POLYMORPHS OF HYDROCHLORIDE SALT OF 5-(3-(ETHYLSULFONYL)PHENYL)-3,8-DIMETHYL-N-(1-METHYLPIPERIDIN-4-YL)-9H-PYRIDO[2,3-B]INDOLE-7-CARBOXAMIDE AND METHODS OF USE THEREFOR

Information

  • Patent Application
  • 20090270442
  • Publication Number
    20090270442
  • Date Filed
    April 13, 2009
    15 years ago
  • Date Published
    October 29, 2009
    14 years ago
Abstract
Polymorphic forms of the hydrochloride salt of 5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-N-(1-methylpiperidin-4-yl)-9h-pyrido[2,3-b]indole-7-carboxamide (referred to herein as Compound 1) which has the formula:
Description
FIELD OF THE INVENTION

The present invention relates generally to polymorphic forms of the hydrochloric acid salt of 5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-N-(1-methylpiperidin-4-yl)-9H-pyrido[2,3-b]indole-7-carboxamide, (referred to herein as “Compound 1”) and methods for their preparation. The present invention also relates to pharmaceutical compositions, kits and articles of manufacture comprising polymorphs of Compound 1, and methods of their use.


DESCRIPTION OF RELATED ART

Compound 1, which has the formula:







is a kinase inhibitor that is described in U.S. Patent Publication No. 2007-0117816, published May 24, 2007 (see Compound 112) and U.S. Patent Application Nos. 60/912,625 and 60/912,629, filed Apr. 18, 2007 (see Compound 83), which are incorporated herein by reference in their entireties.


Phosphoryl transferases are a large family of enzymes that transfer phosphorous-containing groups from one substrate to another. By the conventions set forth by the Nomenclature Committee of the International Union of Biochemistry and Molecular Biology (IUBMB) enzymes of this type have Enzyme Commission (EC) numbers starting with 2.7 . . . . (See, Bairoch A., The ENZYME database in Nucleic Acids Res. 28:204-305 (2000)). Kinases are a class of enzymes that function in the catalysis of phosphoryl transfer. The protein kinases constitute the largest subfamily of structurally related phosphoryl transferases and are responsible for the control of a wide variety of signal transduction processes within the cell. (See, Hardie, G. and Hanks, S. (1995) The Protein Kinase Facts Book, I and II, Academic Press, San Diego, Calif.). Protein kinases are thought to have evolved from a common ancestral gene due to the conservation of their structure and catalytic function. Almost all kinases contain a similar 250-300 amino acid catalytic domain. The protein kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, histidine, etc.). Protein kinase sequence motifs have been identified that generally correspond to each of these kinase families (See, for example, Hanks, S. K.; Hunter, T., FASEB J. 9:576-596 (1995); Kinghton et al., Science, 253:407-414 (1991); Hiles et al., Cell 70:419-429 (1992); Kunz et al., Cell, 73:585-596 (1993); Garcia-Bustos et al., EMBO J., 13:2352-2361 (1994)). Lipid kinases (e.g. PI3K) constitute a separate group of kinases with structural similarity to protein kinases.


Protein and lipid kinases regulate many different cell processes including, but not limited to, proliferation, growth, differentiation, metabolism, cell cycle events, apoptosis, motility, transcription, translation and other signaling processes, by adding phosphate groups to targets such as proteins or lipids. Phosphorylation events catalyzed by kinases act as molecular on/off switches that can modulate or regulate the biological function of the target protein. Phosphorylation of target proteins occurs in response to a variety of extracellular signals (hormones, neurotransmitters, growth and differentiation factors, etc.), cell cycle events, environmental or nutritional stresses, etc. Protein and lipid kinases can function in signaling pathways to activate or inactivate, or modulate the activity of (either directly or indirectly) the targets. These targets may include, for example, metabolic enzymes, regulatory proteins, receptors, cytoskeletal proteins, ion channels or pumps, or transcription factors. Uncontrolled signaling due to defective control of protein phosphorylation has been implicated in a number of diseases and disease conditions, including, for example, inflammation, cancer, allergy/asthma, diseases and conditions of the immune system, disease and conditions of the central nervous system (CNS), cardiovascular disease, dermatology, and angiogenesis.


Initial interest in protein kinases as pharmacological targets was stimulated by the findings that many viral oncogenes encode structurally modified cellular protein kinases with constitutive enzyme activity. These findings pointed to the potential involvement of oncogene related protein kinases in human proliferatives disorders. Subsequently, deregulated protein kinase activity, resulting from a variety of more subtle mechanisms, has been implicated in the pathophysiology of a number of important human disorders including, for example, cancer, CNS conditions, and immunologically related diseases. The development of selective protein kinase inhibitors that can block the disease pathologies and/or symptoms resulting from aberrant protein kinase activity has therefore generated much interest.


Cancer results from the deregulation of the normal processes that control cell division, differentiation and apoptotic cell death. Protein kinases play a critical role in this regulatory process. A partial non-limiting list of such kinases includes abl, Aurora-A, Aurora-B, Aurora-C, ATK, bcr-abl, Blk, Brk, Btk, c-Kit, c-Met, c-Src, CDK1, CDK2, CDK4, CDK6, cRaf1, CSF1R, CSK, EGFR, ErbB2, ErbB3, ErbB4, ERK, Fak, fes, FGFR1, FGFR2, FGFR3, FGFR4, FGFR5, Fgr, FLK-4, Flt-1, Fps, Frk, Fyn, Hck, IGF-1R, INS-R, Jak, KDR, Lck, Lyn, MEK, p38, PDGFR, PIK, PKC, PYK2, Ros, Tie1, Tie2, Trk, Yes and Zap70. In mammalian biology, such protein kinases comprise mitogen activated protein kinase (MAPK) signaling pathways. MAPK signaling pathways are inappropriately activated by a variety of common disease-associated mechanisms such as mutation of ras genes and deregulation of growth factor receptors (Magnuson et al., Seminars in Cancer Biology 5:247-252 (1994)). Therefore the inhibition of protein kinases is an object of the present invention.


Aurora kinases (Aurora-A, Aurora-B, Aurora-C) are serine/threonine protein kinases that have been implicated in human cancer, such as colon, breast and other solid tumors. Aurora-A (also sometimes referred to as AIK) is believed to be involved in protein phosphorylation events that regulate the cell cycle. Specifically, Aurora-A may play a role in controlling the accurate segregation of chromosomes during mitosis. Misregulation of the cell cycle can lead to cellular proliferation and other abnormalities. In human colon cancer tissue, Aurora-A, Aurora-B and Aurora-C have been found to be overexpressed (See, Bischoff et al., EMBO J., 17:3052-3065 (1998); Schumacher et al., J. Cell Biol. 143:1635-1646 (1998); Kimura et al., J. Biol. Chem., 272:13766-13771 (1997)).


Kinase inhibitors are believed to be useful agents for the prevention, delay of progression, and/or treatment of conditions mediated by kinases.


SUMMARY OF THE INVENTION

The present invention provides novel polymorphic forms of Compound 1 and methods of preparing these polymorphic forms, as well as compositions comprising one or more of the novel polymorphs.


Polymorphic Forms

In one aspect, the invention provides polymorphic forms of Compound 1 having the formula:







Various methods are also provided for making Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O, and Form P. Various methods are also provided for manufacturing pharmaceutical compositions, kits and other articles of manufacture comprising one or more of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P.


Amorphous Form:

In one embodiment, the polymorphic form is an amorphous solid having an X-ray powder diffraction pattern (CuKα) comprising a broad diffraction peak at about 25.5 degrees 2-theta (°2θ). In some variations, the X-ray diffraction pattern is substantially as shown in FIG. 1.


Form A:

In another embodiment, the polymorphic form is a monohydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 10.3 and 20.5 degrees 2-theta (°2θ). In some variations, the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 15.5, 17.0 and 19.9°2θ. In other variations, the X-ray diffraction pattern is substantially as shown in FIG. 2.


In yet another embodiment, the polymorphic form is a monohydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 315° C. to about 330° C. In some variations the endotherm is centered at about 327° C. In further variations, the DSC curve is substantially as shown in FIG. 3.


Form B:

In still another embodiment, the polymorphic form is a dimethylacetamide (DMA) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 13.8, 17.1 and 19.7°2θ. In some variations, the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 16.5, 20.1 and 25.0°2θ. In further variations the X-ray diffraction pattern is substantially as shown in FIG. 7.


In a further embodiment, the polymorphic form is a dimethylacetamide (DMA) solvate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 330° C. to about 340° C. In some variations the endotherm is centered at about 337° C. In other variations the polymorphic form has substantially a DSC curve as shown in FIG. 8.


Form C:

In a still further embodiment, the polymorphic form is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.1, 19.8 and 26.4°2θ. In some variations the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 17.7 and 22.0°2θ. In other variations, the X-ray diffraction pattern is substantially as shown in FIG. 11.


In another embodiment, the polymorphic form is an anhydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 332° C. to about 336° C. In some variations the endotherm is centered at about 335° C. In other variations the polymorphic form has a DSC curve substantially as shown in FIG. 12.


Form D:

In still another embodiment, the polymorphic form is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.8, 17.6, and 20.9°2θ. In some variations the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 5.9 and 25.2°2θ. In other variations the X-ray diffraction pattern is substantially as shown in FIG. 16.


In yet another embodiment, the polymorphic form is an anhydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 245° C. to about 255° C. In some variations, the endotherm is centered at about 251° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 17.


Form E:

In still yet another embodiment, the polymorphic form is a N-methylpyrrolidinone (NMP) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.0, 19.6 and 20.2°2θ. In some variations the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 13.9, 25.1 and 26.2 °2θ. In other variations, the X-ray diffraction pattern is substantially as shown in FIG. 20.


In a further embodiment, the polymorphic form is a N-methylpyrrolidinone (NMP) solvate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 215° C. to about 225° C. In some variations, the endotherm is centered at about 221° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 21.


Form F:

In yet a further embodiment, the polymorphic form is a desolvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.0, 17.2, and 25.9°2θ. In some variations, the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 5.2, 10.3 and 20.2°2θ. In other variations, the X-ray diffraction pattern is substantially as shown in FIG. 24.


In another embodiment, the polymorphic form is a desolvate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 323° C. to about 333° C. In some variations, the endotherm is centered at about 328° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 25.


Form G:

In still another embodiment, the polymorphic form is a dimethylformamide (DMF) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.5, 10.9 and 22.0°2θ. In some variations, the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 16.5, 18.4 and 19.5°2θ. In other variations, the X-ray diffraction pattern is substantially as shown in FIG. 27.


In yet another embodiment, the polymorphic form is a dimethylformamide (DMF) solvate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 334° C. to about 338° C. In some variations, the endotherm is centered at about 336° C. In some variations, the polymorphic form has a DSC curve substantially as shown in FIG. 28.


Form I:

In still yet another embodiment, the polymorphic form is a tetrahydrofuran (THF) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.0, 16.7 and 17.4°2θ. In some variations, the X-ray powder diffraction pattern further comprises significant diffraction peaks at about 19.6, 20.2 and 24.6°2θ. In other variations, the X-ray diffraction pattern is substantially as shown in FIG. 31.


In a further embodiment, the polymorphic form is a tetrahydrofuran (THF) solvate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 320° C. to about 340° C. In some variations the endotherm is centered at about 331° C. In other variations, the polymorphic form has substantially a DSC curve substantially as shown in FIG. 32.


Form J:

In a still further embodiment, the polymorphic form is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 4.9, 17.5 and 20.0°2θ. In some variations the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 9.2, 22.1 and 25.2°2θ. In other variations the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 35.


In another embodiment, the polymorphic form is an anhydrate having a differential scanning calorimetry (DSC) curve comprising a forked endotherm centered from about 320° C. to about 330° C. In some variations the forked endotherm is centered at about 326° C. In other variations, the polymorphic form has substantially a DSC curve substantially as shown in FIG. 36.


Form K:

In still another embodiment, the polymorphic form is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.3, 8.5 and 10.5°2θ. In some variations, the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 13.3, 18.6 and 21.3°2θ. In other variations, the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 39.


In yet another embodiment, the polymorphic form is an anhydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 315° C. to about 330° C. In some variations, the endotherm is centered at about 322° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 40.


Form L:

In a further embodiment, the polymorphic form is a channel hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 10.4 and 20.7°2θ. In some variations, the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 15.5, 16.9 and 24.4°2θ. In other variations, the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 43.


In still a further embodiment, the polymorphic form is a channel hydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 320° C. to about 340° C. In some variations, the endotherm is centered at about 333° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 44.


Form M:

In another embodiment, the polymorphic form is a hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.1, 8.2 and 10.2°2θ. In some variations the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 18.1 and 20.6°2θ. In other variations the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 48.


In still another embodiment, the polymorphic form is a hydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 325° C. to about 335° C. In some variations, the endotherm is centered at about 332° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 49.


Form N:

In yet another embodiment, the polymorphic form is a hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 8.4 and 10.3°2θ. In some variations, the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 18.6, 20.0 and 21.0°2θ. In other variations the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 52.


In a further embodiment, the polymorphic form is a hydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 326° C. to about 336° C. In some variations, the endotherm is centered at about 331° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 53.


Form O:

In a still further embodiment, the polymorphic form is a dehydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 6.3, 12.6 and 25.3°2θ. In some variations the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 10.5 and 21.0°2θ. In other variations, the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 56.


In another embodiment, the polymorphic form is a dehydrate having a differential scanning calorimetry (DSC) curve comprising an endotherm centered from about 320° C. to about 330° C. In some variations, the endotherm is centered at about 327° C. In other variations, the polymorphic form has a DSC curve substantially as shown in FIG. 57.


