSC results shown in , a weight loss of 2.0% was observed up to 100 °C and DSC data show a sharp melting peak at 209.6 °C (onset temperature). Also, a purity of 99.4 area% was detected via HPLC in Table 5-4 and the stoichiometry was determined as 1.11 (acid/base) by HPLC/IC.
The pared sulfate Type A (807919A) was used as the starting material of polymorph screening. The solubility data in Table 5-5 were collected adopting the same procedures as n 5.1.1 and used to guide the solvent ion in polymorph ing design.
Table 5-3 Preparation procedures of sulfate Type A (807919—21-A) ation Procedures 1. Weigh 4 g freebase (807919A) into a 200-mL glass vial and dissolve the solids with 80 mL THF at 50 °C. 2. Measure 1.3 g of sulfuric acid (charge ratio of 1 :1, acid/base) and dilute with 20 mL THF. 3. Add the acid solution to the se solution drop by drop with stirring ically at a speed of 1500 rpm.
.°°.\‘F’\.U‘.'P Add ~ 50 mg seed (807919A) into the system and continue to stir at RT overnight. ng for XRPD and DSC analysis, and both s conform to the reference.
Mix the 4-g batch with the 1-g batch (807919A) prepared previously and stir for1 hr.
Vacuum filter and dry the wet cake at 50 °C for 2 hrs followed by vacuum drying at RT overnight.
Collect 6.4 g solids for analysis (approximate yield of 96.8%).
Table 5-4 HPLC purity profile of sulfate Type A (807919A) # RRT Area% # RRT Area% 1 0.84 0.58 3 1.40 0.06 2 1.00 99.36 -- -- -- Table 5-5 Solubility estimation of sulfate Type A (807919A) at RT Solvent Solubility (mg/mL) Solvent Solubility (mg/mL) MeOH 20.0<S<40.0 2-MeTHF S<2.2 EtOH 7.0<S<21.0 1,4-dioxane S<2.1 IPA S<2.2 Anisole S<2.2 IBA S<2.1 ACN S<2.2 Acetone S<2.0 CHCI3 S<2.0 MEK S<2.0 n-heptane S<2.1 MIBK S<2.1 toluene S<2.0 EtOAc S<2.2 DMAc S>40.0 lPAc S<2.0 DMSO S>44.0 Ethyl lactate S<2.2 NMP S>44.0 MTBE S<2.3 H20 S>44.0 THF S<2.3 DCM S<2.3 .2 Abbreviations for Solvents Used The abbreviations for solvents used are listed in Table 5-6.
WO 32725 Table 5-6 Abbreviations of solvents Abbreviation Solvent Abbreviation Solvent MeOH Methanol THF Tetrahyd rofu ran EtOH Ethanol 2-MeTHF 2-Methyltetrahydrofuran IPA Isopropyl alcohol DCM Dichloromethane IBA lsobutyl alcohol CHCI3 Trichloromethane MEK none ACN Acetonitrile MIBK ylpentanone DMSO Dimethylsulfoxide EtOAc Ethyl acetate DMAc N,N-Dimethylacetamide IPAc Isopropyl acetate NMP ylpyrrolidone MTBE Methyl tert-butyl ether .3 Instruments and Methods .3.1 XRPD For XRPD analysis, a PANalytical Empyrean X—ray powder diffractometer was used. The parameters used are listed in Table 5-7.
Table 5-7 Parameters for XRPD test Parameter Value Cu, kd, K011 (A): 1.540598, K012 (A): 1.544426 X-Ray wavelength Kd2/Kd1 intensity ratio: 0.50 X-Ray tube setting 45 kV, 40 mA Divergence slit Automatic Scan mode Continuous Scan range (°2TH) 3° - 40° Step size (°2TH) 0.013 Scan speed (°/min) About 10 .3.2 TGAIDSC TGA data were collected using a TA Q500/Q5000 TGA from TA Instruments. DSC was performed using a TA Q200/Q2000 DSC from TA Instruments. ed parameters used are listed in Table 5-8.
Table 5-8 Parameters for TGA and DSC test Parameters TGA DSC Method Ramp Ramp Sample pan Platinum, open um, crimped Temperature RT — desired temperature Heating rate 10 °C/min Purge gas N2 .3.3 HPLC Agilent 1100 HPLC was utilized to analyze purity and solubility, with detailed method listed in Table 5-9 and Table 5-10.
Table 5-9 HPLC method for purity test HPLC Agilent 1100 with DAD Detector Column a C18, 150><4.6 mm, 5pm A: 0.1% TFA in H20 Mobile phase B: 0.1% TFA in Acetonitrile Time (min) %B 0.0 10 .0 40 Gradient table 18.0 90 .0 90 .1 10 23.0 10 Run time 23.0 min Post time 0.0 min Flow rate 1.0 mL/min Injection volume 5 pL Detector wavelength UV at 228 nm, reference 500 nm Column temperature 40 °C Samplertemperature RT Diluent Acetonitrile:HZO=1 :1 Table 5-10 HPLC method for solubility test HPLC Agilent 1100 with DAD Detector Column Waters Xbridge C18, .6 mm, 5pm A: 0.1% TFA in H20 Mobile phase B: 0.1% TFA in Acetonitrile HPLC Agilent 1100 with DAD Detector Time (min) %B 0.0 10 .0 90 Gradient table 7.0 90 7.1 10 .0 10 Run time 10.0 min Post time 0.0 min Flow rate 1.0 mL/min Injection volume 10 pL Detector wavelength UV at 228 nm, reference 500 nm Column temperature 40 °C Samplertemperature RT Diluent itrile:HZO=1 :1 .3.4 IC lC method for counter-ion t measurement was listed in Table 5-11 below.
Table 5-11 lC method for counter-ion content measurement Parameters Settings Column lonPac A818 Analytical Column (4 x 250 mm) Mobile Phase 25 mM NaOH ion volume 25 pL Flow rate 1.0 mL/min Cell temperature 35 °C Column temperature 35 °C Current 80 mA 8 mins (CI), 28 mins (P043), 12 mins (SO42'/N03') Run Time 14 mins (C204 ') .3.5 PLM Polarized light microscopic picture was captured on Axio Lab. A1 upright microscope at room temperature. .3.6 DVS DVS was measured via a SMS (Surface Measurement Systems) DVS Intrinsic.
The relative humidity at 25 °C were calibrated against deliquescence point of LiCl, )2 and KCI. Actual parameters for DVS test were listed in Table 5-12.
Table 5-12 Parameters for DVS test Parameters DVS Temperature 25 °C Sample size 10 ~ 20 mg Gas and flow rate N2, 200 mL/min dm/dt 0.002%/min Min. dm/dt ity duration 10 min Max. equilibrium time 180 min RH range 0%RH to 95%RH %RH from 0%RH t0 90%RH RH step size %RH from 90%RH t0 95%RH .3.7 1H NMR 1H NMR spectrum was collected on Bruker 400M NMR Spectrometer using 6 as solvent. .3.8 pKa pKa was determined on Sirius T3 TM according to the manufacturer’s instructions and the parameters for pKa test was listed in Table 5-13.
Table 5-13 Parameters for pKa test pH electrode Ag/AgCl, double junction reference Stirrer Overhead, variable speed, computer controlled Temperature probe Thermocouple, Temperature ed with every datapoint ature l Peltier, Range: 12 °C to 70 °C Turbidity detection Turbidity sensor ion dispensers Water, Acid, Base MultiTip dispenser Multi-tip capillary bundle Home position for electrode storage and pH? buffer positions for Electrode storage/calibration_ _ calibration Washes Two static washes and flowing water wash station WO 32725 Purge gas Two internal flow meters, nitrogen supply requied CoSoIvents Methanol, DMSO and MDM System standardisation Sirius Four-PlusTM procedure pH-range 2.0-12.0 Assay volume 0.5 to 3.5 mls .4 Characterization of Crystalline Hits from Salt Screening .4.1 HCI Salt Type B A total of two HCI salt crystal forms were obtained from screening. HCI salt Type A 9- 07-D1) was obtained via solution crystallization (molar charge of 1:1) in THF and Type B 9C2) was generated via reactive crystallization (molar ratio of 2:1, acid/base) in EtOAc at RT. The XRPD patterns were displayed in . XRPD data for HCI salt Type B show (peak shift within i0.2°) primary peaks at 7.4, 24.3, and 26.2; secondary peaks at 6.7, 15.4, and 20.3; and tertiary peaks at 12.7, 19.1, and 28.5.
For HCI salt Type B, TGA and DSC data () show a two-step weight loss of .8% up to 150 °C and multiple endotherms before decomposition and five erms at 103.5 °C, 110.2 °C, 181.1 °C, 249.8 °C and 266.0 °C (peak temperature) before decomposition. The stoichiometry of 1.73 (acid/base) was determined by HPLC/IC and a purity of 99.2 area% was detected by HPLC in Table 5-15.
Table 5-14 HPLC purity profile of HCI salt Type A (807919D1) # RRT Area% # RRT Area% 1 0.79 0.09 2 1.00 99.91 # RRT Area% # RRT Area% 1 0.79 0.71 3 1.48 0.09 2 1.00 99.21 -- -- -- .4.2 e One e crystal form was generated via screening. Sulfate Type A (807919 A3) was produced via solution crystallization in acetone (molar ratio of 1:1) at RT and its XRPD pattern was shown in . As per TGA and DSC data shown in , a weight loss of 0.4% was viewed up to 130 °C and the DSC curve shows an endothermic peak at 210.1 °C (onset temperature). A purity of 99.3 area% was detected by HPLC in Table 5-16 and the stoichiometry of sulfate Type A (807919A3) was determined as 0.98 (acid/base) by HPLC/IC.