Form P:

In still another embodiment, the polymorphic form has an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.0, 9.4 and 10.0°2θ. In some variations, the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 17.2 and 25.7°2θ. In other variations, the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 59.


Methods of Making Polymorphic Forms

In another aspect, the invention provides methods of making polymorphic forms of Compound 1 having the formula:







In one embodiment, the polymorphic form is Form A (e.g., a monohydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 10.3 and 20.5°2θ), and the method comprises treating Compound 1 with water. In some variations, the method further comprises dissolving Compound 1 in DMF. In other variations, the method further comprises adding an antisolvent to Compound 1 dissolved in the solvent, wherein the antisolvent is isopropyl acetate.


In another embodiment, the polymorphic form is Form B (e.g., a dimethylacetamide (DMA) solvate having an X-ray powder diffraction pattern comprising significant diffraction peaks at about 13.8, 17.1 and 19.7°2θ), and the method comprises treating Compound 1 with DMA. In some variations, the method further comprises dissolving Compound 1 in DMA.


In a further embodiment, the polymorphic form is Form C (e.g., an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.1, 19.8 and 26.4°2θ), and the method comprises drying Compound 1. In some variations, the method further comprises drying Compound 1 at a temperature above 50° C. In other variations, the method further comprises drying Compound 1 at a temperature above 70° C.


In still a further embodiment, the polymorphic form is Form C (e.g., an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.1, 19.8 and 26.4°2θ), and the method comprises dissolving Compound 1 in an anhydrous solvent.


In another embodiment, the polymorphic form is Form D (e.g., an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.8, 17.6, and 20.9°2θ), and the method comprises treating Compound 1 with DMA. In some variations, the method further comprises dissolving Compound 1 in DMA. In other variations, the method further comprises adding an antisolvent to Compound 1 dissolved in the solvent, wherein the antisolvent is methyl tert-butylether (MTBE).


In still another embodiment, the polymorphic form is Form E (e.g., a N-methyl pyrrolidinone (NMP) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.0, 19.6 and 20.2°2θ), and the method comprises treating Compound 1 with NMP.


In yet another embodiment, the polymorphic form is Form F (e.g., a desolvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.0, 17.2, and 25.9°2θ), and the method comprises treating Compound 1 with DMA or DMF. In some variations, the method further comprises heating Compound 1.


In another embodiment, the polymorphic form is Form G (e.g., a dimethylformamide (DMF) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.5, 10.9 and 22.0°2θ), and the method comprises treating Compound 1 with DMF.


In still another embodiment, the polymorphic form is Form I (e.g., a tetrahydrofuran (THF) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.0, 16.7 and 17.4°2θ), and the method comprises treating Compound 1 with THF.


In another embodiment, the polymorphic form is Form J (e.g., an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 4.9, 17.5 and 20.0°2θ), and the method comprises treating Compound 1 with isopropyl alcohol.


In still another embodiment, the polymorphic form is Form K (e.g., an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.3, 8.5 and 10.5°2θ), and the method comprises treating Compound 1 with THF. In some variations, the method further comprises dissolving Compound 1 in EtOH. In other variations, the method further comprises adding an antisolvent to Compound 1 dissolved in the solvent, wherein the antisolvent is THF.


In yet another embodiment, the polymorphic form is Form L (e.g., a channel hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 10.4 and 20.7°2θ), and the method comprises treating Compound 1 with water. In some variations, the method further comprises dissolving Compound 1 in methanol. In other variations, the method further comprises adding an antisolvent to Compound 1 dissolved in the solvent, wherein the antisolvent is selected from the group consisting of methyl tert-butylether, isopropyl acetate and heptane.


In a further embodiment, the polymorphic form is Form M (e.g., a hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.1, 8.2 and 10.2°2θ), and the method comprises treating Compound 1 with water.


In a still further embodiment, the polymorphic form is Form N (e.g., a hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 8.4 and 10.3°2θ), and the method comprises treating Compound 1 with water.


In another embodiment, the polymorphic form is Form O (e.g., a dehydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 6.3, 12.6 and 25.3°2θ), and the method comprises treating Compound 1 with water. In some variations, the method further comprises heating Compound 1.


Methods by which the above referenced analyses were performed in order to identify these physical characteristics are described in the Examples section below.


Compositions Comprising Compound 1

In a further aspect, the invention provides pharmaceutical compositions comprising Compound 1 of the formula:







wherein at least a portion of Compound 1 is present as a polymorphic form, such as any polymorphic form described throughout this application.


In some embodiments, Compound 1 is present in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and/or Form P. These forms are described in greater detail below. It is noted that other crystalline and amorphous forms of Compound 1 may also be present in the composition.


In one variation, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1 where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P. The composition may optionally be a pharmaceutical composition. The pharmaceutical composition may optionally further include one or more additional components that do not deleteriously affect the use of Compound 1.


Kits and Articles of Manufacture Comprising Compound 1

The invention also provides kits and other articles of manufacture comprising a composition that comprises Compound 1, wherein Compound 1 is present in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P. In one variation, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1 where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P. The composition in the kits and articles of manufacture may optionally be a pharmaceutical composition. The pharmaceutical composition may optionally further include one or more additional components that do not deleteriously affect the use of Compound 1.


In regard to each of the above embodiments including a pharmaceutical composition, the pharmaceutical composition may be formulated in any manner where at least a portion of Compound 1 is present in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P. Optionally, a portion of Compound 1 is present in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P for a period of time subsequent to administration of the pharmaceutical formulation to a subject.


Methods of Using Polymorphic Forms

Methods of using a pharmaceutical composition, kit and other article of manufacture comprising one or more of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P to treat various diseases mediated by a kinase are also provided.


In one embodiment, the present invention relates to a method of inhibiting kinases comprising administering a composition where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1.


In another embodiment, the present invention relates to a method of inhibiting kinases in a subject (e.g., human body) with Compound 1 by administering Compound 1 where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P, when the compound is administered. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1.


In another embodiment, the present invention relates to a method of inhibiting kinases in a subject (e.g., human body) with Compound 1 by administering Compound 1 where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P for a period of time after the compound has been administered to a subject. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1.


In still another embodiment, the present invention provides a method of treating a disease state for which kinases possess activity that contributes to the pathology and/or symptomology of the disease state, comprising administering to a subject (e.g., human body) a composition where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P when administered. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1.


In still another embodiment, the present invention provides a method of treating a disease state for which kinases possess activity that contributes to the pathology and/or symptomology of the disease state, comprising causing a composition to be present in a subject (e.g., human body) where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P, for a period of time after the composition has been administered to a subject. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1.


In another embodiment, a method is provided for preventing, delaying the progression of, and/or treating conditions mediated by kinases, in particular cancer (e.g., squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, small-cell lung cancer, non small-cell lung cancers (e.g., large cell lung cancer, adenocarcinoma and squamous cell carcinoma), bladder cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, glioma, colorectal cancer, genitourinary cancer, gastrointestinal cancer, thyroid cancer, skin cancer and blood cancers (e.g., multiple myeloma, chronic myelogenous leukemia and acute lymphocytic leukemia)); inflammation; inflammatory bowel disease; psoriasis; transplant rejection; amyotrophic lateral sclerosis; corticobasal degeneration; Down syndrome; Huntington's Disease; Parkinson's Disease; postencephelatic parkinsonism; progressive supranuclear palsy; Pick's Disease; Niemann-Pick's Disease; stroke; head trauma; chronic neurodegenerative diseases; Bipolar Disease; affective disorders; depression; schizophrenia; cognitive disorders; hair loss; contraceptive medication; mild Cognitive Impairment; Age-Associated Memory Impairment; Age-Related Cognitive Decline; Cognitive Impairment No Dementia; mild cognitive decline; mild neurocognitive decline; Late-Life Forgetfulness; memory impairment; cognitive impairment; androgenetic alopecia; dementia related diseases (e.g., Frontotemporal dementia Parkinson's Type, Parkinson dementia complex of Guam, HIV dementia, diseases with associated neurofibrillar tangle pathologies, predemented states, vascular dementia, dementia with Lewy bodies, Frontotemporal dementia and dementia pugilistica); Alzheimer's Disease; arthritis; and others.


In each instance where it is stated that Compound 1 may be present in the composition in a form selected from the group consisting of Amorphous Form, Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form 0 and Form P, it is intended for the invention to encompass compositions where only one form is present, where two forms are present (all combinations) and where three, four or more forms are present (all combinations).





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a X-ray powder diffraction (XRPD) spectrum of Amorphous Form of Compound 1.



FIG. 2 is an XRPD pattern of Form A of Compound 1.



FIG. 3 is a differential scanning calorimetry (DSC) curve of Form A of Compound 1.



FIG. 4 is a thermal gravimetric analysis (TGA) curve of Form A of Compound 1.



FIG. 5 is an 1H NMR spectrum of Form A of Compound 1.



FIG. 6 is a moisture sorption curve of Form A of Compound 1.



FIG. 7 is an XRPD pattern of Form B of Compound 1.



FIG. 8 is a DSC curve of Form B of Compound 1.



FIG. 9 is a TGA curve of Form B of Compound 1.



FIG. 10 is an 1H NMR spectrum of Form B of Compound 1.



FIG. 11 is an XRPD pattern of Form C of Compound 1.



FIG. 12 is a DSC curve of Form C of Compound 1.



FIG. 13 is a TGA curve of Form C of Compound 1.



FIG. 14 is a 1H NMR spectrum of Form C of Compound 1.



FIG. 15 is a moisture sorption curve of Compound 1.



FIG. 16 is an XRPD pattern of Form D of Compound 1.



FIG. 17 is a DSC curve of Form D of Compound 1.



FIG. 18 is a TGA curve of Form D of Compound 1.



FIG. 19 is an is a 1H NMR spectrum of Form D of Compound 1.



FIG. 20 is a XRPD pattern of Form E of Compound 1.



FIG. 21 is a DSC curve of Form E of Compound 1.



FIG. 22 is a TGA curve of Form E of Compound 1.



FIG. 23 is a 1H NMR spectrum of Form E of Compound 1.



FIG. 24 is a XRPD pattern of Form F of Compound 1.



FIG. 25 is a DSC curve of Form F of Compound 1.



FIG. 26 is a 1H NMR spectrum of Form F of Compound 1.



FIG. 27 is a XRPD pattern of Form G of Compound 1.



FIG. 28 is a DSC curve of Form G of Compound 1.



FIG. 29 is a TGA curve of Form G of Compound 1.



FIG. 30 is a 1H NMR spectrum of Form G of Compound 1.



FIG. 31 is a XRPD pattern of Form I of Compound 1.



FIG. 32 is a DSC curve Form I of Compound 1.



FIG. 33 is a TGA curve of Form I of Compound 1.



FIG. 34 is a 1H NMR spectrum of Form I of Compound 1.



FIG. 35 is an XRPD pattern of Form J of Compound 1.



FIG. 36 is a DSC curve of Form J of Compound 1.



FIG. 37 is a TGA curve of Form J of Compound 1.



FIG. 38 is a 1H NMR spectrum of Form J of Compound 1.



FIG. 39 is an XRPD pattern of Form K of Compound 1.



FIG. 40 is a DSC curve of Form K of Compound 1.



FIG. 41 is a TGA curve of Form K of Compound 1.



FIG. 42 is a 1H NMR spectrum of Form K of Compound 1.



FIG. 43 is an XRPD pattern of Form L of Compound 1.



FIG. 44 is a DSC curve of Form L of Compound 1.



FIG. 45 is a TGA curve of Form L of Compound 1.



FIG. 46 is a 1H NMR spectrum of Form L of Compound 1.



FIG. 47 is a moisture sorption curve of Form L of Compound 1.



FIG. 48 is an XRPD pattern of Form M of Compound 1.


Form 49 is a DSC curve of Form M of Compound 1.


Form 50 is a TGA curve of Form M of Compound 1.


Form 51 is a 1H NMR spectrum of Form M of Compound 1.



FIG. 52 is a XRPD pattern of Form N of Compound 1.



FIG. 53 is a DSC curve of Form N of Compound 1.



FIG. 54 is a TGA curve of Form N of Compound 1.



FIG. 55 is a 1H NMR spectrum of Form N of Compound 1.



FIG. 56 is an XRPD pattern of Form O of Compound 1.



FIG. 57 is a DSC curve of Form O of Compound 1.



FIG. 58 is a TGA curve of Form O of Compound 1.



FIG. 59 is a XRPD pattern of Form P of Compound 1.



FIG. 60 illustrates the conversion of forms observed from slurry and humidity chamber studies.





DETAILED DESCRIPTION OF THE INVENTION

The present invention provides novel polymorphs of Compound 1, as well as compositions comprising Compound 1, where at least a portion of Compound 1 is present in the composition in a form selected from the group consisting of crystalline forms (e.g., Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P) and an amorphous form (e.g., Amorphous Form).


Also provided are kits and other articles of manufacture with compositions comprising Compound 1 where at least a portion of Compound 1 is present in the composition in a form selected from the group consisting of crystalline forms (e.g., Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P) and an amorphous form (e.g., Amorphous Form).


Various methods are also provided including methods of making each of the disclosed forms; methods for manufacturing pharmaceutical compositions comprising Compound 1 where at least a portion of Compound 1 is present in the composition in a form selected from the group consisting of crystalline forms (i.e., Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P) and an amorphous form; and methods of using compositions comprising Compound 1 where at least a portion of Compound 1 is present in the composition in a form selected from the group consisting of crystalline forms (e.g., Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P) and an amorphous form (e.g., Amorphous Form).


As one will appreciate, depending on how a composition comprising a given compound is produced and then, once produced, how the composition is stored and manipulated, will influence the crystalline content of the composition. Accordingly, it is possible for a composition to comprise no crystalline content or may comprise higher concentrations of crystalline content.


It is further noted that a compound may be present in a given composition in one or more different polymorphic forms, as well as optionally also being present as an amorphous material. This may be the result of (a) physically mixing two or more different polymorphic forms; (b) having two or more different polymorphic forms be generated from crystallization conditions; (c) having all or a portion of a given polymorphic form convert into another polymorphic form; and (d) having all or a portion of a compound in an amorphous state convert into two or more polymorphic forms; as well as for a host of other reasons.


As can be seen, depending on how a composition comprising a compound is prepared, the percentage, by weight, of that compound in a given polymorphic form can vary from 0% to 100%. According to the present invention, compositions are provided where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% or more of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O Form P and Amorphous Form.


Definitions

“Crystalline”, as the term is used herein, refers to a material that contains a specific compound, which may be hydrated and/or solvated, and has sufficient crystalline content to exhibit a discernable diffraction pattern by XRPD or other diffraction techniques. Often, a crystalline material that is obtained by direct crystallization of a compound dissolved in a solution or interconversion of crystals obtained under different crystallization conditions, will have crystals that contain the solvent used in the crystallization, termed a crystalline solvate. Also, the specific solvent system and physical embodiment in which the crystallization is performed, collectively termed crystallization conditions, may result in the crystalline material having physical and chemical properties that are unique to the crystallization conditions, generally due to the orientation of the chemical moieties of the compound with respect to each other within the crystal and/or the predominance of a specific polymorphic form of the compound in the crystalline material.


Depending upon the polymorphic form(s) of the compound that are present in a composition, various amounts of the compound in an amorphous solid state may also be present, either as a side product of the initial crystallization, and/or a product of degradation of the crystals comprising the crystalline material. Thus, crystalline, as the term is used herein, contemplates that the composition may include amorphous content; the presence of the crystalline material among the amorphous material being detectable by, among other methods, the composition having a diffraction pattern with individual, discernable peaks.


The amorphous content of a crystalline material may be increased by grinding or pulverizing the material, which is evidenced by broadening of diffraction and other spectral lines relative to the crystalline material prior to grinding. Sufficient grinding and/or pulverizing may broaden the lines relative to the crystalline material prior to grinding to the extent that the XRPD or other crystal specific spectrum may become undiscernable, making the material substantially amorphous or quasi-amorphous.


Continued grinding would be expected to increase the amorphous content and further broaden the XRPD pattern with the limit of the XRPD pattern being so broadened that it can no longer be discerned above noise. When the XRPD pattern is broadened to the limit of being indiscernible, the material may be considered to no longer be a crystalline material, but instead be wholly amorphous. For material having increased amorphous content and wholly amorphous material, no peaks should be observed that would indicate grinding produces another form.


“Amorphous”, as the term is used herein, refers to a composition comprising a compound that contains too little crystalline content of the compound to yield a diffraction pattern, by XRPD or other diffraction techniques, having individual, discernable peaks. Glassy materials are a type of amorphous material. Glassy materials do not have a true crystal lattice, and technically resemble very viscous non-crystalline liquids. Rather than being true solids, glasses may better be described as quasi-solid amorphous material.


“Broad” or “broadened”, as the term is used herein to describe spectral lines, including XRPD, NMR, IR and Raman spectroscopy lines, is a relative term that relates to the line width of a baseline spectrum. The baseline spectrum is often that of an unmanipulated crystalline form of a specific compound as obtained directly from a given set of physical and chemical conditions, including solvent composition and properties such as temperature and pressure. For example, broadened can be used to describe the spectral lines of a XRPD spectrum of ground or pulverized material comprising a crystalline compound relative to the material prior to grinding. In materials where the constituent molecules, ions or atoms, as solvated or hydrated, are not tumbling rapidly, line broadening is indicative of increased randomness in the orientation of the chemical moieties of the compound, thus indicative of an increased amorphous content. When comparisons are made between crystalline materials obtained via different crystallization conditions, broader spectral lines indicate that the material producing the relatively broader spectral lines has a higher level of amorphous material.


“About” as the term is used herein, refers to an estimate that the actual value falls within 5% of the value cited.


“Forked” as the term is used herein to describe DSC endotherms and exotherms, refers to overlapping endotherms or exotherms having distinguishable peak positions.


Preparation and Characterization of the Polymorphs

A. Preparation of Compound 1


Various methods may be used to synthesize Compound 1. A representative method for synthesizing Compound 1 is provided in Example 1. It is noted, however, that other synthetic routes may also be used to synthesize Compound 1.


B. Preparation of the Polymorphs of Compound 1


General methods for precipitating and crystallizing a compound may be applied to prepare the various polymorphs described herein. These general methods are known to those skilled in the art of synthetic organic chemistry and pharmaceutical formulation, and are described, for example, by J. March, “Advanced Organic Chemistry: Reactions, Mechanisms and Structure,” 4th Ed. (New York: Wiley-Interscience, 1992).


In general, a given polymorph of a compound may be obtained by direct crystallization of the compound or by crystallization of the compound followed by inter-conversion from another polymorphic form or from an amorphous form. Depending on the method by which a compound is crystallized, the resulting composition may contain different amounts of the compound in crystalline form as opposed to as an amorphous material. Also, the resulting composition may contain differing mixtures of different polymorphic forms of the compound.


Compositions comprising a higher percentage of crystalline content (e.g., forming crystals having fewer lattice defects and proportionately less glassy material) are generally prepared when conditions are used that favor slower crystal formation, including those slowing solvent evaporation and those affecting kinetics. Crystallization conditions may be appropriately adjusted to obtain higher quality crystalline material as necessary. Thus, for example, if poor crystals are formed under an initial set of crystallization conditions, the solvent temperature may be reduced and ambient pressure above the solution may be increased relative to the initial set of crystallization conditions in order to slow crystallization.


Precipitation of a compound from solution, often affected by rapid evaporation of solvent, is known to favor the compound forming an amorphous solid as opposed to crystals. A compound in an amorphous state may be produced by rapidly evaporating solvent from a solvated compound, or by grinding, pulverizing or otherwise physically pressurizing or abrading the compound while in a crystalline state.


Compound 1 as prepared by the method described in Example I may be used as the starting material for preparation of other polymorphic forms. The methods for testing the solubility of Compound 1 are described in Example 3, and the solubilities of Compound 1 in various solvents are summarized in Table 16. Good solubility was observed in dioxane, MeOH, DMF, DMA, NMP, AcOH and EtOH. Poor solubility was observed in acetone, MeCN, MTBe, EtOAc, IPAc, IPA, THF, 2-Me-THF, DCM, MEK, cyclohexane, heptane and water.


Methods by which the various polymorphic forms may be prepared are described in the Examples section. Specific methods by which Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P may be prepared are summarized below, including in Tables 17-30, 34, 36a and 36b.


C. Polymorphs of Compound 1


Fifteen crystalline forms and one amorphous solid were identified by conducting a polymorph screen. Described herein are Form A, Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P and Amorphous Form of Compound 1. As described in greater detail below, Forms B, E, G, and I were found to be solvates of DMA, NMP, DMF, and THF respectively. Forms A, L, M, and N were found to be hydrates where Form A was confirmed to be a monohydrate and Form L was found to be a channel hydrate. The remaining forms were found to be either anhydrates (C, F, J, K, O) or likely anhydrates (D, P). Where possible, the results of each test for each different polymorph are provided.


Various tests were performed in order to physically characterize the polymorphs of Compound 1 including X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), solution proton nuclear magnetic resonance (1H-NMR), and moisture sorption and desorption analysis (M S/Des). Detailed experimental conditions for each of the analytical techniques are described in Example 2. The characterization of Forms A, B, C, D, E, F, G, 1, J, K, L, M, N, O, and P and Amorphous Form are described below, as are methods for testing the stabilities of the various forms of Compound 1, and the conditions under which the polymeric forms interconvert are also described below.


1. Form A


Based on the available characterization data, Form A appears to be a monohydrate polymorphic form of Compound 1 that is stable at ambient conditions. Form A was characterized by a variety of techniques, including XRPD, DSC, TGA, 1H-NMR and moisture sorption analysis. Table 36a summarizes some of these results. Preparation and scale-up studies related to Form A are presented in Examples 6-10. For example, Form A could be obtained successfully from a water re-slurry (e.g., a binary solvent system, such as MeCN/water) for approximately 4-5 hours at ambient temperature.


Form A is consistent with a monohydrate based on Karl Fischer (KF) and moisture sorption data (FIG. 6). For example, KF analysis of a sample of Form A showed 3.7% water, consistent with a monohydrate (the theoretical wt % for a monohydrate is 3.2%). KF analysis of another sample of Form A showed 3.1% water before heating and 3.0% water after heating. The moisture sorption curve (FIG. 6) shows the hydrate to be stable from 5 to 90% RH, with a maximum moisture uptake of 4.2 wt % at 90% RH. The experiment did not time out (>4 hours) at any point consistent with the hydrated form being stable during the experiment.


A sample of Form A that was dried for one hour at 80° C. to remove water and XRPD analysis following drying showed a pattern consistent with Form A (Table 38 and Example 12). The equilibration to roughly 1 mole of water under the wide ranges of humidity is consistent with the dehydrated material rapidly reconverting to the hydrated Form A upon exposure to ambient laboratory conditions. Further characterization of Form A by XRPD and KF following heating was performed and further confirmed this assessment. XRPD showed the same pattern before and after heating. Form A was also found to be the isolated form when Forms C, L, or N were slurried in water. See Table 34 and FIG. 60. Solubility measurement results showed similar values for both the DI water and the phosphate buffer slurries after equilibration overnight and 1 week, indicating 3 to 4 mg/mL as shown in Table 37. Form A also did not show a change in form upon exposure to 0 and 95% RH for one week, further consistent with a stable monohydrate form as shown in Table 38.



FIG. 2 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form A. The XRPD pattern confirms that Form A is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 1.









TABLE 1







Characteristic XRPD Peaks (CuKα) of Form A











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
4.6000
19.19426
144
2301


2
5.2024
16.97298
2211
15919


3
8.3967
10.52186
128
1187


4
10.2853
8.59367
938
9850


5
14.9200
5.93296
97
939


6
15.2800
5.79398
192
737


7
15.5306
5.70105
728
4707


8
16.2800
5.44027
242
2573


9
16.8000
5.27303
493
2925


10
17.0400
5.19930
865
6197


11
18.0457
4.91174
84
698


12
19.0254
4.66096
224
1633


13
19.8973
4.45864
484
5068


14
20.4800
4.33308
925
6275


15
20.7600
4.27527
673
4921


16
21.2000
4.18752
100
840


17
21.8213
4.06968
110
707


18
22.4272
3.96108
432
3171


19
23.2097
3.82927
148
1069


20
23.7324
3.74611
274
2115


21
24.3450
3.65321
232
1396


22
24.9029
3.57262
148
1107


23
25.2469
3.52471
167
920


24
25.9630
3.42910
418
3520


25
26.8560
3.31707
383
3177


26
27.2400
3.27117
120
972


27
28.2240
3.15932
76
740


28
30.6000
2.91921
67
411


29
30.8400
2.89703
68
963


30
32.1070
2.78554
99
588


31
32.7664
2.73098
86
1644


32
36.0157
2.49169
92
550


33
36.8551
2.43685
68
1234


34
39.2195
2.29521
203
1876









This unique set of XRPD peak positions or a subset thereof can be used to identify Form A. One such subset comprises peaks at about 5.2, 10.3 and 20.5°2θ. Another subset comprises peaks comprises peaks at about 15.5, 17.0 and 19.9°2θ.



FIG. 3 shows a characteristic DSC thermogram of Form A. An endotherm was observed at approximately 327° C. (peak maximum). FIG. 4 is a TGA thermogram of Form A, showing a weight loss of approximately 2.4% at a temperature below 100° C. The theoretical weight loss for a monohydrate is 3.2%.


Form A was further characterized by solution 1H NMR. The spectrum is reported in FIG. 5. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form A are presented below in the Examples section.


2. Form B


Based on the available characterization data, Form B appears to be a DMF solvate polymorphic form of Compound 1. Form B was characterized by a variety of techniques, including XRPD, DSC, TGA, and 1H-NMR. Table 36b summarizes some of these results.



FIG. 7 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form B. The XRPD pattern confirms that Form B is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 2.









TABLE 2







Characteristic XRPD Peaks (CuKα) of Form B











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.1200
28.29512
36
52


2
4.2012
21.01534
46
346


3
5.2400
16.85127
74
454


4
5.5845
15.81249
363
2252


5
6.9185
12.76628
280
1756


6
8.9600
9.86159
50
280


7
9.1743
9.63172
178
977


8
11.1284
7.94443
140
955


9
11.4724
7.70698
111
599


10
12.4362
7.11177
134
909


11
13.1090
6.74824
155
865


12
13.4800
6.56334
129
844


13
13.7939
6.41468
860
4077


14
14.1435
6.25689
286
2040


15
14.5200
6.09549
88
400


16
15.2800
5.79398
106
692


17
15.4800
5.71957
64
316


18
15.8606
5.58316
136
684


19
16.2000
5.46695
50
170


20
16.4932
5.37042
629
4273


21
16.8400
5.26059
240
0


22
17.1041
5.17996
863
5415


23
18.0000
4.92411
39
160


24
18.2901
4.84665
539
3482


25
19.2400
4.60946
57
282


26
19.6698
4.50970
1013
6807


27
20.1135
4.41120
818
4984


28
20.8577
4.25546
303
1985


29
21.1200
4.20320
110
456


30
21.8030
4.07305
438
2257


31
22.2845
3.98612
420
3169


32
23.2986
3.81486
276
1641


33
23.7908
3.73704
32
104


34
24.2800
3.66284
260
1229


35
24.4800
3.63337
238
1274


36
24.9602
3.56454
558
3435


37
25.4395
3.49846
68
339


38
25.8000
3.45039
120
892


39
26.3343
3.38158
631
3405


40
26.6800
3.33855
77
641


41
27.0381
3.29514
258
1441


42
27.6966
3.21827
83
418


43
28.3728
3.14309
174
788


44
29.1200
3.06412
60
240


45
29.5200
3.02350
199
1568


46
29.8400
2.99180
113
672


47
30.3757
2.94025
61
277









This unique set of XRPD peak positions or a subset thereof can be used to identify Form B. One such subset comprises peaks at about 13.8, 17.1 and 19.7°2θ. Another subset comprises peaks at about 16.5, 20.1 and 25.0°2θ.