Table 5-16 HPLC purity profile of sulfate Type A (807919-07—A3) # RRT Area% # RRT Area% 1 0.79 0.64 3 1.48 0.08 2 1.00 99.28 -- -- -- .4.3 Phosphate One phosphate crystal form was obtained from screening. Phosphate Type A (807919-07—E5) was obtained via solution llization (molar ratio of 1:1) in MeOH/HZO (9:1, v/v) at RT, and its XRPD pattern was shown in . TGA and DSC curves () show a weight loss of 0.5% up to 150 °C and an endotherm at 254.5 °C (onset temperature) possibly due to melting along with decomposition. Also, a purity of 99.9 area% was detected by HPLC in Table 5-17 and the stoichiometry was determined as 0.92 base) for phosphate Type A (807919-07—E5) via C.
Table 5-17 HPLC purity profile of phosphate Type A (807919E5) # RRT Area% # RRT Area% 1 0.79 0.08 2 1.00 99.92 .4.4 Glycolate One glycolate crystal form was obtained via screening. Glycolate Type A (807919- 07-B9) was generated via reactive crystallization (molar ratio of 1:1) in EtOH at RT. The XRPD pattern was displayed in . XRPD data for glycolate Type A provide (peak shift within i0.2°) primary peaks at 9.3, 11.8, and 22.5; secondary peaks at 14.4, 19.7, and 25.6; and tertiary peaks at 13.1, 18.0, and 21.5.
As per TGA and DSC data in , a weight loss of 0.9% was observed up to 130 °C and the DSC result shows a sharp endothermic peak at 206.0 °C (onset temperature) before decomposition. Also, a purity of 99.7 area% was ed by HPLC in Table 5-18 and the stoichiometry of glycolate Type A (807919-07—B9) was determined as 1.04 (acid/base) by 1H NMR.
Table 5-18 HPLC purity profile of glycolate Type A (807919B9) # RRT Area% # RRT Area% 1 0.79 0.18 3 1.48 0.08 2 1.00 99.74 -- -- -- .4.5 Maleate One maleate crystal form was obtained via screening. Maleate Type A (807919- 07-D4) was generated via reactive llization (molar ratio of 1:1) in THF at RT. The XRPD pattern was yed in .
TGA and DSC results in show a weight loss of 1.4% up to 150 °C and an endothermic peak at 223.8 °C (onset temperature) possibly due to melting. Also, a purity of 99.3 area% was detected by HPLC in Table 5-19 and the stoichiometric ratio was speculated as 0.98 (acid/base) by 1H NMR.
Table 5-19 HPLC purity profile of maleate Type A (807919-07—D4) # RRT Area% # RRT Area% 1 0.79 0.67 3 1.48 0.09 2 1.00 99.25 -- -- -- .4.6 Malate One malate crystal form was obtained via screening. Malate Type A (807919—07— B10) was generated via reactive llization (charge molar ratio of 1:1) in EtOH at RT.
The XRPD pattern was displayed in .
As per TGA and DSC data in , a weight loss of 0.8% was viewed up to 130 °C and DSC result shows a sharp endotherm at 193.3 °C (onset temperature), possibly due to melting. Also, a purity of 99.9 area% was detected by HPLC in Table 5-20 and the stoichiometric ratio was determined as 1.04 freebase) by 1H NMR.
Table 5-20 HPLC purity profile of malate Type A (807919B10) # RRT Area% # RRT Area% 1 0.79 0.07 3 1.48 0.07 2 1.00 99.87 -- -- -- .4.7 Adipate One adipate l form was obtained from screening. e Type A (807919B14) was produced via ve crystallization (molar ratio of 1:1) in EtOH at RT. XRPD pattern and TGA/DSC curves were shown in and respectively.
A weight loss of 1.0% was observed up to 130 °C in TGA and DSC result shows an endothermic peak at 217.7 °C (onset temperature) possibly due to melting. Also, a purity of 100.0 area% was detected by HPLC in Table 5—21 and the stoichiometry of adipate Type A (807919-07—B14) was ated as 0.52 (acid/base) by 1H NMR.
Table 5—21 HPLC purity profile of adipate Type A (807919B14) # RRT Area% # RRT Area% 1 0.79 0.05 2 1.00 99.95 .4.8 Hippurate One hippurate crystal form was obtained via screening. Hippurate Type A (807919—07—B1 1) was generated via reactive crystallization (molar ratio of 1:1) in EtOH at RT.
XRPD n was displayed in . XRPD data for hippurate Type A provide (peak shift within i0.2°) primary peaks at 5.9, 9.5, and 12.1; secondary peaks at 18.9, 21.2, and 25.2; and tertiary peaks at 10.8, 23.3, and 29.2.
TGA and DSC results in show a weight loss of 2.8% before 130 °C and an endothermic peak at 213.9 °C (onset temperature) before decomposition. Also, a purity of 99.6 area% was detected by HPLC in Table 5—22. The stoichiometric ratio was determined as 1.00 (acid/base) by 1H NMR.
Table 5-22 HPLC purity profile of hippurate Type A (807919B11) # RRT Area% # RRT Area% 1 0.79 0.05 3 1.48 0.08 2 1.00 99.61 -- -- -- .4.9 te A total of three te crystal forms were obtained via screening. Tartrate Type A (807919-07—A6), Type B (807919—07—E6), and Type C (807919—07—B6) were generated via reactive crystallization in acetone, MeOH/HZO (9:1, v/v), and EtOH at RT respectively, with a molar charge ratio of 1:1. The XRPD pattern for te Type A is displayed in .
XRPD data for tartrate Type A provide (peak shift within i0.2°) primary peaks at 6.3, 18.2, and 20.9; secondary peaks at 9.1, 19.4, and 25.9; and tertiary peaks at 16.3, 23.1, and 23.6.
WO 32725 The XRPD pattern for Tartrate Type B is displayed in . XRPD data for tartrate Type B provide (peak shift within i0.2°) primary peaks at 8.9, 14.5, and 23.5; secondary peaks at 11.3, 16.9, and 24.2; and tertiary peaks at 9.9, 13.4, and 15.4.
The XRPD pattern for Tartrate Type C is yed in . XRPD data for tartrate Type C provide (peak shift within i0.2°) primary peaks at 7.2, 10.2, and 11.1; ary peaks at 9.3, 13.8, and 18.9; and tertiary peaks at 8.7, 20.4, and 23.5.
For Type A, a weight loss of 2.1% was observed up to 130 °C and DSC data () shows an endothermic peak at 161.4 °C (onset temperature) before decomposition. For Type B, TGA and DSC results in show a weight loss of 3.2% up to 130 °C and two ermic peaks at 144.9 °C and 245.8 °C (peak temperature) before decomposition. For Type C, a weight loss of 2.1% was viewed up to 150 °C in TGA and DSC result () shows an endotherm at 72.2 °C (peak ature) before melting at 240.9 °C (onset temperature) before decomposition.
Also, the stoichiometric ratio was determined as 1.02 (acid/base) for Type A, 0.52 (acid/base) for Type B and C by 1H NMR. .4.10 Fumarate A total of three fumarate crystal forms were obtained via screening. Fumarate Type A (807919—07—A7), Type B (807919—07—E7), and Type C (807919—07—C7) were generated via reactive crystallization (molar ratio of 1:1) in e, MeOH/HZO (9:1, v/v), and EtOAc at RT respectively. The XRPD pattern for fumarate Type A is shown in .
XRPD data for fumarate Type A provide (peak shift within i0.2°) primary peaks at 6.4, 9.9, and 18.4; secondary peaks at 7.9, 23.7, and 26.0; and ry peaks at 7.2, 13.3, and 25.2.
] The XRPD patterns for fumarate Type B are shown in . XRPD data for fumarate Type B provide (peak shift within i0.2°) primary peaks at 10.0, 12.4, and 17.2; secondary peaks at 15.3, 19.1, and 20.3; and tertiary peaks at 17.2, 22.6, and 24.9.
The XRPD patterns for fumarate Type C are shown in . XRPD data for fumarate Type C provide (peak shift within i0.2°) primary peaks at 6.7, 9.1, and 26.6; secondary peaks at 11.2, 15.4, and 20.1; and tertiary peaks at 13.7, 19.4, and 21.1.
For Type A, TGA and DSC data shows a weight loss of 0.8% up to 150 °C and three endothermic peaks at 229.9 °C, 238.0 °C and 252.9 °C (peak temperature) before decomposition (). For Type B, TGA and DSC data shows a weight loss of 4.2% up to 100 °C and four endothermic peaks at 109.6 °C, 226.8 °C, 237.9 °C and 255.9 °C (peak temperature) before decomposition (). For Type C, TGA and DSC data show a weight loss of 0.4% up to 100 °C and four endothermic peaks at 156.3 °C, 237.8 °C and 248.8 °C (peak temperature) before decomposition ().
Also, the stoichiometry was determined as 0.81, 0.61, and 1.03 (acid/base) for Type A ~ 0 by 1H NMR .4.11 Citrate Two citrate crystal forms were obtained via ing. Citrate Type A (807919-07— A8) and Type B (807919B8) were produced via reactive crystallization (molar ratio of 1:1) in acetone and EtOH at RT. The XRPD pattern for citrate Type A is shown in . XRPD data for citrate Type A provide (peak shift within i0.2°) primary peaks at 6.3, 11.5, and 21.3; secondary peaks at 14.9, 17.6, and 19.6; and tertiary peaks at 5.7, 10.0, and 26.3.
The XRPD patterns for citrate Type B are shown in . XRPD data for citrate Type B provide (peak shift within i0.2°) primary peaks at 6.0, 10.0, and 18.2; secondary peaks at 8.2, 12.1, and 21.5; and tertiary peaks at 11.0, 13.4, and 19.2.
For Type A, a weight loss of 0.3% was viewed up to 100 °C and DSC result () shows a sharp endothermic peak at 163.1 °C (onset temperature) before decomposition.
For Type B, TGA and DSC data in shows a weight loss of 2.3% before 100 °C and a sharp endothermic peak at 195.3 °C (onset temperature) before decomposition.
Also, the stoichiometric ratio was determined as 1.02 and 0.53 (acid/base) for Type A and B by 1H NMR in 8 and 9, respectively. .4.12 Lactate ] Two lactate crystal forms were obtained via screening. e Type A (807919- ) and Type B (807919A12) were generated via reactive llization (molar ratio of 1:1) in EtOAc and acetone at RT. The XRPD pattern for lactate Type A is displayed in . XRPD data for lactate Type A provide (peak shift within i0.2°) primary peaks at 5.6, 7.5, and 9.0; secondary peaks at 6.7, 10.1, and 22.3; and tertiary peaks at 8.4, 13.2, and 19.2.
The XRPD patterns for e Type B are shown in . XRPD data for lactate Type B provide (peak shift within i0.2°) primary peaks at 5.8, 7.6, and 9.4; secondary peaks at 6.8, 11.6, and 14.0; and tertiary peaks at 8.5, 18.8, and 25.6.
For Type A, a weight loss of 4.9% was observed up to 100 °C in TGA and DSC result in show endothermic peaks at 85.4 °C, 159.5 °C and 169.4 °C (peak temperature) before decomposition. For Type B, TGA and DSC result in shows a weight loss of 0.6% up to 100 °C and two endothermic peaks at 142.9 °C and 160.2 °C (peak temperature) before decomposition.
] Also, the stoichiometric ratio was determined as 1.07 and 0.96 (acid/base) for lactate Type A and B by 1H NMR .4.13 Succinate Two succinate l forms were obtained via screening. Succinate Type A (807919—07—C13) and Type B (807919-07—E13) were generated via reactive crystallization (molar ratio of 1:1) in EtOAc and MeOH/HZO (9:1, v/v) at RT. The XRPD pattern for succinate Type A is displayed in . XRPD data for succinate Type A provide (peak shift within i0.2°) primary peaks at 6.4, 7.1, and 9.9; secondary peaks at 11.3, 18.4, and 23.1; and tertiary peaks at 5.0, 20.1, and 24.9.
The XRPD pattern for succinate Type B is shown in . XRPD data for succinate Type B provide (peak shift within i0.2°) y peaks at 10.5, 17.5, and 23.9; secondary peaks at 8.7, 12.2, and 14.1; and tertiary peaks at 16.6, 19.7, and 22.3.
] For Type A, TGA and DSC results in show a weight loss of 2.0% up to 130 °C and a sharp endothermic peak at 174.4 °C (onset ature) before decomposition.
For Type B, data (0) show a weight loss of 4.6% up to 100 °C and three endothermic peaks at 103.5 °C, 188.4 °C and 209.6 °C (peak temperature) before decomposition. Also, the stoichiometric ratio was determined as 1.00 and 0.52 (acid/base) by 1H NMR. .4.14 Tosylate Two tosylate crystal forms were obtained via screening. Tosylate Type A (807919- 07-B15) and Type B (807919-07—D15) were generated via reactive crystallization (molar ratio of 1:1) in EtOH and THF at RT. The XRPD pattern for tosylate Type A is displayed in 1. XRPD data for tosylate Type A provide (peak shift within i0.2°) primary peaks at 4.8, 9.3, and 19.2; secondary peaks at 14.9, 16.3, and 19.7; and ry peaks at 20.7, 24.6, and 27.9.
The XRPD pattern for tosylate Type B is shown in 2. XRPD data for tosylate Type B provide (peak shift within i0.2°) y peaks at 7.7, 8.6, and 10.0; secondary peaks at 13.5, 15.5, and 19.9; and tertiary peaks at 17.4, 23.3, and 27.8.
For Type A, TGA and DSC results in 3 show a weight loss of 1.3% up to 130 °C and a sharp endothermic peak at 201.5 °C (onset temperature) before decomposition.
For Type B, data (4) show a weight loss of 5.3% up to 150 °C and four endothermic peaks at 61.1 °C, 185.4 °C, 189.9 °C and 201.9 °C (peak temperature) before osition.
Also, the stoichiometric ratio was determined as 0.89 and 0.92 (acid/base) for Type A and B by 1H NMR.
WO 32725 .4.15 Mesylate One te crystal form was obtained via screening. Mesylate Type A (807919- 07-A16) was generated via reactive crystallization (molar ratio of 1:1) in acetone at RT. The XRPD pattern was displayed in 5. XRPD data for mesylate Type A e (peak shift within i0.2°) primary peaks at 8.6, 12.7, and 25.8; secondary peaks at 14.2, 18.6, and 19.5; and tertiary peaks at 16.4, 17.4, and 21.3.
As per TGA and DSC data in 6, it shows a weight loss of 1.5% up to 130 °C and a sharp endothermic peak at 206.6 °C (onset temperature) before decomposition.
Also, the stoichiometric ratio was determined as 0.93 base) by 1H NMR. .4.16 Oxalate Two oxalate crystal forms were obtained via screening. e Type A (807919- 07-B17) and Type B (807919D17) were produced via reactive crystallization (molar ratio of 1:1) in EtOH and THF at RT respectively. The XRPD pattern for oxalate Type A is yed in 7. XRPD data for oxalate Type A provide (peak shift within i0.2°) primary peaks at 9.2, 19.1, and 23.4; secondary peaks at 14.5, 17.7, and 25.0; and tertiary peaks at 11.5, 22.6, and 30.2.
The XRPD pattern for oxalate Type B is shown in 8. XRPD data for oxalate Type B provide (peak shift within i0.2°) primary peaks at 5.4, 18.0, and 23.1; secondary peaks at 9.9, 10.9, and 27.8; and tertiary peaks at 13.0, 16.9, and 24.2.
For Type A, TGA and DSC data (9) show a weight loss of 0.7% up to 130 °C and a sharp endothermic peak at 227.3 °C (onset temperature) before decomposition.
For Type B, TGA and DSC results (0) show a weight loss of 1.8% up to 130 °C and two endothermic peaks at 190.5 °C and 218.2 °C (peak temperature) before decomposition.Also, the stoichiometric ratio was speculated as 0.92 and 1.02 (acid/base) for Type A and B by HPLC/IC. .4.17 Gentisate ] A total of two gentisate crystal forms were obtained via screening. Gentisate Type A (807919—07—A18) and Type B (807919E18) were ted via reactive crystallization (molar ratio of 1:1) in acetone and MeOH/HZO (9:1, v/v) at RT respectively. The XRPD pattern for genistate Type A is displayed in 1. XRPD data for gentisate Type A provide (peak shift within i0.2°) primary peaks at 6.7, 10.0, and 22.9; secondary peaks at 7.1, 8.4, and 16.4; and tertiary peaks at 14.4, 18.4, and 20.5.
The XRPD pattern for gentisate Type B is shown in 2. XRPD data for gentisate Type B provide (peak shift within i0.2°) primary peaks at 6.3, 10.1, and 24.2; ary peaks at 12.7, 20.6, and 26.2; and tertiary peaks at 11.1, 14.8, and 15.8.
For Type A, TGA and DSC data (3) show a weight loss of 4.9% up to 150 °C and an endothermic peak at 210.4 °C (onset temperature) and an exothermic peak at 145.5 °C (peak temperature) before decomposition. For Type B, TGA and DSC results in 4 show a weight loss of 1.7% up to 150 °C and an endothermic peak at 208.2 °C (onset temperature) and an exothermic peak at 148.5 °C (peak ature) before decomposition. Also, the stoichiometric ratio was determined as 0.99 and 1.00 base) for Type A and B by 1H NMR. .4.18 Benzoate A total of two benzoate crystal forms were obtained from screening. Benzoate Type A 9A19) and Type B (807919-07—E19) were produced via reactive crystallization (molar ratio of 1:1) in acetone and MeOH/HZO (9:1, v/v) at RT. The XRPD pattern for benzoate Type A is shown in 5. XRPD data for benzoate Type A provide (peak shift within i0.2°) y peaks at 7.9, 20.8, and 21.5; ary peaks at 12.0, 15.6, and 23.9; and tertiary peaks at 8.7, 19.9, and 29.4.
The XRPD pattern for benzoate Type B is shown in 6. XRPD data for benzoate Type B provide (peak shift within i0.2°) primary peaks at 7.7, 12.5, and 18.8; secondary peaks at 13.5, 22.6, and 26.7; and tertiary peaks at 19.8, 21.4, and 24.4.
For Type A, TGA and DSC results (7) show a weight loss of 0.6% up to 130 °C and an endothermic peak at 199.0 °C (onset temperature) before decomposition. For Type B, TGA and DSC data in 8 show a weight loss of 4.0% up to 100 °C and two endothermic peaks at 102.2 °C and 199.8 °C (onset temperature) before decomposition.
Also, the stoichiometry was ated as 0.98 and 0.99 (acid/base) for Type A and B by 1H .4.19 Nitrate Two nitrate crystal forms were obtained via screening. Nitrate Type A (807919 D20) and Type B (807919-07—B20) were ted via solution crystallization (molar ratio of 1:1) in THF and EtOH at RT. The XRPD pattern for nitrate Type A is displayed in 9.
XRPD data for nitrate Type A provide (peak shift within i0.2°) primary peaks at 17.2, 20.5, and 21.6; secondary peaks at 9.1, 10.1, and 12.0; and tertiary peaks at 14.5, 16.2, and 25.0.
The XRPD pattern for nitrate Type B is shown in 0. XRPD data for nitrate Type B provide (peak shift within i0.2°) primary peaks at 6.7, 9.1, and 25.5; secondary peaks at 9.7, 12.7, and 15.6; and tertiary peaks at 14.4, 20.1, and 26.8.