FIG. 8 shows a characteristic DSC thermogram of Form B, showing multiple events, with an endotherm observed near the temperature range observed for bound weight loss by TGA and followed by an exothermic event consistent with re-crystallization to an anhydrous form. The first endotherm is centered at about 211° C.; the second endotherm is forked having peaks at about 331° C. and at about 338° C. The exotherm is centered at about 245° C.



FIG. 9 is a TGA thermogram of Form B.


Form B was further characterized by solution 1H NMR. The spectrum is reported in FIG. 10. The spectrum is consistent with one molar equivalent of solvent present, as well as the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form B are presented below in the Examples section.


3. Form C


Based on the available characterization data, Form C appears to be an anhydrous polymeric form of Compound 1 that is stable under ambient non-aqueous conditions. Form C can be prepared by slurrying Form A in anhydrous MeCN and MeOH.


Under humid conditions, Form C can be converted to Form A. For example, Form C can be converted to Form A after equilibrating at 95% RH (% relative humidity) for one week at ambient temperature (FIG. 60).


Form C is consistent with an anhydrate based on KF and moisture sorption data showing 1.4% water where 3.2% is theoretical for a monohydrate. The moisture sorption curve showed Form C to be slightly hygroscopic, with a maximum water uptake of 1.9% at 90% RH. The experiment did not time out (>4 hours) at any point and hysteresis was not observed upon desorption. After drying the material at 80° C. for one hour to remove water the sample was analyzed by XRPD and showed a pattern consistent to the starting form. Form C was found to be a stable anhydrate form in non-aqueous environments based on the results of slurry studies presented in Table 34. Form C converted to the monohydrate Form A in water slurries as well as in acetonitrile/water slurries at different ratios (Tables 34 and 35). The humidity chamber study showed that Form C converted to Form A at 95% RH after one week (Table 38).


Form C was characterized by several techniques including XRPD, DSC, TGA, 1H-NMR and moisture sorption analysis. Table 36a summarizes some of these results. FIG. 11 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form C. The XRPD pattern confirms that Form C is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 3.









TABLE 3







Characteristic XRPD Peaks (CuKα) of Form C











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.3217
26.57743
50
259


2
3.5296
25.01242
36
152


3
4.3562
20.26793
35
216


4
4.5614
19.35660
41
189


5
5.3966
16.36262
36
204


6
6.0000
14.71837
41
364


7
6.3449
13.91905
117
890


8
10.5188
8.40343
183
1392


9
11.0558
7.99643
182
1275


10
11.4000
7.75576
144
831


11
12.6166
7.01048
162
1414


12
16.0235
5.52677
42
397


13
16.6400
5.32337
124
1064


14
17.1193
5.17539
1147
9431


15
17.5200
5.05792
294
0


16
17.7200
5.00128
258
2227


17
18.6642
4.75034
138
1167


18
19.4400
4.56248
88
492


19
19.7870
4.48325
1108
7932


20
20.4216
4.34534
36
451


21
21.0406
4.21888
148
911


22
21.9798
4.04069
199
2076


23
22.3600
3.97283
114
0


24
22.7737
3.90159
175
1653


25
23.1517
3.83874
113
761


26
23.5362
3.77689
158
1131


27
24.4723
3.63449
116
692


28
25.2931
3.51838
140
822


29
25.9600
3.42949
42
261


30
26.3864
3.37503
309
1819


31
26.7600
3.32875
92
637


32
27.0579
3.29277
58
308


33
28.1746
3.16475
60
500


34
28.5565
3.12329
119
977


35
30.4944
2.92907
50
421


36
31.4279
2.84417
127
997


37
31.7600
2.81518
39
247


38
36.8369
2.43801
40
436


39
40.1507
2.24410
47
863









This unique set of XRPD peak positions or a subset thereof can be used to identify Form C. One such subset comprises peaks at about 17.1, 19.8 and 26.4°2θ. Another subset comprises peaks at about 17.7 and 22.0°2θ.



FIG. 12 shows a characteristic DSC thermogram of Form C. An endotherm which onsets at about 314° C. and centered from about 332° C. to about 336° C. and the peak maximum was observed at approximately 335° C.



FIG. 13 is a TGA thermogram of Form C. TGA analysis showed no weight loss or only small weight losses likely due to residual solvents.


Form C was further characterized by solution 1H NMR. The spectrum is reported in FIG. 14. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1. A moisture sorption/desorption analysis is shown in FIG. 15.


Further details related to the preparation and characterization of Form C are presented below in the Examples section.


4. Form D


Based on the available characterization data, Form D appears to be an anhydrous polymorphic form of Compound 1. Form D was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36a summarizes some of these results.



FIG. 16 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form D. The XRPD pattern confirms that Form D is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 4.









TABLE 4







Characteristic XRPD Peaks (CuKα) of Form D











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
5.8720
15.03892
49
1203


2
7.7510
11.39688
67
1686


3
8.8400
9.99519
27
729


4
10.7800
8.20039
7
70


5
12.2700
7.20772
30
753


6
14.4000
6.14602
7
102


7
17.5948
5.03658
143
6341


8
20.8766
4.25165
78
2675


9
23.3666
3.80392
43
994


10
25.1866
3.53301
43
1180


11
26.8200
3.32144
8
145


12
29.2000
3.05590
8
120


13
30.8200
2.89887
7
177


14
37.5200
2.39518
8
177









This unique set of XRPD peak positions or a subset thereof can be used to identify Form D. One such subset comprises peaks at about 7.8, 17.6, and 20.9°2θ. Another subset comprises peaks at about 5.9 and 25.2°2θ.



FIG. 17 shows a characteristic DSC thermogram of Form D. An endothermic event centered from about 245° C. to about 255° C. with peak maximum at about 249° C. was observed. An exothermic event which was centered at about 264° C. was also observed.



FIG. 18 is a TGA thermogram of Form D. TGA analysis showed no weight loss or only small weight losses likely due to residual solvents.


Form D was further characterized by solution 1H NMR. The spectrum is reported in FIG. 19. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form D are presented below in the Examples section.


5. Form E


Based on the available characterization data, Form E appears to be a NMP solvate polymorphic form of Compound 1. Form E was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36b summarizes some of these results.



FIG. 20 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form E. The XRPD pattern confirms that Form E is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 5.









TABLE 5







Characteristic XRPD Peaks (CuKα) of Form E











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.2796
26.91851
43
161


2
3.4796
25.37172
34
179


3
4.9246
17.92979
44
275


4
5.4866
16.09441
168
1284


5
6.9449
12.71781
203
1689


6
8.8400
9.99519
56
343


7
9.1607
9.64598
228
1460


8
10.9822
8.04986
70
468


9
11.5489
7.65610
66
425


10
12.5213
7.06363
110
629


11
13.1242
6.74046
84
491


12
13.4000
6.60234
65
453


13
13.8611
6.38373
648
4350


14
14.1600
6.24964
172
0


15
14.5200
6.09549
78
1225


16
15.3611
5.76357
96
732


17
15.8393
5.59062
130
755


18
16.4050
5.39909
520
3202


19
17.0185
5.20582
1073
6834


20
17.9600
4.93498
50
188


21
18.2600
4.85458
350
3028


22
19.5778
4.53068
976
5631


23
19.8000
4.48034
236
1186


24
20.2445
4.38295
755
4547


25
20.8611
4.25478
253
1612


26
21.1600
4.19535
119
669


27
21.7900
4.07545
401
2126


28
22.0000
4.03702
295
1689


29
22.4800
3.95190
142
1162


30
23.4391
3.79231
236
1552


31
23.8134
3.73355
66
518


32
24.3019
3.65959
422
2213


33
24.7600
3.59291
52
240


34
25.1167
3.54269
509
3622


35
25.6577
3.46920
108
622


36
26.2135
3.39689
597
3619


37
26.5200
3.35833
84
630


38
26.9200
3.30933
41
186


39
27.1968
3.27627
211
1123


40
27.6243
3.22653
41
156


41
27.9202
3.19300
48
256


42
28.5154
3.12770
160
893


43
29.2544
3.05035
187
1398


44
29.7186
3.00375
147
960


45
30.0400
2.97234
54
308


46
30.8020
2.90052
263
1475


47
31.1200
2.87160
50
348









This unique set of XRPD peak positions or a subset thereof can be used to identify Form E. One such subset comprises peaks at about 17.0, 19.6 and 20.2°2θ. Another subset comprises peaks at about 13.9, 25.1 and 26.2°2θ.



FIG. 21 shows a characteristic DSC thermogram of Form E, showing multiple events, with an endotherm observed near the temperature range observed for bound weight loss by TGA and followed by an exothermic event consistent with re-crystallization to an anhydrous form. The first endothermic event was centered at approximately 220° C. (peak maximum). The second endothermic event onset at about 318° C. and was centered at 336°. The exothermic event was centered at about 228° C. (peak maximum).



FIG. 22 is a TGA thermogram of Form E.


Form E was further characterized by solution 1H NMR. The spectrum is reported in FIG. 23. The spectrum is consistent with one molar equivalent of solvent present, as well as the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form E are presented below in the Examples section.


6. Form F


Based on the available characterization data, Form F appears to be a desolvate polymorphic form of Compound 1. Form F can be observed after de-solvating Forms B or G by heating them in a TGA instrument to 230-250° C. Form F was characterized by techniques including XRPD, DSC, and 1H-NMR. Table 36a summarizes some of these results.



FIG. 24 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form F. The XRPD pattern confirms that Form F is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 6.









TABLE 6







Characteristic XRPD Peaks (CuKα) of Form F











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.2450
27.20545
41
158


2
4.2764
20.64596
56
359


3
5.1988
16.98472
234
2476


4
5.5200
15.99711
130
596


5
6.4800
13.62916
61
129


6
7.0130
12.59447
755
5647


7
7.3200
12.06693
106
630


8
8.6057
10.26679
48
322


9
9.6000
9.20554
47
367


10
9.9600
8.87361
133
1450


11
10.3084
8.57447
347
2173


12
13.3040
6.64977
24
345


13
14.4843
6.11043
156
979


14
15.1888
5.82856
39
297


15
15.8400
5.59038
23
119


16
16.2000
5.46695
110
1185


17
16.8400
5.26059
276
2913


18
17.2400
5.13943
441
3111


19
17.7200
5.00128
84
833


20
18.8800
4.69653
40
400


21
19.4883
4.55129
153
1491


22
20.1886
4.39496
269
2680


23
20.7200
4.28343
218
2060


24
21.0000
4.22695
230
1402


25
21.6475
4.10196
114
1120


26
22.5920
3.93256
72
634


27
23.1216
3.84367
166
1263


28
23.8612
3.72617
44
315


29
24.3634
3.65049
80
566


30
25.2400
3.52566
255
1302


31
25.4400
3.49839
191
1576


32
25.9281
3.43363
370
2674


33
26.5600
3.35336
161
1517


34
26.7600
3.32875
140
0


35
27.0800
3.29013
77
1043


36
27.4450
3.24720
40
168


37
27.6339
3.22543
33
109


38
28.6027
3.11835
27
102


39
29.4300
3.03254
41
291


40
30.0671
2.96972
44
333


41
31.1600
2.86801
28
139


42
31.4666
2.84076
31
242


43
31.8143
2.81050
28
200


44
35.4395
2.53087
24
151


45
37.2440
2.41229
40
506


46
39.3022
2.29057
32
222


47
42.4711
2.12671
30
174









This unique set of XRPD peak positions or a subset thereof can be used to identify Form F. One such subset comprises peaks at about 7.0, 17.2, and 25.9°2θ. Another subset comprises peaks at about 5.2, 10.3 and 20.2°2θ.



FIG. 25 shows a characteristic DSC thermogram of Form F. An endotherm was observed to onset at about 304° C. and centered from about 323° C. to about 333° C.; peak maximum is at about 328° C.


Form F was further characterized by solution 1H NMR. The spectrum is reported in FIG. 26. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


7. Form G


Based on the available characterization data, Form G appears to be an DMF solvate polymorphic form of Compound 1. Form G was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36b summarizes some of these results.



FIG. 27 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form G. The XRPD pattern confirms that Form G is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 7.









TABLE 7







Characteristic XRPD Peaks (CuKα) of Form G











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
5.4847
16.09999
2359
10474


2
6.9827
12.64905
710
3038


3
9.2290
9.57475
309
1561


4
10.9344
8.08494
2101
8785


5
13.4725
6.56697
321
1670


6
13.9654
6.33629
669
2821


7
15.4400
5.73430
257
1234


8
15.6800
5.64706
280
1375


9
16.4628
5.38027
1566
9294


10
17.0020
5.21083
1161
5256


11
18.4415
4.80720
1105
6367


12
19.5223
4.54344
1678
7240


13
20.6429
4.29926
625
2760


14
20.9709
4.23275
460
2591


15
21.9517
4.04579
6414
24463


16
24.1289
3.68544
315
3198


17
24.5621
3.62141
466
1782


18
25.1330
3.54043
205
1452


19
25.6216
3.47401
228
1195


20
26.3248
3.38278
1026
4893


21
27.1244
3.28485
310
1289


22
29.0166
3.07480
381
2124


23
31.1516
2.86876
518
2161


24
34.4828
2.59887
216
1340









This unique set of XRPD peak positions or a subset thereof can be used to identify Form G. One such subset comprises peaks at about 5.5, 10.9 and 22.0 degrees °2θ. Another subset comprises peaks at about 16.5, 18.4 and 19.5°2θ.



FIG. 28 shows a characteristic DSC thermogram of Form G. The thermogram shows a broad endotherm centered at about 201° C. and a second endotherm which onset at approximately 314° C. and centered from about 334° C. to about 338° C. This second endotherm peaked at approximately 336° C. (peak maximum).



FIG. 29 is a TGA thermogram of Form G.


Form G was further characterized by solution 1H NMR. The spectrum is reported in FIG. 30. The spectrum is consistent with one molar equivalent of solvent present, as well as the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form G are presented below in the Examples section.


8. Form I


Based on the available characterization data, Form I appears to be a THF solvate polymorphic form of Compound 1. Form I was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36b summarizes some of these results.



FIG. 31 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form I. The XRPD pattern confirms that Form I is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 8.









TABLE 8







Characteristic XRPD Peaks (CuKα) of Form I











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.8726
22.79778
37
165


2
4.0783
21.64836
38
96


3
4.2951
20.55611
27
131


4
4.7018
18.77890
41
313


5
5.3200
16.59804
49
269


6
5.6645
15.58934
216
1319


7
6.3279
13.95641
33
96


8
6.5600
13.46313
67
326


9
6.9855
12.64399
564
3563


10
7.5900
11.63827
23
88


11
7.7960
11.33120
18
86


12
9.0400
9.77450
23
45


13
9.2887
9.51335
90
643


14
11.3004
7.82389
77
465


15
12.0000
7.36928
17
47


16
12.2694
7.20808
79
478


17
12.8880
6.86346
50
267


18
13.6000
6.50569
35
323


19
13.9480
6.34415
281
1624


20
14.2400
6.21471
45
314


21
15.2400
5.80910
49
579


22
15.4800
5.71957
78
0


23
15.6400
5.66141
92
670


24
15.8800
5.57639
33
156


25
16.4400
5.38768
136
563


26
16.7316
5.29443
345
2743


27
17.3561
5.10531
481
3013


28
17.7200
5.00128
18
88


29
18.5670
4.77499
273
1927


30
18.8800
4.69653
34
219


31
19.5848
4.52908
261
2408


32
20.1598
4.40118
260
2026


33
21.0841
4.21028
140
1329


34
21.3600
4.15651
55
266


35
21.7815
4.07702
121
674


36
22.2000
4.00110
38
167


37
22.7480
3.90594
135
1389


38
23.5760
3.77060
35
392


39
24.2800
3.66284
43
269


40
24.5571
3.62214
196
1801


41
25.1200
3.54223
26
141


42
25.8800
3.43991
64
778


43
26.1600
3.40372
107
0


44
26.4000
3.37332
158
1391


45
26.7600
3.32875
18
106


46
27.2628
3.26849
21
171


47
28.1166
3.17115
22
158









This unique set of XRPD peak positions or a subset thereof can be used to identify Form I. One such subset comprises peaks at about 7.0, 16.7 and 17.4°2θ. Another subset comprises peaks at about 19.6, 20.2 and 24.6°2θ.



FIG. 32 shows a characteristic DSC thermogram of Form I, showing multiple events, with an endotherm observed near the temperature range observed for bound weight loss by TGA and followed by an exothermic event consistent with re-crystallization to an anhydrous form. The first endothermic event was centered at about 206° C. The exothermic event was centered at about 242° C. The second endothermic event onset at about 314° C. and centered from about 320° C. to about 340° C. and peak maximum centered at approximately 336° C.



FIG. 33 is a TGA thermogram of Form I.


Form I was further characterized by solution 1H NMR. The spectrum is reported in FIG. 34. The spectrum is consistent with one half molar equivalent of THF present, as well as the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form I are presented below in the Examples section.


9. Form J


Based on the available characterization data, Form J appears to be an anhydrous polymorphic form of Compound 1. Form J was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36a summarizes some of these results.



FIG. 35 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form J. The XRPD pattern confirms that Form J is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 9.









TABLE 9







Characteristic XRPD Peaks (CuKα) of Form J











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
4.1200
21.42934
15
80


2
4.4800
19.70812
90
736


3
4.8696
18.13218
491
3649


4
5.2400
16.85127
31
231


5
6.8000
12.98849
28
173


6
7.2697
12.15031
69
675


7
8.4600
10.44328
19
208


8
8.8800
9.95026
49
381


9
9.1910
9.61425
173
1428


10
9.7334
9.07968
131
1178


11
10.4330
8.47234
103
1118


12
13.4420
6.58181
16
146


13
14.6589
6.03804
109
1062


14
15.3600
5.76398
57
694


15
16.0400
5.52112
37
707


16
16.3600
5.41384
86
0


17
16.9600
5.22364
124
0


18
17.4800
5.06940
197
3351


19
18.1600
4.88108
29
0


20
18.4800
4.79728
22
241


21
19.6400
4.51647
113
931


22
20.0400
4.42722
199
2536


23
20.9600
4.23492
79
1497


24
22.0616
4.02589
123
2087


25
22.7855
3.89960
28
106


26
23.0022
3.86335
23
167


27
24.0000
3.70494
29
199


28
24.7200
3.59863
95
1539


29
25.2440
3.52511
167
2054


30
25.8000
3.45039
44
0


31
26.1200
3.40884
32
0


32
26.3960
3.37382
46
673


33
28.2075
3.16113
15
69


34
28.7378
3.10399
17
143


35
29.7000
3.00559
15
89


36
30.5186
2.92681
17
112


37
31.7350
2.81734
15
71


38
32.7628
2.73127
17
39


39
33.1700
2.69866
16
228


40
40.5466
2.22310
16
112









This unique set of XRPD peak positions or a subset thereof can be used to identify Form J. One such subset comprises peaks at about 4.9, 17.5 and 20.0°2θ. Another subset comprises peaks at about 9.2, 22.1 and 25.2°2θ.



FIG. 36 shows a characteristic DSC thermogram of Form J. The thermogram shows a first endotherm centered at about 219° C., a forked exotherm with peaks centered at about 223° C. and 236° C., followed by a forked endotherm which onset at 302° C. with peaks centered at approximately 323° C., 328° C. and 338° C.



FIG. 37 is a TGA thermogram of Form J. TGA analysis showed no weight loss or only small weight losses likely due to residual solvents.


Form J was further characterized by solution 1H NMR. The spectrum is reported in FIG. 38. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form J are presented below in the Examples section.


10. Form K


Based on the available characterization data, Form K appears to be an anhydrous polymorphic form of Compound 1. Form K was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36a summarizes some of these results.



FIG. 39 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form K. The XRPD pattern confirms that Form K is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 10.









TABLE 10







Characteristic XRPD Peaks (CuKα) of Form K











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
4.5600
19.36254
69
1117


2
5.2901
16.69179
1302
14236


3
8.0800
10.93355
102
765


4
8.5269
10.36149
606
7757


5
9.4800
9.32180
76
0


6
10.5407
8.38602
502
8225


7
13.2675
6.66798
231
4157


8
15.5200
5.70492
145
1273


9
15.8800
5.57639
196
2279


10
17.9600
4.93498
122
1574


11
18.5502
4.77928
329
5144


12
19.4400
4.56248
39
317


13
20.0400
4.42722
45
666


14
20.7200
4.28343
29B
3200


15
21.3381
4.16073
396
6209


16
23.5200
3.77945
75
977


17
23.8800
3.72328
63
0


18
24.4000
3.64510
71
1089


19
26.0000
3.42430
61
975


20
26.4000
3.37332
64
900


21
29.0400
3.07238
45
953


22
29.4000
3.03557
44
0


23
29.7600
2.99966
53
581


24
30.0800
2.96848
67
711


25
39.2819
2.29171
57
897


26
41.8467
2.15699
41
736









This unique set of XRPD peak positions or a subset thereof can be used to identify Form K. One such subset comprises peaks at about 5.3, 8.5 and 10.5°2θ. Another subset comprises peaks at about 13.3, 18.6 and 21.3°2θ.



FIG. 40 shows a characteristic DSC thermogram of Form K. An endotherm which onset at about 306° C. and centered at about 322° C. (peak maximum) was observed.



FIG. 41 is a TGA thermogram of Form K. TGA analysis showed no weight loss or only small weight losses likely due to residual solvents.


Form K was further characterized by solution 1H NMR. The spectrum is reported in FIG. 42. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form K are presented below in the Examples section.


11. Form L


Based on the available characterization data, Form L appears to be a channel hydrate polymorphic form of Compound 1 that is stable at ambient conditions. Form L was characterized by techniques including XRPD, DSC, TGA, 1H-NMR and moisture sorption analysis. Table 36a summarizes some of these results.


Form L is consistent with a channel hydrate based on KF and moisture sorption data (FIG. 47). KF analysis showed 2.9% water where 3.2% is theoretical for a monohydrate. The moisture sorption curve showed Form L to be moderately hygroscopic with a maximum water uptake of 3.9% at 90% RH. The shape of the curve is consistent with water able to be freely bound/removed based on temperature and relative humidity without significantly affecting the unit cell (i.e. form). The experiment did not time out (>4 hours) at any point and hysteresis was not observed upon desorption. Slurry experiments showed Form L to convert to the monohydrate Form A in water and to the anhydrate Form C in all other solvents. This is consistent with Form C being more thermodynamically stable than Form L at ambient temperature in non-aqueous environments.



FIG. 43 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form L. XRPD analysis which showed the same pattern before and after drying at 80° C. for one hour. The XRPD pattern confirms that Form L is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 11.









TABLE 11







Characteristic XRPD Peaks (CuKα) of Form L











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
4.6800
18.86633
216
3765


2
5.2436
16.83971
3437
28787


3
10.3883
8.50870
1268
10874


4
15.5487
5.69445
945
8156


5
16.2000
5.46695
103
0


6
16.8800
5.24822
317
3104


7
17.2000
5.15129
161
2040


8
17.7805
4.98440
165
1404


9
19.7600
4.48931
109
1601


10
20.2400
4.38392
227
3043


11
20.7515
4.27700
1547
11336


12
21.8685
4.06100
146
1064


13
23.1344
3.84157
290
2388


14
24.3913
3.64638
667
5099


15
25.3923
3.50486
224
1910


16
25.9912
3.42544
174
1251


17
26.6000
3.34841
155
1724


18
26.9200
3.30933
262
1535


19
27.2400
3.27117
104
1360


20
29.4781
3.02770
190
1539


21
32.0605
2.78948
237
2340


22
39.3236
2.28937
287
2215


23
42.7408
2.11391
107
1358









This unique set of XRPD peak positions or a subset thereof can be used to identify Form L. One such subset comprises peaks at about 5.2, 10.4 and 20.7°2θ. Another subset comprises peaks at about 15.5, 16.9 and 24.4°2θ.



FIG. 44 shows a characteristic DSC thermogram of Form L. An endotherm which onset at about 303° C. and centered at approximately 333° C. (peak maximum) was observed. FIG. 45 is a TGA thermogram of Form L, showing a weight loss of approximately 1.7% at a temperature below 100° C. The theoretical weight loss for a monohydrate is 3.2%.


Form L was further characterized by solution 1H NMR. The spectrum is reported in FIG. 46. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form L are presented below in the Examples section.


12. Form M


Based on the available characterization data, Form M appears to be an hydrate polymorphic form of Compound 1. Form M was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36a summarizes some of these results.



FIG. 48 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form M. The XRPD pattern confirms that Form M is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 12.









TABLE 12







Characteristic XRPD Peaks (CuKα) of Form M











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
4.4800
19.70812
84
988


2
4.6400
19.02888
167
0


3
5.1309
17.20934
1349
13342


4
5.6000
15.76875
48
896


5
8.2465
10.71316
551
5560


6
9.4816
9.32023
136
1393


7
10.2400
8.63159
557
6743


8
12.6000
7.01968
69
638


9
13.1200
6.74261
80
1022


10
15.0400
5.88589
118
1017


11
15.3200
5.77894
171
1404


12
17.2435
5.13839
122
1623


13
18.1029
4.89635
358
4950


14
19.5200
4.54397
72
713


15
20.0400
4.42722
309
3010


16
20.5600
4.31640
243
3303


17
22.2434
3.99339
65
947


18
23.6532
3.75847
48
438


19
24.7200
3.59863
46
484


20
25.0400
3.55337
75
826


21
25.7674
3.45468
132
1296


22
26.2400
3.39352
54
545


23
28.5600
3.12291
42
615


24
29.0000
3.07652
46
497


25
30.0818
2.96830
51
618









This unique set of XRPD peak positions or a subset thereof can be used to identify Form M. One such subset comprises peaks at about 5.1, 8.2 and 10.2°2θ. Another subset comprises peaks at about 18.1 and 20.6°2θ.



FIG. 49 shows a characteristic DSC thermogram of Form M. An endotherm onset at about 309° C. and centered at about 332° C. (peak maximum) was observed.