For Type A, TGA and DSC results in 1 show a weight loss of 1.6% up to 130 °C and a sharp endothermic peak at 212.5 °C (onset temperature) before decomposition.
For Type B, TGA and DSC results (2) show a weight loss of 0.6% up to 130 °C and a sharp ermic peak at 217.6 °C (onset temperature) before decomposition. Also, the iometric ratio was determined as 1.04 (acid/base) for both Type A and B by HPLC/IC. .5 Experiments of rph Screening for Sulfate .5.1 Anti-solvent Addition A total of 18 anti-solvent addition experiments were carried out. About 15 mg of starting sulfate (807919A) was ved in 0.1-2.5 mL solvent to obtain a clear solution, and the solution was magnetically stirred followed by addition of 0.2 mL anti-solvent per step until precipitate appeared or the total amount of anti-solvent reached 15.0 mL. The obtained precipitate was isolated for XRPD analysis. s in Table 5-23 show that no new form was obtained.
Table 5-23 Summary of anti-solvent on experiments Experiment ID Solvent (v:v) Anti-solvent Solid Form 807919A1 MIBK Sulfate Type A 807919A2 EtOAc Sulfate Type A 807919A3 MeOH 1,4-dioxane Sulfate Type A 807919A4** CHCI3 e Type A A5 Toluene Sulfate Type A 807919A6 n-heptane Sulfate Type A 807919A7 EtOH MEK Sulfate Type A 807919A8 lPAc Sulfate Type A 807919A9 MTBE Sulfate Type A lPA/ACN (1:1) 807919A10* Toluene Sulfate Type A A11 ane Sulfate Type A lPA/DCM (3:2) 807919A12* Toluene Sulfate Type A 807919A13** Ethyl lactate Clear 807919A14 THF Sulfate Type A 807919A15 MIBK Sulfate Type A 807919A16 2-MeTHF Sulfate Type A Experiment ID Solvent (v:v) Anti-solvent Solid Form 807919A17 Acetone Sulfate Type A 807919A18 MTBE Sulfate Type A *: solid was observed after stirring the clear solution from anti-solvent on at 5 °C for 2 days. **: no solid was obtained via stirring the clear solution at 5 °C and then evaporation was .5.2 Solid Vapor ion Solid vapor diffusion experiments were conducted using 13 different solvents.
Approximate 10 mg of starting sulfate (807919—21-A) was weighed into a 3-mL vial, which was placed into a 20—mL vial with 2 mL of volatile solvent. The 20-mL vial was sealed with a cap and kept at RT for 7 days ng solvent vapor to interact with sample. The solids were tested by XRPD and the results summarized in Table 5-24 show that sulfate Type A and B were generated.
Table 5-24 Summary of solid vapor diffusion experiments Experiment ID Solvent Solid Form 807919A1 H20 Sulfate Type A 807919A2 DCM Sulfate Type A A3 EtOH e Type A 807919A4 MeOH Sulfate Type A 807919A5 ACN Sulfate Type A 807919A6 THF Sulfate Type A 807919A7 CHCI3 Sulfate Type A 807919A8 Acetone Sulfate Type A 807919A9 DMF e Type A 807919A10 EtOAc Sulfate Type A 807919A11 1,4-dioxane Sulfate Type A 807919A12 IPA e Type A 807919A13 DMSO Sulfate Type B .5.3 Liquid Vapor Diffusion Ten liquid vapor diffusion ments were conducted. Approximate 15 mg of starting sulfate (807919—21-A) was dissolved in appropriate solvent to obtain a clear solution in a 3-mL vial. This solution was then placed into a 20—mL vial with 3 mL of volatile solvents.
The 20-mL vial was sealed with a cap and kept at RT allowing sufficient time for organic vapor to interact with the solution. The precipitates were isolated for XRPD analysis. After 7 days, solids were isolated for XRPD analysis. The results summarized in Table 5—25 show that only sulfate Type A was obtained.
Table 5—25 Summary of liquid vapor diffusion experiments Experiment ID Solvent (v:v) Anti-solvent Solid Form A1 MEK Sulfate Type A 807919A2 MeOH lPAc Sulfate Type A 807919A3 2-MeTHF Sulfate Type A 807919A4 Ethyl lactate Sulfate Type A 807919A5 1,4-dioxane Sulfate Type A 807919A6 lPA/ACN (1:1) Acetone Sulfate Type A 807919A7 lPA/DCM (3:2) MTBE Sulfate Type A A8 NMP THF Sulfate Type A 807919A9 DMSO EtOAc e Type A 807919A10 DMAc CHCI3 Sulfate Type A .5.4 Slow Evaporation Slow ation experiments were performed under eight ions. Briefly, ~15 mg of starting sulfate (807919A) was dissolved in 0.1-2.5 mL of solvent in a 3-mL glass vial. If not dissolved completely, suspensions were filtered using a nylon membrane (pore size of 0.45 pm) and the filtrates would be used d for the follow-up steps. The visually clear solutions were subjected to evaporation at desired temperature with vials sealed by PARAFILM®. The solids were ed for XRPD is, and the results summarized in Table 5-26 indicated that a mixture of sulfate Type A and hemi-sulfate Type A were generated.
Table 5—26 y of slow evaporation experiments ment ID Solvent (v:v) Temperature Solid Form 807919A1 MeOH Sulfate Type A 807919A2 EtOH Sulfate Type A 807919A3 IPA/ACN (1:1) Sulfate Type A 807919A4 IPA/DCM (3:2) Sulfate Type A Sulfate Type A+Hemi- 807919A5 H20 sulfate Type A 807919A6 DMSO 50 °C Sulfate Type A Experiment ID Solvent (v:v) Temperature Solid Form 807919A7 NMP Sulfate Type A 807919A8 DMAc Sulfate Type A .5.5 Polymer induced Crystallization Polymer induced crystallization experiments were performed with two sets of polymer mixtures in three solvents. Approximate 15 mg of starting sulfate (807919A) was dissolved in appropriate solvent to obtain a clear solution in a 3-mL vial. About 2 mg of polymer mixture was added into 3-mL glass vial. All the samples were subjected to evaporation at RT to induce precipitation. The solids were ed for XRPD analysis.
Results summarized in Table 5-27 show that only e Type A was produced.
Table 5-27 Summary of polymer induced crystallization experiments Experiment ID Solvent (v:v) Polymer Solid Form 807919-28—A1 MeOH Sulfate Type A 807919-28—A2 EtOH Polymer mixture A Sulfate Type A 807919-28—A3 lPA/ACN (1:1) Sulfate Type A 807919-28—A4 MeOH Sulfate Type A 807919-28—A5 EtOH Polymer mixture B e Type A 807919-28—A6 lPA/ACN (1:1) Sulfate Type A polyvinyl acetate (PVAC), ellose (HPMC), methyl cellulose (MC) (mass ratio of 1 :1 :1 :1 :1 :1) Polymer mixture B: prolactone (PCL), polyethylene glycol (PEG), poly(methy| rylate) (PMMA) sodium alginate (SA), and hydroxyethyl cellulose (HEC) (mass ratio of 1 :1 :1 :1 :1). .5.6 Slurry at RT ] Slurry conversion experiments were conducted at RT in different solvent systems.
About15 mg of ng sulfate (807919A) was suspended in 0.5 mL of solvent in a 1.5- mL glass vial. After the sion was stirred magnetically for 5 days at RT, the remaining solids were isolated for XRPD analysis. Results summarized in Table 5-28 indicated that hemi-sulfate Type A was ted besides e Type A.
Table 5-28 Summary of slurry conversion experiments at RT Experiment ID Solvent (v:v) Solid Form 807919A1 IPA Sulfate Type A 807919A2 IBA Sulfate Type A 807919A3 MEK Sulfate Type A WO 32725 Experiment ID Solvent (v:v) Solid Form 807919A4 MIBK Sulfate Type A 807919A5 EtOAc Sulfate Type A 807919A6 lPAc Sulfate Type A 807919A7 Ethyl lactate Sulfate Type A 807919A8 MTBE e Type A 807919A9 THF Sulfate Type A 807919A10 2-MeTHF Sulfate Type A 807919A11 1,4-dioxane Sulfate Type A 807919A12 Anisole Sulfate Type A 807919A13 ACN e Type A 807919A14 DCM Sulfate Type A 807919A15 MeOH/toluene (1 :3) Sulfate Type A 807919A16 EtOH/n-heptane (1:3) Sulfate Type A 807919A17 e Sulfate Type A A18 Acetone/H20 (aW=0.2) Sulfate Type A 807919A19 Acetone/H20 (aW=0.4) Sulfate Type A 807919A20 Acetone/H20 (aW=0.6) Sulfate Type A 807919A21 Acetone/H20 (aW=0.8) Hemi-sulfate Type A .5.7 Slurry at 50 °C Slurry conversion experiments were also ted at 50 °C in different solvent s. About 15 mg of starting sulfate (807919A) was suspended in 0.3 mL of solvent in a 1.5-mL glass vial. After the sion was stirred for about 6 days at 50 °C, the remaining solids were isolated for XRPD analysis. Results summarized in Table 5-29 indicated that only sulfate Type A was obtained.
Table 5-29 Summary of slurry conversion experiments at 50 °C ment ID Solvent (v:v) Solid Form 807919A1 IPA Sulfate Type A 807919A2 IBA Sulfate Type A 807919A3 MEK Sulfate Type A 807919A4 MIBK Sulfate Type A 807919A5 EtOAc Sulfate Type A 807919A6 lPAc Sulfate Type A Experiment ID Solvent (v:v) Solid Form 807919A7 Ethyl lactate Sulfate Type A 807919A8 MTBE Sulfate Type A 807919A9 THF Sulfate Type A 807919A10 2-MeTHF Sulfate Type A 807919A11 oxane Sulfate Type A 807919A12 Anisole Sulfate Type A 807919A13 ACN Sulfate Type A 807919A14 CHCI3 Sulfate Type A 807919A15 MeOH/toluene (1 :3) e Type A 807919A16 EtOH/n-heptane (1:3) Sulfate Type A 807919A17 Acetone Sulfate Type A .5.8 Slow g Slow cooling ments were conducted in seven solvent systems. About 20 mg of starting e (807919—21-A) was suspended in 1.0 mL t in a 3-mL glass vial at RT.
The suspension was then heated to 50 °C, equilibrated for about two hours and filtered to a new vial using a nylon membrane (pore size of 0.45 pm). Filtrates were slowly cooled down to 5 °C at a rate of 0.1 °C/min. Clear solutions were transferred to cooling at -20 °C for 2 days and the final clear solutions were subjected to evaporation at RT. Results summarized in Table 5-30 indicated sulfate Type A and B were generated.
Table 5-30 Summary of slow cooling experiments Experiment ID Solvent (v:v) Solid Form 807919A1* oluene (1:1) Sulfate Type A 807919A2 EtOH/n-heptane (1:1) N/A 807919A3 IPA/ACN (1:1) Sulfate Type A 807919A4 lPA/DCM (3:2) Sulfate Type A 807919A5 DMSO/EtOAc (1:3) e Type A + B A6 NMP/MIBK (1:3) Clear 807919A7 DMAc/MTBE (1:3) Sulfate Type A N/A: limited solid for XRPD analysis. *: solid was obtained at -20 °C.
Example 2 1. Summary Polymorph screening for resiquimod freebase was med and its polymorphism towards identifying a suitable crystal form for further ceutical development was evaluated.
The starting material (Batch No.: 144875-48—9) as received was characterized by X—ray powder diffraction (XRPD), thermo-gravimetric analysis (TGA), and ential scanning calorimetry (DSC). The characterization results indicated the starting material conformed to freebase Type A - an anhydrate.
Using Type A as starting material, a polymorph screening was performed under 100 conditions through methods of anti-solvent addition, ation, slow cooling, slurry conversion, vapor diffusion and polymer-induced crystallization. Based on the XRPD comparison, a total of eight crystal forms were isolated, including one anhydrate (Type A), two metastable forms (Type C and F), four solvates (Type B, D, E, and G), and one acetate/acetic acid co-crystal (sample H). The characterization results were ized in Table 2-1. As the inter-conversion illustration displayed in 4, all metastable forms and solvates converted to Type A after storage at ambient conditions or heating experiments, ting Type A was the thermodynamically stable form at room temperature (RT, 20 1r 3 Type A was ed as a leading form and further evaluated on hygroscopicity and solid-state stability. Hygroscopicity was assessed using dynamic vapor sorption (DVS) at 25 °C, and result ted Type A was non-hygroscopic. Physicochemical stability was investigated under 25 °C/60%RH and 40 °C/75%RH for one week, and 80 °C for 24 hours.
No crystal form change or decrease of HPLC purity was observed, indicating good physical and chemical stability for Type A under tested conditions. 2. Characterization of Crystal Forms Polymorph ing was performed under 100 experimental conditions, with eight crystal forms obtained, including one anhydrate (Type A), two metastable forms (Type C/F), four solvates (Type B/D/E/G), and one acetate/acetic acid stal (sample H). The inter-conversion relationships among these forms were studied via storage and g experiments, with the results illustrated in 4.
Table 2-1 Characterization summary of l forms from ing Crystal Form Crystallization Endotherm Wt Loss Solvent Speculate (Sample ID) Conditions (peak, °C) (%) Residual d Form Type A , Not disclosed 194.1 2.5 N/A Anhydrate (807920A) Crystal Form Crystallization Endotherm Wt Loss Solvent Speculate e ID) Conditions (peak, °C) (%) Residual d Form Type C Solid vapor diffusion Metastabl N/A N/A N/A (807920A11) 1,4-dioxane e form Type F Slow cooling Metastabl N/A N/A N/A (807920A4) MEK e form Type B Evaporation 158.4, 192.9 10.8 Toluene 4.3% (807920A13) IBA/Toluene Type B Evaporation 146.7, 191.9 9.3 EtOAc 5.2% phi (807920A7) EtOAc c forms Solution vapor Type B diffusion 143.7, 194.4 6.5 THF 6.1% (807920A2) THF/H20 Type D Solution vapor DMAc 94.1,193.2 18.4 DMAC 18.3% (807920A9) diffusion DMAc/MTBE solvate Type E Slurry NMP 134.0, 187.4 25.5 NMP 22.9% (807920A3) NMP/MTBE solvate Type G Fast cooling Anisole Anisole 84.7, 193.9 14.6 (807920F) Anisole 12.4% solvate olvent addition Acetate/ Sample H Acetic acid Ethyl lactate 151.6,156.9 12.1 acetic acid (807920A1) 8.2% /n-heptane/Acetic acid co-crystal N/A: no data was collected due to the form transformation to Type A.
N/A*: no data was ble.
Preparation procedures for l forms described below are described in Table 2-1a.
Table 2-3 Preparation procedures of salts Crystal Form ation Procedures 1. Weigh 15.3 mg freebase Type A into 2.0 mL IBA to get a clear solution. 2. Add 15.0 mL toluene as anti-solvent to the system and clear solution was obtained.
Isomorphism Type B 3. Transfer the solution to ng at 5 °C 2 days and clear solution was (807920A2) observed.
Method 1 4. Transferthe solution to ation to dryness at RT.
Isolate the solids for analysis.
Isomorphism Type B 1- Weigh 8.5 mg freebase Type A into 2.0 mL EtOAc to get a clear Crystal Form Preparation Procedures (807920A2) on.
Method 2 Transferthe solution to evaporation at RT Isolate the solids for analysis.
Weigh 60.1 mg freebase Type A into 4.0 mL THF to get a clear solution.
Isomorphism Type B Pippte 1.0 mL freebase solution to a 3-mL glass vial. (807920A2) Seal the vial into the 20-mL glass vial with 3-mL H20 and keep the Method 3 system at RT for 6 days. e the solids for analysis.
Weigh 15.3 mg freebase Type A to 0.1 mL DMAc to get a clear solution in a 3-mL glass vial.
DMAc Solvate Type D Seal the vial into the 20-mL glass vial with 3-mL MTBE and keep the (807920A9) system at RT for 6 days.
Isolate the solids for analysis.
Weigh 30.6 mg freebase Type A to 0.3 mL NMP to get a clear solution at RT.
NMP Solvate Type E Add 0.6 mL MTBE to induce precipitate. (807920A3) er the sample to stirring at 4 °C overnight.
Isolate the solids for analysis.
Crystal Form Preparation Procedures 1. Weigh 59.0 mg freebase Type A to 4.5 mL anisole and stir at 50 °C for 1 hrto get a clear solution.
Anisole Solvate Type G (807920F) Keep the sample at 50 °C for another 30 min and then transferto -20 3. Isolate the solids for analysis after 4 days. 1. Weigh 10.1 mg freebase Type A into a 3-mL glass vial.
Metastable Form Type 2. Seal the vial into the 20-mL glass vial with 2-mL 1,4-dioxane and keep C the system at RT. 0A11) 3. Isolate the solids for analysis after 7 days. 1. Weigh 14.4 mg freebase Type A to 1.0 mL MEK and stir at 50 °C for 2 hrs to get a suspension.
Metastable Form Type 2. FIIterthe suspension and transfer the solution to slow cooling (50 °C to_ _ _ _ °C, 0.1 °C/min). (807920A4) 3. Isolate the solids for analysis after 14 days._ _ 2.1 Anhydrate (Type A) Starting material (Batch No.: 1448759, with a CP ID 807920A) was characterized by XRPD, TGA, DSC, and HPLC. The XRPD result in 4 conformed to the Type A reference (807919A). XRPD ns are displayed in 1 and provide (peak shift within i0.2°) y peaks at 8.9, 12.4, and 17.7; ary peaks at 19.7, 21.5, and 23.4; and tertiary peaks at 16.5, 20.1, and 26.7.
TGA and DSC data for the anhydrate Type A crystal form show (5) a weight loss of 2.5% up to 100 °C and a sharp endothermic peak at 193.0 °C (onset ature) before decomposition. Also, a purity of 99.4 area% was detected by HPLC in Table 2-2. Considering all the results, Type A was deemed as an ate.
Table 2-2 HPLC purity profile of Type A (807920A) # RRT Area% # RRT Area% 1 0.75 0.08 4 1.00 99.41 2 0.83 0.24 5 1.07 0.10 3 0.87 0.05 6 1.61 0.13 2.2 Metastable Form 2.2.1 Type C Type C was ted in 1,4-dioxane system. Type C sample (807920—11-A11) was obtained via solid vapor diffusion in 1,4-dioxane. As XRPD pattern displayed in 6, Type C converted to Type A after dried at ambient ions ght, suggesting metastable form for Type C at ambient conditions. XRPD patterns e (peak shift within i0.2°) primary peaks at 9.6, 18.7, and 19.8; secondary peaks at 12.1, 14.5, and 21.2; and tertiary peaks at 17.4, 20.7, and 28.5. 2.2.2 Type F Type F sample (807920A4) was obtained via slow cooling in MEK and the XRPD n was displayed in 7. XRPD patterns provide (peak shift within i0.2°) primary peaks at 10.4, 16.5, and 21.2; secondary peaks at 8.3, 20.7, and 28.8; and tertiary peaks at 12.5, 17.7, and 24.8.