FIG. 50 is a TGA thermogram of Form M, showing a weight loss of approximately 6.0% at a temperature below 200° C. The theoretical weight loss for a monohydrate is 3.2%.


Form M was further characterized by solution 1H NMR. The spectrum is reported in FIG. 51. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form M are presented below in the Examples section.


13. Form N


Based on the available characterization data, Form N appears to be an hydrate polymorphic form of Compound 1. Form N was characterized by techniques including XRPD, DSC, TGA, and 1H-NMR. Table 36a summarizes some of these results.



FIG. 52 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form N. The XRPD pattern confirms that Form N is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 13.









TABLE 13







Characteristic XRPD Peaks (CuKα) of Form N











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
4.4800
19.70812
56
933


2
5.1562
17.12495
1350
11632


3
5.6400
15.65701
93
691


4
6.6800
13.22154
66
546


5
6.9600
12.69025
88
689


6
8.4202
10.49255
663
5474


7
9.4400
9.36121
43
934


8
10.2867
8.59251
822
8368


9
12.8000
6.91045
100
1018


10
13.2749
6.66428
221
1576


11
13.8514
6.38818
85
641


12
14.9200
5.93296
64
502


13
15.3909
5.75248
328
3053


14
16.3600
5.41384
48
583


15
17.3939
5.09430
211
2105


16
17.8000
4.97898
68
481


17
18.2000
4.87044
82
319


18
18.5708
4.77402
644
5287


19
19.4182
4.56756
279
2241


20
19.9899
4.43820
378
3716


21
20.6400
4.29985
268
2535


22
20.9600
4.23492
366
3328


23
21.6254
4.10610
55
605


24
22.4400
3.95885
58
399


25
22.7200
3.91069
45
344


26
23.9792
3.70810
284
2728


27
24.3600
3.65099
61
438


28
25.2400
3.52566
94
1563


29
25.6800
3.46624
93
0


30
26.1946
3.39930
209
2806


31
28.4000
3.14014
60
491


32
28.6000
3.11864
70
514


33
29.3610
3.03951
112
1487


34
29.8000
2.99573
67
0


35
30.4800
2.93043
79
1313


36
30.8000
2.90071
52
358


37
40.5600
2.22239
64
773


38
40.9200
2.20367
42
0


39
41.2800
2.18528
41
525









This unique set of XRPD peak positions or a subset thereof can be used to identify Form N. One such subset comprises peaks at about 5.2, 8.4 and 10.3°2θ. Another subset comprises peaks at about 18.6, 20.0 and 21.0°2θ.



FIG. 53 shows a characteristic DSC thermogram of Form N. An endotherm which onset at about 313° C. and centered at about 333° C. (peak maximum) was observed.



FIG. 54 is a TGA thermogram of Form N, showing a weight loss of approximately 6.2% at a temperature below 200° C. The theoretical weight loss for a monohydrate is 3.2%.


Form N was further characterized by solution 1H NMR. The spectrum is reported in FIG. 55. Chemical assignments were not performed; however, the spectra are consistent with the known chemical structure of Compound 1.


Further details related to the preparation and characterization of Form N are presented below in the Examples section.


14. Form O


Form O appears to be a dehydrate polymorphic form of Compound 1. It can be obtained by drying Form N under the TGA conditions. Form O was characterized by techniques including XRPD, DSC and TGA. Table 36a summarizes some of these results.



FIG. 56 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form O. The XRPD pattern confirms that Form O is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 14.









TABLE 14







Characteristic XRPD Peaks (CuKα) of Form O











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.1200
28.29512
39
124


2
3.3622
26.25737
51
214


3
3.7593
23.48462
31
211


4
4.6121
19.14393
20
178


5
5.6613
15.59815
46
286


6
5.8400
15.12125
59
300


7
6.3387
13.93265
398
3172


8
6.8533
12.88759
22
71


9
7.4819
11.80618
20
124


10
10.1200
8.73367
49
307


11
10.5047
8.41467
352
2325


12
11.0000
8.03687
16
102


13
11.3828
7.76744
106
645


14
12.2400
7.22532
50
407


15
12.6104
7.01392
520
3330


16
13.0000
6.80458
19
150


17
15.7200
5.63278
17
86


18
16.0000
5.53483
60
393


19
16.2400
5.45357
59
319


20
16.6866
5.30861
31
277


21
17.1722
5.15957
102
722


22
17.6000
5.03511
87
976


23
18.3600
4.82836
23
99


24
18.6000
4.76660
52
264


25
18.8711
4.69873
118
886


26
19.8255
4.47463
102
801


27
21.0419
4.21863
261
1777


28
21.6800
4.09588
39
241


29
21.9200
4.05157
95
626


30
22.3353
3.97717
123
922


31
22.7755
3.90129
117
1174


32
24.4755
3.63403
23
142


33
24.8800
3.57585
28
106


34
25.2945
3.51819
407
2742


35
26.4067
3.37248
196
1444


36
26.7200
3.33364
73
557


37
28.5798
3.12079
50
458


38
31.3200
2.85372
19
86


39
31.5200
2.83607
53
255


40
31.7600
2.81518
55
367


41
33.9470
2.63865
16
91


42
34.6000
2.59033
21
123


43
34.9334
2.56637
41
379


44
37.2382
2.41265
25
189


45
38.9265
2.31181
30
194


46
43.4496
2.08105
35
170


47
43.6566
2.07167
28
115









This unique set of XRPD peak positions or a subset thereof can be used to identify Form O. One such subset comprises peaks at about 6.3, 12.6 and 25.3°2θ. Another subset comprises peaks at about 10.5 and 21.0°2θ.



FIG. 57 shows a characteristic DSC thermogram of Form O. An endotherm was observed at approximately 327° C. (peak maximum). FIG. 58 is a TGA thermogram of Form O.


Further details related to the preparation and characterization of Form O are presented below in the Examples section.


15. Form P


Form P appears to be a metastable form of Compound 1. Form P was characterized by techniques including XRPD. FIG. 59 shows a characteristic XRPD spectrum (CuKα, λ=1.5418 Å) of Form P. The XRPD pattern confirms that Form P is crystalline. Major X-Ray diffraction lines expressed in °2θ and their relative intensities are summarized in Table 15.









TABLE 15







Characteristic XRPD Peaks (CuKα) of Form P











Peak No.
2θ (°)
d-spacing
Intensity
I/Io














1
3.0991
28.48590
49
145


2
3.3181
26.60625
54
133


3
3.5309
25.00321
37
153


4
4.3600
20.25027
69
604


5
5.0151
17.60644
825
6787


6
5.6008
15.76650
36
311


7
6.4000
13.79934
29
159


8
6.8318
12.92810
35
240


9
7.2314
12.21458
35
190


10
8.5233
10.36586
26
160


11
8.8800
9.95026
32
168


12
9.4048
9.39616
203
1815


13
9.9851
8.85136
224
1682


14
10.3600
8.53188
54
490


15
14.6400
6.04580
27
426


16
14.9594
5.91742
106
809


17
16.8400
5.26059
39
0


18
17.2400
5.13943
72
901


19
18.0443
4.91212
45
442


20
18.8254
4.71003
34
372


21
19.7600
4.48931
60
533


22
20.0400
4.42722
91
523


23
20.3200
4.36684
35
187


24
20.6410
4.29965
28
224


25
21.4996
4.12984
32
388


26
22.7816
3.90025
35
337


27
24.0405
3.69879
27
178


28
24.7600
3.59291
36
257


29
25.0000
3.55896
56
379


30
25.6989
3.46373
71
1049


31
27.7606
3.21100
30
503


32
28.3785
3.14247
27
187


33
37.8316
2.37616
26
251









This unique set of XRPD peak positions or a subset thereof can be used to identify Form P. One such subset comprises peaks at about 5.0, 9.4 and 10.0°2θ. Another subset comprises peaks at about 17.2 and 25.7°2θ.


Further details related to the preparation and characterization of Form P are presented below in the Examples section.


Indications for Use of Compound 1

The present invention also relates to methods to alter, preferably to reduce kinase activity within a subject by administrating Compound 1 in a form selected from the group consisting of Forms A, B, C, D, E, F, G, 1, J, K, L, M, N, O, and P and Amorphous Form.


Kinases are believed to contribute to the pathology and/or symptomology of several different diseases such that reduction of the activity of one or more kinases in a subject through inhibition may be used to therapeutically address these disease states. Examples of various diseases that may be treated using Compound 1 of the present invention are described herein. It is noted that additional diseases beyond those disclosed herein may be later identified as the biological roles that kinases play in various pathways becomes more fully understood.


Compound 1 may be used to treat or prevent cancer. In one embodiment, Compound 1 is used in a method comprising administering a therapeutically effective amount of Compound 1 or a composition comprising Compound 1 to a mammalian species in need thereof. In particular embodiments, the cancer is selected from the group consisting of squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, small-cell lung cancer, non small-cell lung cancers (e.g., large cell lung cancer, adenocarcinoma and squamous cell carcinoma), bladder cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, glioma, colorectal cancer, genitourinary cancer, gastrointestinal cancer, thyroid cancer, skin cancer, kidney cancer, rectal cancer, colonic cancer, cervical cancer, mesothelioma, pancreatic cancer, liver cancer, uterus cancer, cerebral tumor cancer, urinary bladder cancer and blood cancers including multiple myeloma, chronic myelogenous leukemia and acute lymphocytic leukemia. In other embodiments, Compound 1 is useful for inhibiting growth of cancer, for suppressing metastasis of cancer, for suppressing apoptosis and the like.


In another embodiment, Compound 1 is used in a method for treating inflammation, inflammatory bowel disease, psoriasis, or transplant rejection, comprising administration to a mammalian species in need thereof a therapeutically effective amount of Compound 1 or a composition comprising Compound 1.


In another embodiment, Compound 1 is used in a method for preventing or treating amyotrophic lateral sclerosis, corticobasal degeneration, Down syndrome, Huntington's Disease, Parkinson's Disease, postencephelatic parkinsonism, progressive supranuclear palsy, Pick's Disease, Niemann-Pick's Disease, stroke, head trauma and other chronic neurodegenerative diseases, Bipolar Disease, affective disorders, depression, schizophrenia, cognitive disorders, hair loss and contraceptive medication, comprising administration to a mammalian species in need thereof of a therapeutically effective amount of Compound 1 or a composition comprising Compound 1.


In yet another embodiment, Compound 1 is used in a method for preventing or treating mild Cognitive Impairment, Age-Associated Memory Impairment, Age-Related Cognitive Decline, Cognitive Impairment No Dementia, mild cognitive decline, mild neurocognitive decline, Late-Life Forgetfulness, memory impairment and cognitive impairment and androgenetic alopecia, comprising administering to a mammal, including man in need of such prevention and/or treatment, a therapeutically effective amount of Compound 1 or a composition comprising Compound 1.


In a further embodiment, Compound 1 is used in a method for preventing or treating dementia related diseases, Alzheimer's Disease and conditions associated with kinases, comprising administration to a mammalian species in need thereof of a therapeutically effective amount of Compound 1 or a composition comprising Compound 1. In one particular variation, the dementia related diseases are selected from the group consisting of Frontotemporal dementia Parkinson's Type, Parkinson dementia complex of Guam, HIV dementia, diseases with associated neurofibrillar tangle pathologies, predemented states, vascular dementia, dementia with Lewy bodies, Frontotemporal dementia and dementia pugilistica.


In another embodiment, Compound 1 is used in a method for treating arthritis comprising administration to a mammalian species in need thereof of a therapeutically effective amount of Compound 1 or a composition comprising Compound 1.


Compositions, according to the present invention, may be administered, or coadministered with other active agents. These additional active agents may include, for example, one or more other pharmaceutically active agents. Coadministration in the context of this invention is intended to mean the administration of more than one therapeutic agent, one of which includes Compound 1. Such co-administration may also be coextensive, that is, occurring during overlapping periods of time or may be sequential, that is, occurring during non-overlapping periods of time. Examples of co-administration of Compound 1 with other active ingredients in a combination therapy are described in U.S. Patent Publication No. 2007-0117816, published May 24, 2007 (see Compound 112) and U.S. Patent Application Nos. 60/912,625 and 60/912,629, filed Apr. 18, 2007 (see Compound 83), which are incorporated herein by reference in their entireties.


For oncology indications, Compound 1 may be administered in conjunction with other agents to inhibit undesirable and uncontrolled cell proliferation. Examples of other anti-cell proliferation agents that may be used in conjunction with Compound 1 include, but are not limited to, retinoid acid and derivatives thereof, 2-methoxyestradiol, ANGIOSTATIN™ protein, ENDOSTATIN™ protein, suramin, squalamine, tissue inhibitor of metalloproteinase-1, tissue inhibitor of metalloproteinase-2, plasminogen activator inhibitor-1, plasminogen activator inhibitor-2, cartilage-derived inhibitor, paclitaxel, platelet factor 4, protamine sulfate (clupeine), sulfated chitin derivatives (prepared from queen crab shells), sulfated polysaccharide peptidoglycan complex (sp-pg), staurosporine, modulators of matrix metabolism, including for example, proline analogs ((l-azetidine-2-carboxylic acid (LACA)), cishydroxyproline, d,l-3,4-dehydroproline, thiaproline, beta.-aminopropionitrile fumarate, 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone, methotrexate, mitoxantrone, heparin, interferons, 2 macroglobulin-serum, chimp-3, chymostatin, beta.-cyclodextrin tetradecasulfate, eponemycin; fumagillin, gold sodium thiomalate, d-penicillamine (CDPT), beta.-1-anticollagenase-serum, alpha.2-antiplasmin, bisantrene, lobenzarit disodium, n-2-carboxyphenyl-4-chloroanthronilic acid disodium or “CCA”, thalidomide; angostatic steroid, carboxyaminoimidazole; metalloproteinase inhibitors such as BB94. Other anti-angiogenesis agents that may be used include antibodies, preferably monoclonal antibodies against these angiogenic growth factors: bFGF, aFGF, FGF-5, VEGF isoforms, VEGF-C, HGF/SF and Ang-1/Ang-2. Ferrara N. and Alitalo, K. “Clinical application of angiogenic growth factors and their inhibitors” (1999) Nature Medicine 5:1359-1364.


In another embodiment, a therapeutic method is provided that comprises administering Compound 1. In another embodiment, a method of inhibiting cell proliferation is provided that comprises contacting a cell with an effective amount of Compound 1. In another embodiment, a method of inhibiting cell proliferation in a patient is provided that comprises administering to the patient a therapeutically effective amount of Compound 1.


In another embodiment, a method of treating a condition in a patient which is known to be mediated by one or more kinases, or which is known to be treated by kinase inhibitors, is provided comprising administering to the patient a therapeutically effective amount of Compound 1. In another embodiment, a method is provided for using Compound 1 in order to manufacture a medicament for use in the treatment of a disease state which is known to be mediated by one or more kinases, or which is known to be treated by kinase inhibitors.


In another embodiment, a method is provided for treating a disease state for which kinases possess activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering Compound 1 to a subject such that Compound 1 is present in the subject in a therapeutically effective amount for the disease state.


The present invention relates generally to a method comprising administering between 1 mg/day and 500 mg/day of Compound 1 to a patient, optionally between 1 mg/day and 400 mg/day of Compound 1, optionally between 1 mg/day and 250 mg/day of Compound 1, optionally between 2.5 mg/day and 200 mg/day of Compound 1, optionally between 2.5 mg/day and 150 mg/day of Compound 1, and optionally between 5 mg/day and 100 mg/day of Compound 1 (in each instance based on the molecular weight of the free base form of Compound 1). Specific dosage amounts that may be used include, but are not limited to 2.5 mg, 5 mg, 6.25 mg, 10 mg, 12.5 mg, 20 mg, 25 mg, 50 mg, 75 mg, 100 mg, 200 mg, 250 mg, 400 mg and 500 mg of Compound 1 per day. It is noted that the dosage may be administered as a daily dose or weekly dose, once daily or multiple doses per day. It is noted that Compound 1 may be administered in a form selected from the group consisting of Forms A, B, C, D, E, F, G, I, J, K, L, M, N, O, and P and Amorphous Form. However, the dosage amounts and ranges provided herein are always based on the molecular weight of the free base form of Compound 1.


Compound 1 may be administered by any route of administration. In particular embodiments, however, the method of the present invention is practiced by administering Compound 1 orally.


Pharmaceutical Compositions Comprising Compound 1 where at Least One of Form A Through Form P, or Amorphous Form is Present


Compound 1 may be used in various pharmaceutical compositions where at least a portion of Compound 1 is present in the composition in a form selected from the group consisting of Forms A, B, C, D, E, F, G, I, J, K, L, M, N, O, and P and Amorphous Form. The pharmaceutical composition should contain a sufficient quantity of Compound 1 to reduce kinase activity in vivo sufficiently to provide the desired therapeutic effect. Such pharmaceutical compositions may comprise Compound 1 present in the composition in a range of between 0.005% and 100% (weight/weight), optionally 0.1-95%, and optionally 1-95%.


In particular embodiments, the pharmaceutical compositions comprise at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1 in a form selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, Amorphous Form, and mixtures thereof. In another embodiment, a particular polymorphic form selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, Amorphous Form, and mixtures thereof may comprise at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of the total amount of Compound 1 (weight/weight) in the pharmaceutical composition.


In addition to Compound 1, the pharmaceutical composition may comprise one or more additional components that do not deleteriously affect the use of Compound 1. For example, the pharmaceutical compositions may include, in addition to Compound 1, conventional pharmaceutical carriers; excipients; diluents; lubricants; binders; wetting agents; disintegrating agents; glidants; sweetening agents; flavoring agents; emulsifying agents; solubilizing agents; pH buffering agents; perfuming agents; surface stabilizing agents; suspending agents; and other conventional, pharmaceutically inactive agents. In particular, the pharmaceutical compositions may comprise lactose, mannitol, glucose, sucrose, dicalcium phosphate, magnesium carbonate, sodium saccharin, carboxymethylcellulose, magnesium stearate, calcium stearate, sodium crosscarmellose, talc, starch, natural gums (e.g., gum acaciagelatin), molasses, polyvinylpyrrolidine, celluloses and derivatives thereof, povidone, crospovidones acetate, sodium citrate, cyclodextrine derivatives, sorbitan monolaurate, triethanolamine sodium acetate, triethanolamine oleate, biocompatible polymers, such as collagen, ethylene vinyl acetate, polyanhydrides, polyglycolic acid, polyorthoesters, polylactic acid and others such agents.


Pharmaceutical compositions according to the present invention may be adapted for administration by any of a variety of routes. For example, pharmaceutical compositions according to the present invention can be administered orally, parenterally, intraperitoneally, intravenously, intraarterially, topically, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery (for example, by catheter or stent), subcutaneously, intraadiposally, intraarticularly, or intrathecally, optionally in a slow release dosage form. In particular embodiments, the pharmaceutical compounds are administered orally, by inhalation or by injection subcutaneously, intramuscularly, intravenously or directly into the cerebrospinal fluid.


In general, the pharmaceutical compositions of the present invention may be prepared in a gaseous, liquid, semi-liquid, gel, or solid form, and formulated in a manner suitable for the route of administration to be used.


Compositions according to the present invention are optionally provided for administration to humans and animals in unit dosage forms or multiple dosage forms, such as tablets, capsules, pills, powders, dry powders for inhalers, granules, sterile parenteral solutions or suspensions, oral solutions or suspensions, oil-water emulsions, sustained release formulations, such as, but not limited to, implants and microencapsulated delivery systems, containing suitable quantities of Compound 1. Methods of preparing such dosage forms are known in the art, and will be apparent to those skilled in this art; for example, see Remington's Pharmaceutical Sciences, 19th Ed. (Easton, Pa.: Mack Publishing Company, 1995).


Unit-dose forms, as used herein, refers to physically discrete units suitable for human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of Compound 1 sufficient to produce the desired therapeutic effect, in association with a pharmaceutical carrier, vehicle or diluent. Examples of unit-dose forms include ampoules and syringes, and individually packaged tablets or capsules. Unit-dose forms may be administered in fractions or multiples thereof. A multiple-dose form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dose form. Examples of multiple-dose forms include vials, bottles of tablets or capsules, or bottles of pints or gallons. Hence, multiple dose form may be viewed as a multiple of unit-doses that are not segregated in packaging.


In general, the total amount of Compound 1 in a pharmaceutical composition according to the present invention should be sufficient to provide a desired therapeutic effect. This amount may be delivered as a single per day dosage, multiple dosages per day to be administered at intervals of time, or as a continuous release dosage form. Compound 1 may advantageously be used when administered to a patient at a daily dose of between 1 mg/day and 250 mg/day of Compound 1, optionally between 2.5 mg and 200 mg of Compound 1, optionally between 2.5 mg and 150 mg of Compound 1, and optionally between 5 mg and 100 mg of Compound 1 (in each instance based on the molecular weight of the free base form of Compound 1). Specific dosage amounts that may be used include, but are not limited to 2.5 mg, 5 mg, 6.25 mg, 10 mg, 12.5 mg, 20 mg, 25 mg, 50 mg, 75 mg, and 100 mg of Compound 1 per day. It may be desirable for Compound 1 to be administered one time per day. Accordingly, pharmaceutical compositions of the present invention may be in the form of a single dose form comprising between 1 mg/day and 250 mg/day of Compound 1, optionally between 2.5 mg and 200 mg of Compound 1, optionally between 2.5 mg and 150 mg of Compound 1, and optionally between 5 mg and 100 mg of Compound 1. In specific embodiments, the pharmaceutical composition comprises 2.5 mg, 5 mg, 6.25 mg, 10 mg, 12.5 mg, 20 mg, 25 mg, 50 mg, 75 mg or 100 mg of Compound 1.


A. Formulations for Oral Administration


Oral pharmaceutical dosage forms may be as a solid, gel or liquid where at least a portion of Compound 1 is present in the composition in a form selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form.


In certain embodiments, Compound 1 is provided as solid dosage forms. Examples of solid dosage forms include, but are not limited to pills, tablets, troches, capsules, granules, and bulk powders. More specific examples of oral tablets include compressed, chewable lozenges, troches and tablets that may be enteric-coated, sugar-coated or film-coated. Examples of capsules include hard or soft gelatin capsules. Granules and powders may be provided in non-effervescent or effervescent forms. The powders may be prepared by lyophilization or by other suitable methods.


The tablets, pills, capsules, troches and the like may optionally contain one or more of the following ingredients, or compounds of a similar nature: a binder; a diluent; a disintegrating agent; a lubricant; a glidant; a coloring agent; a sweetening agent; a flavoring agent; and a wetting agent.


Examples of binders that may be used include, but are not limited to, microcrystalline cellulose, gum tragacanth, glucose solution, acacia mucilage, gelatin solution, sucrose and starch paste.


Examples of diluents that may be used include, but are not limited to, lactose, sucrose, starch, kaolin, salt, mannitol and dicalcium phosphate.


Examples of disintegrating agents that may be used include, but are not limited to, crosscarmellose sodium, sodium starch glycolate, alginic acid, corn starch, potato starch, bentonite, methylcellulose, agar and carboxymethylcellulose.


Examples of lubricants that may be used include, but are not limited to, talc, starch, magnesium or calcium stearate, lycopodium and stearic acid.


Examples of glidants that may be used include, but are not limited to, colloidal silicon dioxide.


Examples of coloring agents that may be used include, but are not limited to, any of the approved certified water soluble FD and C dyes, mixtures thereof; and water insoluble FD and C dyes suspended on alumina hydrate.


Examples of sweetening agents that may be used include, but are not limited to, sucrose, lactose, mannitol and artificial sweetening agents such as sodium cyclamate and saccharin, and any number of spray-dried flavors.


Examples of flavoring agents that may be used include, but are not limited to, natural flavors extracted from plants such as fruits and synthetic blends of compounds that produce a pleasant sensation, such as, but not limited to peppermint and methyl salicylate.


Examples of wetting agents that may be used include, but are not limited to, propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate and polyoxyethylene lauryl ether.


Examples of anti-emetic coatings that may be used include, but are not limited to, fatty acids, fats, waxes, shellac, ammoniated shellac and cellulose acetate phthalates.


Examples of film coatings that may be used include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000 and cellulose acetate phthalate.


When the dosage form is a pill, tablet, torches, or the like, Compound 1 may optionally be provided in a composition that protects it from the acidic environment of the stomach. For example, the composition can be formulated in an enteric coating that maintains its integrity in the stomach and releases the active compound in the intestine. The composition may also be formulated in combination with an antacid or other such ingredient.


When the dosage unit form is a capsule, it may optionally additionally comprise a liquid carrier such as a fatty oil. In addition, dosage unit forms may optionally additionally comprise various other materials that modify the physical form of the dosage unit, for example, coatings of sugar and other enteric agents.


Compound 1 may also be administered as a component of an elixir, emulsion, suspension, microsuspension, syrup, wafer, sprinkle, chewing gum or the like. A syrup may optionally comprise, in addition to the active compounds, sucrose as a sweetening agent and certain preservatives, dyes and colorings and flavors.


Alternatively, liquid or semi-solid oral formulations may be prepared by dissolving or dispersing the active compound or salt in vegetable oils, glycols, triglycerides, propylene glycol esters (e.g. propylene carbonate) and other such carriers, and encapsulating these solutions or suspensions in hard or soft gelatin capsule shells. Other useful formulations include those set forth in U.S. Pat. Nos. Re 28,819 and 4,358,603.


Examples of oral formulations that may be used to administer Compound 1 has been described in U.S. patent application Ser. No. 11/531,671, filed Sep. 13, 2006, the disclosure of which is herein expressly incorporated by reference in its entirety.


Exemplary tablet formulations are provided below. It is noted that the examples are, by way of illustration but not limitation. It is also noted that Compound 1 is present in the formulation in a form selected from the group consisting of one or more of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form. It is also noted that the formulations provided herein may be varied as is known in the art.