After storage at ambient conditions for 2 days, Type F converted to Type A, indicating Type F was metastable at ambient ions. 2.3 Solvate 2.3.1 Type B lsomorphism occurred to Type B. It can be prepared in several solvent systems, including IBA/toluene, EtOAc, THF/H20, and etc. XRPD patterns of Type B were displayed in 8. XRPD patterns provide (peak shift within i0.2°) primary peaks at 6.2, 16.3, and 21.4; secondary peaks at 12.3, 22.3, and 24.7; and tertiary peaks at 20.4, 27.0, and 28.4.
Three batches of the isomorphism Type B crystal form were produced. Batch 1: TGA and DSC data (9) show a weight loss of 10.8% up to 165 °C and two endothermic peaks at 158.4 °C and 192.9 °C (peak temperature) before decomposition.
Batch 2: TGA and DSC data (0) show a weight loss of 9.3% up to 150 °C and two ermic peaks at 146.7 °C and 191.9 °C (peak temperature) before decomposition.
Batch 3: TGA and DSC data (1) show a weight loss of 6.5% up to 160 °C and two endothermic peaks at 143.7 °C and 194.4 °C (peak temperature) before osition.
Also, 1H NMR confirm a EtOAc t of 5.2% in Type B sample (807920-08—A7), which suggested EtOAc solvate for Type B (807920A7). Also, 1H NMR confirm a THF content of 6.1% in Type B sample (807920A2), which suggested THF solvate for Type B (807920A2).
Table 2-3 HPLC purity profiles of Type B (807920A13) before and after heating RRT Area (%) Initial Type B Type B heated to 160 °C 0.69 0.09 -- 0.75 0.13 0.09 0.83 0.38 0.29 0.87 0.09 0.06 1.00 99.09 99.51 1.04 0.10 -- 1.07 0.12 0.05 2.3.2 Type D Type D sample (807920A9) was prepared via solution vapor diffusion in DMAc/MTBE. XRPD pattern was shown in 7. XRPD patterns are yed in 2 and provide (peak shift within i0.2°) primary peaks at 8.7, 17.6, and 23.9; secondary peaks at 11.2, 21.2, and 22.8; and tertiary peaks at 9.1, 15.5, and 16.9. TGA and DSC data (3) show a weight loss of 18.4% up to 90 °C and two endothermic peaks at 87.2 °C and 190.2 °C (onset temperature) before decomposition. Also, 1H NMR confirm a DMAc content of 18.3% in Type D sample, indicating DMAc solvate for Type D. 2.3.3 Type E Type E was generated in NMP/MTBE system. Type E sample was obtained via slurry in NMP/MTBE (1:2, v/v) at RT. The XRPD pattern is displayed in 4. XRPD patterns provide (peak shift within i0.2°) y peaks at 8.7, 17.9, and 23.9; ary peaks at 16.9, 21.3, and 22.9; and tertiary peaks at 9.2, 11.2, and 12.5. TGA and DSC data (5) show a weight loss of 25.5% up to 120 °C and two endothermic peaks at 134.0 °C and 187.4 °C (peak temperature) before decomposition. Also, 1H NMR shows NMP content of 22.9% was detected, which was consistent with the TGA weight loss, indicating Type E was a NMP solvate. 2.3.4 Type G ] Type G was generated in anisole system. Type G sample (807920F) was obtained via fast cooling from 50 °C to -20 °C and the XRPD pattern yed in 6.
XRPD ns provide (peak shift within i0.2°) y peaks at 9.7, 13.3, and 19.2; secondary peaks at 8.9, 13.8, 28.0; and tertiary peaks at 12.4, 20.6, and 23.4. TGA and DSC data (7) show a weight loss of 14.6% up to 100 °C and two endothermic peaks at 64.5 °C and 193.0 °C (onset temperature) before decomposition. 1H NMR results indicate anisole of 12.4% was detected, which was consistent with the second weight loss in TGA, suggesting anisole solvate for Type G. 2.4 Salt/Co-crystal (Sample H) Sample H (807920A1) was obtained via anti-solvent addition in ethyl lactate/n- heptane with additional acetic acid (molar ratio 04:1, acid/base) and a mixture of Type A and sample H was generated via anti-solvent addition in ethyl Iactate/n-heptane (with acetic acid content detected in ethyl lactate). The XRPD patterns shown in 8 provide (peak shift within i0.2°) primary peaks at 9.7, 13.3, and 19.2; secondary peaks at 8.9, 13.8, 28.0; and tertiary peaks at 12.4, 20.6, and 23.4. TGA and DSC data (9) show a weight loss of 14.6% up to 100 °C and two endothermic peaks at 64.5 °C and 193.0 °C (onset temperature) before decomposition. Also, the acetic acid content of 0.47:1 (molar ratio, acid/base) was determined by 1H NMR. Combined with the terization data, sample H was speculated as an acetate/acetic acid co-crystal. 3. Evaluation of Leading Type A Since all solvates and metastable forms converted to Type A after storage or heating experiments, anhydrate Type A was the dynamically stable form at RT and selected to be further ted on hygroscopicity and state stability. Results show: 1) Type A was non-hygroscopic as evidenced by the limited water uptake in DVS; 2) Type A had good physicochemical properties under 25 °C/60%RH and 40 °C/75%RH for one week, and 80 °C for 24 hours. 3.1 Hygroscopicity DVS rm plot was collected at 25 °C to investigate the solid form ity as a function of humidity for anhydrate Type A (807919A). Solids were pre-dried at 0%RH to remove the unbounded solvent or water before started. As DVS plot shown in 0, a water uptake of 0.1% was observed up to 80%RH, ting Type A (807919A) was non-hygroscopic. Also, no form change was observed after DVS test (0). 3.2 Solid-state Stability ] Physicochemical stability of Type A (807919A) was evaluated under 25 °C/60%RH and 40 °C/75%RH for one week, and 80 °C (closed) for 24 hours. Stability samples were terized by XRPD and HPLC, with the results summarized in Table 3-1 and 2. No change was observed in HPLC purity or crystal form, suggesting good physical and chemical stability for Type A (807919A) under tested conditions.
Table 3—1 Stability evaluation summary of Type A (807919A) l Form HPLC Purity HPLC Purity Form Condition — (Sample ID) (Initial, area%) Area% % of Initial Change se Type A 99.32 25 °C/60%RH, 1 week 99.15 99.8 No (807919A)* 40 °C/75%RH, 1 week 99.28 100.0 N0 Freebase Type A 99.15 80 °c, 24 hrs 99.23 100.1 No (807919A) *: Data was ted in salt screening section. 4. Conclusion Using compound 001 freebase Type A as the starting material, a total of 100 polymorph screening experiments were set up, and XRPD analysis of the solids revealed that eight crystal forms were obtained. Form fication s show that there were one anhydrate (Type A), two metastable forms (Type C/F), four solvates (Type B/D/E/G), and one acetate/acetic acid co-crystal (sample H). The conversion results show Type B~G all converted to Type A after heating or storage, indicating good physical stability for Type A.
Type A was further evaluated by copicity and solid-state stability. The results show Type A was groscopic and possessed good physicochemical properties under 25 RH and 40 °C/75%RH for one week, and 80 °C for 24 hours. Combined with the characterization results, Type A was recommended for further pharmaceutical development.
. Other .1 Sample Information Starting materials as received were used directly in polymorph screening and evaluation experiments, with the detailed information provided in Table 5-1.
Table 5-1 Detailed ation of starting materials nd Batch No. CP ID l Form 144875-48—9 807920A Type A NA 807919A Type A NA: no information was available. .2 Abbreviation for Solvents Used The solvent abbreviations are listed in Table 5-2.
Table 5-2 Abbreviations of solvents Abbreviation Solvent Abbreviation Solvent MeOH Methanol THF Tetrahydrofuran EtOH Ethanol 2-MeTHF 2-Methyltetrahydrofuran IPA lsopropyl alcohol DCM Dichloromethane lBA lsobutyl alcohol CHCI3 Trichloromethane Abbreviation Solvent Abbreviation Solvent MEK none ACN Acetonitrile MIBK 4-MethyIpentanone DMSO Dimethylsulfoxide EtOAc Ethyl acetate DMAc N,N-Dimethylacetamide IPAc Isopropyl e NMP 1-MethyIpyrrolidone MTBE Methyl tert-butyl ether .3 Instruments and Methods .3.1 XRPD For XRPD analysis, a PANaIyticaI Empyrean X—ray powder diffract meter was used. The XRPD parameters used are listed in Table 5-3.
Table 5-3 Parameters for XRPD test Parameters XRPD (Reflection Mode) Cu, kd, K011 (A): 1.540598, K012 (A): 1.544426 X-Ray wavelength Kd2/Kd1 intensity ratio: 0.50 X-Ray tube setting 45 kV, 40 mA Divergence slit Automatic Scan mode uous Scan range (°2TH) 3°-40° Step size (°2TH) 0.0130 Scan speed (°/min) About 7 .3.2 TGA and DSC TGA data were collected using a TA Q500/Q5000 TGA from TA Instruments. DSC was performed using a TA 2000 DSC from TA Instruments. Detailed parameters used are listed in Table 5-4.
Table 5—4 Parameters for TGA and DSC test ters TGA DSC Method Ramp Ramp Sample pan Platinum, open Aluminum, crimped Temperature RT - desired temperature 25 °C - desired temperature Heating rate 10 °C/min 10 °C/min Purge gas N2 N2 .3.3 HPLC Agilent 1100 HPLC was utilized to e purity, with detailed method was listed in Table 5-5.
Table 0-1 HPLC method for purity test HPLC Agilent 1100 with DAD Detector Column Alltima C18, 150><4.6 mm, 5pm A: 0.1% TFA in H20 Mobile phase B: 0.1% TFA in Acetonitrile Time (min) %B 0.0 10 .0 40 Gradient table 18.0 90 .0 90 .1 10 23.0 10 Run time 23.0 min Post time 0.0 min Flow rate 1.0 mL/min Injection volume 5 uL or wavelength UV at 228 nm, reference 500 nm Column temperature 40 °C Samplertemperature RT Diluent Acetonitrile:HZO=1 :1 .3.4 DVS ] DVS was measured via a SMS (Surface Measurement Systems) DVS Intrinsic.
The relative humidity at 25 °C were calibrated against deliquescence point of LiCl, Mg(N03)2 and KCI. Actual parameters for DVS test were listed in Table 5-6.
Table 5-6 Parameters for DVS test Parameters DVS Temperature 25 °C Sample size 10 ~ 20 mg Gas and flow rate N2, 200 mL/min dm/dt 0.002%/min Min. dm/dt stability duration 10 min Max. equilibrium time 180 min RH range 0%RH to 95%RH %RH from 0%RH t0 90%RH RH step size %RH from 90%RH t0 95%RH .3.5 Solution NMR Solution NMR was collected on Bruker 400M NMR Spectrometer using DMSO-d6. .4 Polymorph Screening ] The lity of ng material (807920A) was estimated at RT.
Approximately 2 mg solids were added into a 3-mL glass vial. Solvents were then added step wise (100 pL per step) into the vials until the solids were dissolved or a total volume of 1 mL was reached. Results summarized in Table 5-7 were used to guide the solvent selection in rph ing.
Polymorph screening experiments were performed using different crystallization or solid transition methods. The methods utilized and crystal forms identified are summarized in Table 5-8.
Table 5-7 Approximate solubility of starting material (807920A) at RT Solvent Solubility (mg/mL) t Solubility (mg/mL) n-heptane S<2.0 2-MeTHF 7.0<S<21.0 H20 S<2.1 Acetone 7.1 <S<25.0 MTBE S<2.2 IPA 8.3<S<25.0 toluene S<2.2 IBA 8.3<S<25.0 Anisole S<2.3 THF 21 .0<S<42.0 MEK 2.0<S<6.7 Ethyl lactate <44.0 lPAc 2.1<S<7.0 CHCI3 22.0<S<44.0 MIBK 2.2<S<7.3 EtOH 23.0<S<46.0 EtOAc 2.3<S<7.7 NMP S>40.0 WO 32725 Solvent Solubility (mg/mL) Solvent Solubility (mg/mL) ACN 2.5<S<8.3 DMSO S>44.0 1,4-dioxane 6.7<S<20.0 MeOH S>46.0 DCM 6.7<S<20.0 DMAc S>48.0 Table 5-8 Summary of polymorph screening experiments Method No. of Experiments Crystal Form Anti-solvent Addition 20 Type A, B, G, sample H Slow Evaporation 10 Type A, B Slow Cooling 10 Type A, B, F, G r-induced Crystallization 6 Type A, B Solid Vapor Diffusion 13 Type A, C Solution Vapor Diffusion 10 Type A, B, D, E Slurry at RT/50 °C 31 Type A, C Total 100 Type A~G, sample H .4.1 Anti-solvent Addition A total of 20 anti-solvent addition experiments were carried out. About 15 mg of starting material (807920—05-A) was dissolved in 0.1-2.3 mL solvent to obtain a clear solution, and the solution was magnetically d followed by addition of 0.2 mL anti-solvent per step till precipitate appeared or the total amount of anti-solvent reached 15.0 mL. The obtained itate was isolated for XRPD analysis. Results in Table 5-9 show that Type B, G, and sample H were generated besides Type A.
Table 5-9 Summary of anti-solvent on experiments Experiment ID Solvent Anti-solvent Solid Form 807920A1 MeOH Type A 807920A2** IPA Clear A3** Acetone Type A 807920A4** THF H20 Type A 807920A5 DMSO Type A 807920A6 DMAc Type A A7 NMP Type A 807920A8 EtOH Type A n-heptane 807920A9 THF Type A+B Experiment ID Solvent olvent Solid Form 807920A10 2-MeTHF Type A+B 807920A11 Ethyl lactate Type A+Sample H A12 CHCI3 Type A+B 807920A13** IBA Type B 807920A14 DCM Toluene Type A 807920A15* 1,4-dioxane Type A A16* MeOH Type A 807920A17** Acetone MTBE Type B 807920A18 CHCI3 Type A 807920A19** EtOH Type G Anisole A20 DCM Clear *: solids were observed after stirring the clear solution from anti-solvent addition at 5 °C for 2 days. **: no solid was obtained via stirring the clear solution at 5 °C and then evaporation was employed. .4.2 Slow Evaporation Slow evaporation experiments were performed under ten conditions. y, ~15 mg of starting material (807920-05—A) was dissolved in 1.0-2.0 mL of solvent in a 3-mL glass vial. If not dissolved completely, suspensions were filtered using a nylon membrane (pore size of 0.45 pm) and the filtrates would be used instead for the -up steps. The visually clear solutions were subjected to evaporation at RT with vials sealed by Parafilm®. The solids were isolated for XRPD analysis, and the results summarized in Table 5-10 indicated that Type A and B were obtained.
Table 5-10 Summary of slow evaporation experiments Experiment ID Solvent (v:v) Solid Form 807920-08—A1 MeOH Type A -08—A2 IPA Type A 807920-08—A3 Acetone Type A 807920-08—A4 DCM Type A 807920-08—A5 THF Type A -08—A6 ACN Type A+B 807920-08—A7 EtOAc Type B 807920-08—A8 EtOH/HZO (1:1) Type A 807920-08—A9 2-MeTHF/n-heptane (1:1) Type B WO 32725 ment ID Solvent (v:v) Solid Form -08—A1 o CHCI3/n-heptane (1:1) Type A+B .4.3 Slow g Slow cooling experiments were conducted in ten solvent systems. About 15 mg of ng material (807920-05—A) was suspended in 1.0 mL of solvent in a 3-mL glass vial at RT. The suspension was then heated to 50 °C, equilibrated for about two hours and filtered using a nylon membrane (pore size of 0.45 pm). Filtrates were slowly cooled down to 5 °C at a rate of 0.1 °C/min. The obtained solids were kept isothermal at 5 °C before isolated for XRPD analysis. Clear solutions were transferred to -20 °C and if it was still clear, they were subjected to evaporation at RT. Results summarized in Table 5—11 indicated Type B, F, and G were generated besides Type A.
Table 5—11 Summary of slow cooling experiments Experiment ID Solvent (v:v) Solid Form 807920A1 ACN Type A 807920A2 EtOAc Type A 807920A3 lPAc Type A 807920A4 MEK Type F 807920A5 MIBK Type A+B 807920A6* Anisole Type G 807920A7 Acetone/H20 (1 :3) Type A 807920A8 EtOH/n-heptane (1:3) Type A 807920A9 THF/toluene (1:3) Type A+B 807920A10 CHCI3/MTBE (1:3) Type B No solids were obtained via slow cooling and all samples were erred to -20 °C. *: limited solid was obtained and system was subjected to evaporation at RT. .4.4 Polymer induced Crystallization Polymer d crystallization ments were performed with two sets of polymer mixtures in seven solvents. Approximate 15 mg of starting material (807920A) was dissolved in appropriate solvent to obtain a clear solution in a 3-mL vial. About 2 mg of polymer mixture was added into 3-mL glass vial. All the samples were subjected to evaporation at RT to induce precipitation. The solids were isolated for XRPD analysis.
Results summarized in Table 5-12 show that Type A and B were produced.
Table 5-12 Summary of polymer induced crystallization experiments Experiment ID Solvent Polymer Solid Form 807920A1 MeOH Type A 807920A2 Acetone Polymer mixture A Type A 807920A3 THF Type A 807920A4 MeOH Type B 807920A5 Acetone Polymer e B Type A 807920A6 THF Type A polyvinyl acetate (PVAC), hypromellose (HPMC), methyl cellulose (MC) (mass ratio of 1 :1 :1 :1 :1 :1) Polymer mixture B: polycaprolactone (PCL), polyethylene glycol (PEG), poly(methyl methacrylate) (PMMA) sodium te (SA), and hydroxyethyl cellulose (HEC) (mass ratio of 1 :1 :1 :1 :1). .4.5 Solid Vapor ion Solid vapor diffusion experiments were conducted using 13 different solvents.
Approximate 10 mg of starting material (807920A) was d into a 3-mL vial, which was placed into a 20-mL vial with 2 mL of volatile solvent. The 20-mL vial was sealed with a cap and kept at RT for 7 days allowing solvent vapor to ct with sample. The solids were tested by XRPD and the results summarized in Table 5-13 show that Type A and C were generated.
Table 5-13 Summary of solid vapor diffusion experiments Experiment ID Solvent Solid Form 807920A1 H20 Type A 807920A2 DCM Type A 807920A3 EtOH Type A 807920A4 MeOH Type A A5 ACN Type A 807920A6 THF Type A 807920A7 CHCI3 Type A 807920A8 Acetone Type A 807920A9 DMF Type A 807920A10 EtOAc Type A 807920A11 1,4-dioxane Type C 807920A12 IPA Type A 807920A13 DMSO Type A .4.6 Liquid Vapor Diffusion Ten liquid vapor diffusion experiments were ted. Approximate 15 mg of starting material (807920A) was dissolved in appropriate solvent to obtain a clear solution in a 3—mL vial. This solution was then placed into a 20-mL vial with 3 mL of volatile ts. The 20-mL vial was sealed with a cap and kept at RT allowing sufficient time for organic vapor to interact with the solution. The precipitates were isolated for XRPD analysis.
After 6 days, solids were isolated for XRPD analysis. The results ized in Table 5-14 show that Type B, D, and E were generated besides Type A.
Table 5-14 Summary of liquid vapor diffusion experiments Experiment ID Solvent Anti-solvent Solid Form 807920A1 EtOH Type A 807920A2 THF Type B 807920A3 CHCI3 Type A+B Toluene 807920A4 2-MeTHF Type A 807920A5 Acetone Type A 807920A6 IBA n-heptane Clear 807920A7 DCM Type A 807920A8 Ethyl lactate Clear 807920A9 DMAc MTBE Type D 807920A10 NMP Type E .4.7 Slurry at RT Slurry conversion ments were ted at RT in different solvent systems.
About 15 mg of starting material (807920A) was suspended in 0.5 mL of solvent in a 1.5- mL glass vial. After the sion was stirred magnetically for 3 days at RT, the remaining solids were ed for XRPD analysis. Results summarized in Table 5-15 ted that only Type A was obtained.
Table 5-15 Summary of slurry conversion experiments at RT Experiment ID Solvent (v:v) Solid Form 807920A1 ACN Type A 807920A2 EtOAc Type A 807920A3 lPAc Type A 807920A4 MEK Type A Experiment ID Solvent (v:v) Solid Form 807920A5 MIBK Type A 807920A6 Anisole Type A 807920A7 2-MeTHF Type A 807920A8 1,4-dioxane Type A 807920A9 IPA Type A 807920A10 IBA Type A 807920A11 MeOH/HZO (1 :3) Type A 807920A12 THF/n-heptane (1:3) Type A 807920A13 DCM/toluene (1:3) Type A 807920A14 acetone/H20 (aw=0.2) Type A 807920A15 acetone/H20 (aW =0.4) Type A 807920A16 acetone/H20 (aW =0.6) Type A 807920A17 acetone/H20 (aW =0.8) Type A .4.8 Slurry at 50 °C Slurry conversion experiments were also conducted at 50 °C in different solvent systems. About 15 mg of ng material 0-05—A) was ded in 0.3 mL of t in a 1.5—mL glass vial. After the suspension was stirred for about 3 days at 50 °C, the remaining solids were isolated for XRPD analysis. Results summarized in Table 5-16 indicated that Type A and C were obtained.
Table 5-16 Summary of slurry conversion ments at 50 °C Experiment ID Solvent (v:v) Solid Form 807920A1 ACN Type A 807920A2 EtOAc Type A 807920A3 IPAc Type A 807920A4 MEK Type A 807920A5 MIBK Type A 807920A6 Anisole Type A 807920A7 2-MeTHF Type A 807920A8 1,4-dioxane Type C A9 IPA Type A 807920A10 IBA Type A 807920A11 MeOH/HZO (1 :5) Type A WO 32725 Experiment ID Solvent (v:v) Solid Form A12 THF/n-heptane (1 :5) Type A 807920A13 CHCIg/toluene (1:5) Type A 807920A14 H20 Type A The preceding disclosures are illustrative embodiments. It should be appreciated by those of skill in the art that the techniques disclosed herein elucidate representative techniques that function well in the practice of the t disclosure. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments that are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
Unless otherwise indicated, all numbers expressing quantities of ingredients, properties such as molecular weight, reaction conditions, and so forth used in the specification and claims are to be understood as being modified in all instances by the term .” Accordingly, unless indicated to the ry, the numerical parameters set forth in the specification and attached claims are approximations that may vary depending upon the desired ties sought to be obtained by the present invention. At the very least, and not as an attempt to limit the application of the doctrine of equivalents to the scope of the claims, each numerical parameter should at least be construed in light of the number of reported significant digits and by applying ordinary rounding techniques. hstanding that the numerical ranges and parameters setting forth the broad scope of the invention are approximations, the cal values set forth in the specific es are reported as precisely as possible. Any numerical value, however, inherently contains certain errors necessarily resulting from the standard deviation found in their respective testing measurements.
The terms “a,” “an,” “the” and similar referents used in the context of describing the invention (especially in the context of the ing claims) are to be construed to cover both the singular and the plural, unless otherwise indicated herein or clearly contradicted by context. Recitation of ranges of values herein is merely intended to serve as a shorthand method of referring individually to each separate value falling within the range. Unless otherwise indicated , each dual value is orated into the specification as if it were individually recited herein. A|| methods described herein can be performed in any suitable order unless otherwise indicated herein or otherwise clearly contradicted by context.
The use of any and all examples, or ary language (e.g., “such as”) provided herein is intended merely to better illuminate the ion and does not pose a limitation on the scope of the invention otherwise claimed. No language in the ication should be construed as indicating any non-claimed element essential to the practice of the invention.
Groupings of alternative ts or embodiments of the invention disclosed herein are not to be construed as limitations. Each group member may be referred to and claimed individually or in any ation with other members of the group or other elements found herein. It is anticipated that one or more members of a group may be included in, or deleted from, a group for s of convenience and/or patentability. When any such inclusion or deletion occurs, the specification is deemed to contain the group as modified thus fulfilling the written description of all Markush groups used in the appended claims.
Certain embodiments of this invention are described herein, including the best mode known to the ors for carrying out the invention. Of course, ions on these described embodiments will become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventor expects skilled artisans to employ such variations as appropriate, and the inventors intend for the ion to be practiced othenNise than specifically described herein. Accordingly, this invention includes all modifications and lents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless othenNise indicated herein or othenNise clearly contradicted by context.
Furthermore, numerous references have been made to s and printed publications hout this specification. Each of the above-cited references and printed publications are individually incorporated herein by reference in their entirety.
Specific ments disclosed herein may be further limited in the claims using consisting of or and consisting essentially of language. When used in the claims, whether as filed or added per amendment, the transition term “consisting of’ excludes any element, step, or ingredient not specified in the claims. The transition term “consisting ially of” limits the scope of a claim to the specified materials or steps and those that do not materially affect the basic and novel characteristic(s). Embodiments of the invention so claimed are inherently or expressly described and enabled herein.
In closing, it is to be understood that the embodiments of the invention disclosed herein are illustrative of the principles of the t invention. Other cations that may be employed are within the scope of the invention. Thus, by way of example, but not of limitation, alternative configurations of the t invention may be ed in accordance WO 32725 with the teachings herein. Accordingly, the present invention is not limited to that precisely as shown and described.
We

Claims (8)

claim:
1. A composition comprising resiquimod in the form of a sulfate salt in crystal form A.
2. The composition of claim 1, n the sulfate salt is a monosulfate salt.
3. The composition of claim 1, wherein the e salt is an anhydrate.
4. The composition of claim 1, wherein l form A is characterized by x-ray powder diffraction spectrum that comprises peaks at about 7 to about 8 degrees 26, about 13.5 to about 14.5 degrees 26, about 19 to about 20 degrees 26, and about 19.5 to about 20.5 degrees 26.
5. The composition of claim 1, wherein the crystal form A is stable at room temperature at least about 2 days.
6. The composition of claim 1, wherein the crystal form A is stable at room temperature at least about 1 week.
7. A dosage form comprising the composition of claim 1.
8. A ition comprising a crystal form of a compound of Formula I: N X
NZ789638A 2017-06-23 Pharmaceutical compositions NZ789638A (en)

Publications (1)

Publication Number Publication Date
NZ789638A true NZ789638A (en) 2022-07-01

Family

ID=

Similar Documents

Publication Publication Date Title
US20230132111A1 (en) Pharmaceutical compositions
RU2754856C2 (en) Crystal forms of triazolopyrimidine compound
JP6946194B2 (en) Solid form of compounds that regulate kinases
TWI642667B (en) Pyridino pyrimidine derivatives, preparation method and medical use thereof
KR102342776B1 (en) (S)-2-((2-((S)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1 Polymorphic and solid forms of ,2-d][1,4]oxazepin-9-yl)amino)propanamide, and processes for their preparation
TW202214589A (en) Shp2 inhibitor and application thereof
BR112015004816B1 (en) Crystal of a compound and medicine
JP6963598B2 (en) Thienopyrimidine compounds, their production methods, pharmaceutical compositions and their applications
CN110267945A (en) The benzazolyl compounds that cyano replaces and its purposes as LSD1 inhibitor
KR20170070140A (en) Method for treating cancer
US10626107B2 (en) Crystal form, salt type of substituted 2-hydro-pyrazole derivative and preparation method therefor
TW202208375A (en) Salt and crystal forms of 4-amino-5-(6-(4-methylpiperazin-1-yl)-1h-benzo[d]imidazol-2-yl)thieno[2,3-b]pyridin-6(7h)-one
JP2016513704A (en) Crystalline forms of tyrosine kinase inhibitors and salts thereof
CN106957315B (en) N- replaces benzenesulfonyl-azaindole oxybenzamide class compound and its prepares the purposes of drug
ES2772682T3 (en) Imidazooxazine crystal, pharmaceutical composition containing said crystal, and method of producing said crystal
US9884856B2 (en) Crystal form of Dabrafenib mesylate and preparation method thereof
JP2017529356A (en) Crystal forms of tyrosine kinase inhibitors and salts thereof
JP2019525960A (en) Succinate forms and compositions of breton-type tyrosine kinase inhibitors
SG190914A1 (en) Quinoline derivative compound, method for preparing same, and pharmaceutical composition containing same
BR112021004546A2 (en) crystalline forms of a quinazole compound and its hydrochloride salts
NZ789638A (en) Pharmaceutical compositions
KR20130130802A (en) Compositions and methods of using crystalline forms of wortmannin analogs
WO2022199699A1 (en) Crystal form of salt of nitrogen-containing fused heterocyclic compound, preparation method therefor, and application thereof
CA3211477A1 (en) Salt and solid forms of a kinase inhibitor
TWI443096B (en) Monohydrate crystalline form of 3-(5-(4-(3-fluoropropyl)-piperazin-1-yl)benzimidazol-2-yl)-1-azaazulen-2-one, preparation method, and pharmaceutical composition thereof