12.5 mg of Compound 1 (weight of free base form) per tablet


Core Tablet Formulation











(1)
Compound 1
 17.0 mg



(2)
Lactose Monohydrate, NF, Ph, Eur
224.6 mg




(FOREMOST 316 FAST FLO)



(3)
Microcrystalline Cellulose, NF, Ph, Eur
120.1 mg




(AVICEL PH 102)



(4)
Croscarmellose Sodium, NF, Ph, Eur
 32.0 mg




(AC-DO-SOL)



(5)
Colloidal Silicon Dioxide, NF, Ph, Eur
 3.2 mg




(CAB-O-SIL M-5P)



(6)
Magnesium Stearate, NF, Ph, Eur
 3.2 mg




(MALLINCKRODT, Non-bovine Hyqual)











TOTAL (per tablet)
400.0 mg







25 mg of Compound 1 (weight of free base form) per tablet


Core Tablet Formulation











(1)
Compound 1
 34.0 mg



(2)
Lactose Monohydrate, NF, Ph, Eur
207.6 mg




(FOREMOST 316 FAST FLO)



(3)
Microcrystalline Cellulose, NF, Ph, Eur
120.1 mg




(AVICEL PH 102)



(4)
Croscarmellose Sodium, NF, Ph, Eur
 32.0 mg




(AC-DO-SOL)



(5)
Colloidal Silicon Dioxide, NF, Ph, Eur
 3.2 mg




(CAB-O-SIL M-5P)



(6)
Magnesium Stearate, NF, Ph, Eur
 3.2 mg




(MALLINCKRODT, Non-bovine Hyqual)











TOTAL (per tablet)
400.0 mg







50 mg of Compound 1 (weight of free base form) per tablet


Core Tablet Formulation











(1)
Compound 1
 68.0 mg



(2)
Lactose Monohydrate, NF, Ph, Eur
173.6 mg




(FOREMOST 316 FAST FLO)



(3)
Microcrystalline Cellulose, NF, Ph, Eur
120.1 mg




(AVICEL PH 102)



(4)
Croscarmellose Sodium, NF, Ph, Eur
 32.0 mg




(AC-DO-SOL)



(5)
Colloidal Silicon Dioxide, NF, Ph, Eur
 3.2 mg




(CAB-O-SIL M-5P)



(6)
Magnesium Stearate, NF, Ph, Eur
 3.2 mg




(MALLINCKRODT, Non-bovine Hyqual)











TOTAL (per tablet)
400.0 mg







Film Coat (12.0 mg in total)



(1) Opadry II 85F18422, White - Portion 1 (COLORCON)



(2) Opadry II 85F18422, White - Portion 2 (COLORCON)



(3) Opadry II 85F18422, White - Portion 3 (COLORCON)






B. Injectables, Solutions and Emulsions


Compound 1 present in a form or a mixture of forms selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form may be formulated for parenteral administration. Parenteral administration generally characterized by injection, either subcutaneously, intramuscularly or intravenously. Implantation of a slow-release or sustained-release system, such that a constant level of dosage is maintained (see, e.g., U.S. Pat. No. 3,710,795) is also contemplated herein. The percentage of active compound contained in such parenteral compositions is highly dependent on the route of administration and the indication of disease to be treated.


Injectables may be prepared in any conventional form. These formulations include, but are not limited to, sterile solutions, suspensions, microsuspensions, and emulsions ready for injection, and solid forms, e.g., lyophilized or other powders including hypodermic tablets, ready to be combined with a carrier just prior to use. Generally, the resulting formulation may be a solution, microsuspension, suspension and emulsion. The carrier may be an aqueous, non-aqueous liquid, or a solid vehicle that can be suspended in liquid.


Examples of carriers that may be used in conjunction with injectables according to the present invention include, but are not limited to water, saline, dextrose, glycerol or ethanol. The injectable compositions may also optionally comprise minor amounts of non-toxic auxiliary substances such as wetting or emulsifying agents, pH buffering agents, stabilizers, solubility enhancers, and other such agents, such as for example, sodium acetate, sorbitan monolaurate, triethanolamine oleate and cyclodextrins.


When administered intravenously, examples of suitable carriers include, but are not limited to physiological saline or phosphate buffered saline (PBS), and solutions containing thickening and solubilizing agents, such as glucose, polyethylene glycol, and polypropylene glycol and mixtures thereof.


Examples of pharmaceutically acceptable carriers that may optionally be used in parenteral preparations include, but are not limited to aqueous vehicles, nonaqueous vehicles, antimicrobial agents, isotonic agents, buffers, antioxidants, local anesthetics, suspending and dispersing agents, emulsifying agents, sequestering or chelating agents and other pharmaceutically acceptable substances.


Examples of aqueous vehicles that may optionally be used include Sodium Chloride Injection, Ringers Injection, Isotonic Dextrose Injection, Sterile Water Injection, Dextrose and Lactated Ringers Injection.


Examples of nonaqueous parenteral vehicles that may optionally be used include fixed oils of vegetable origin, cottonseed oil, corn oil, sesame oil and peanut oil.


Antimicrobial agents in bacteriostatic or fungistatic concentrations may be added to parenteral preparations, particularly when the preparations are packaged in multiple-dose containers and thus designed to be stored and multiple aliquots to be removed. Examples of antimicrobial agents that may used include phenols or cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoic acid esters, thimerosal, benzalkonium chloride and benzethonium chloride.


Examples of isotonic agents that may be used include sodium chloride and dextrose. Examples of buffers that may be used include phosphate and citrate. Examples of antioxidants that may be used include sodium bisulfate. Examples of local anesthetics that may be used include procaine hydrochloride. Examples of suspending and dispersing agents that may be used include sodium carboxymethylcellulose, hydroxypropyl methylcellulose and polyvinylpyrrolidone. Examples of emulsifying agents that may be used include Polysorbate 80 (TWEEN 80). A sequestering or chelating agent of metal ions includes EDTA.


Pharmaceutical carriers may also optionally include ethyl alcohol, polyethylene glycol and propylene glycol for water miscible vehicles and sodium hydroxide, hydrochloric acid, citric acid or lactic acid for pH adjustment.


The concentration of Compound 1 in the parenteral formulation may be adjusted so that an injection administers a pharmaceutically effective amount sufficient to produce the desired pharmacological effect. The exact concentration of Compound 1 and/or dosage to be used will ultimately depend on the age, weight and condition of the patient or animal as is known in the art.


Unit-dose parenteral preparations may be packaged in an ampoule, a vial or a syringe with a needle. All preparations for parenteral administration should be sterile, as is known and practiced in the art.


Injectables may be designed for local and systemic administration. Typically a therapeutically effective dosage is formulated to contain a concentration of at least about 0.1% w/w up to about 90% w/w or more, preferably more than 1% w/w of Compound 1 to the treated tissue(s). Compound 1 may be administered at once, or may be divided into a number of smaller doses to be administered at intervals of time. It is understood that the precise dosage and duration of treatment will be a function of the location of where the composition is parenterally administered, the carrier and other variables that may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test data. It is to be noted that concentrations and dosage values may also vary with the age of the individual treated. It is to be further understood that for any particular subject, specific dosage regimens may need to be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations. Hence, the concentration ranges set forth herein are intended to be exemplary and are not intended to limit the scope or practice of the claimed formulations.


Compound 1 may optionally be suspended in micronized or other suitable form or may be derivatized to produce a more soluble active product or to produce a prodrug. The form of the resulting mixture depends upon a number of factors, including the intended mode of administration and the solubility of the compound in the selected carrier or vehicle. The effective concentration is sufficient for ameliorating the symptoms of the disease state and may be empirically determined.


C. Powders


Compound 1 in a form or a mixture of forms selected from the group consisting of one or more of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form may be prepared as powders, which can be reconstituted for administration as solutions, emulsions and other mixtures. The powders may also be formulated as solids or gels.


Powders of Compound 1 may be prepared by grinding, spray drying, lyophilization and other techniques that are well known in the art. Sterile, lyophilized powder may be prepared by dissolving Compound 1 in a sodium phosphate buffer solution containing dextrose or other suitable excipient. Subsequent sterile filtration of the solution followed by lyophilization under standard conditions known to those of skill in the art provides the desired formulation. Briefly, the lyophilized powder may optionally be prepared by dissolving dextrose, sorbitol, fructose, corn syrup, xylitol, glycerin, glucose, sucrose or other suitable agent, about 1-20%, preferably about 5 to 15%, in a suitable buffer, such as citrate, sodium or potassium phosphate or other such buffer known to those of skill in the art at, typically, about neutral pH. Then, Compound 1 is added to the resulting mixture, preferably above room temperature, more preferably at about 30-35° C., and stirred until it dissolves. The resulting mixture is diluted by adding more buffer to a desired concentration. The resulting mixture is sterile filtered or treated to remove particulates and to insure sterility, and apportioned into vials for lyophilization. Each vial may contain a single dosage or multiple dosages of Compound 1.


D. Topical Administration


Compound 1 present in a form or a mixture of forms selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form may also be administered as topical mixtures. Topical mixtures may be used for local and systemic administration. The resulting mixture may be a solution, suspension, microsuspension, emulsions or the like and are formulated as creams, gels, ointments, emulsions, solutions, elixirs, lotions, suspensions, tinctures, pastes, foams, aerosols, irrigations, sprays, suppositories, bandages, dermal patches or any other formulations suitable for topical administration.


Compound 1 may be formulated for topical applications to the respiratory tract. These pulmonary formulations can be in the form of an aerosol, solution, emulsion, suspension, microsuspension for a nebulizer, or as a microfine powder for insufflation, alone or in combination with an inert carrier such as lactose. In such a case, the particles of the formulation will typically have diameters of less than 50 microns, preferably less than 10 microns. Examples of aerosols for topical application, such as by inhalation are disclosed in U.S. Pat. Nos. 4,044,126, 4,414,209, and 4,364,923, which describe aerosols for delivery of a steroid useful for treatment inflammatory diseases, particularly asthma.


Compound 1 may also be formulated for local or topical application, such as for topical application to the skin and mucous membranes, such as in the eye, in the form of gels, creams, and lotions and for application to the eye or for intracisternal or intraspinal application. Topical administration is contemplated for transdermal delivery and also for administration to the eyes or mucosa, or for inhalation therapies. Nasal solutions or suspensions of Compound 1 alone or in combination with other pharmaceutically acceptable excipients can also be administered.


E. Formulations for Other Routes of Administration


Depending upon the disease state being treated, Compound 1 present in a form or a mixture of forms selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form may be formulated for other routes of administration, such as topical application, transdermal patches, and rectal administration. For example, pharmaceutical dosage forms for rectal administration are rectal suppositories, capsules and tablets for systemic effect. Rectal suppositories as used herein mean solid bodies for insertion into the rectum that melt or soften at body temperature releasing one or more pharmacologically or therapeutically active ingredients. Pharmaceutically acceptable substances utilized in rectal suppositories are bases or vehicles and agents to raise the melting point. Examples of bases include cocoa butter (theobroma oil), glycerin-gelatin, carbowax, (polyoxyethylene glycol) and appropriate mixtures of mono-, di- and triglycerides of fatty acids. Combinations of the various bases may be used. Agents to raise the melting point of suppositories include spermaceti and wax. Rectal suppositories may be prepared either by the compressed method or by molding. The typical weight of a rectal suppository is about 2 to 3 gm. Tablets and capsules for rectal administration may be manufactured using the same pharmaceutically acceptable substance and by the same methods as for formulations for oral administration.


Kits and Articles of Manufacture Comprising Compound 1 Polymorphs

The present invention is also directed to kits and other articles of manufacture for treating diseases associated with kinases. It is noted that diseases are intended to cover all conditions for which kinases possess activity that contributes to the pathology and/or symptomology of the condition.


In one embodiment, a kit is provided that comprises a pharmaceutical composition comprising Compound 1 where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in a form selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form; and instructions for use of the kit. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1. The instructions may indicate the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also comprise packaging materials. The packaging material may comprise a container for housing the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.


In another embodiment, an article of manufacture is provided that comprises a pharmaceutical composition comprising Compound 1 where greater than 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97% or 99% of Compound 1 (by weight) is present in the composition in a form selected from the group consisting of Form A, Form B, Form C, Form D, Form E, Form F, Form G, From I, Form J, Form K, Form L, Form M, Form N, Form O, Form P, and Amorphous Form; and packaging materials. Optionally, the composition comprises at least 0.1%, 0.25%, 0.5%, 1%, 5%, 10%, 25%, 50%, 75%, 80%, 85%, 90%, 95%, 97%, or 99% of Compound 1. The packaging material may comprise a container for housing the composition. The container may optionally comprise a label indicating the disease state for which the composition is to be administered, storage information, dosing information and/or instructions regarding how to administer the composition. The kit may also optionally comprise additional components, such as syringes for administration of the composition. The kit may comprise the composition in single or multiple dose forms.


It is noted that the packaging material used in kits and articles of manufacture according to the present invention may form a plurality of divided containers such as a divided bottle or a divided foil packet. The container can be in any conventional shape or form as known in the art which is made of a pharmaceutically acceptable material, for example a paper or cardboard box, a glass or plastic bottle or jar, a re-sealable bag (for example, to hold a “refill” of tablets for placement into a different container), or a blister pack with individual doses for pressing out of the pack according to a therapeutic schedule. The container that is employed will depend on the exact dosage form involved, for example a conventional cardboard box would not generally be used to hold a liquid suspension. It is feasible that more than one container can be used together in a single package to market a single dosage form. For example, tablets may be contained in a bottle that is in turn contained within a box. Typically the kit includes directions for the administration of the separate components. The kit form is particularly advantageous when the separate components are preferably administered in different dosage forms (e.g., oral, topical, transdermal and parenteral), are administered at different dosage intervals, or when titration of the individual components of the combination is desired by the prescribing physician.


One particular example of a kit according to the present invention is a so-called blister pack. Blister packs are well known in the packaging industry and are being widely used for the packaging of pharmaceutical unit dosage forms (tablets, capsules, and the like). Blister packs generally consist of a sheet of relatively stiff material covered with a foil of a preferably transparent plastic material. During the packaging process recesses are formed in the plastic foil. The recesses have the size and shape of individual tablets or capsules to be packed or may have the size and shape to accommodate multiple tablets and/or capsules to be packed. Next, the tablets or capsules are placed in the recesses accordingly and the sheet of relatively stiff material is sealed against the plastic foil at the face of the foil which is opposite from the direction in which the recesses were formed. As a result, the tablets or capsules are individually sealed or collectively sealed, as desired, in the recesses between the plastic foil and the sheet. Preferably the strength of the sheet is such that the tablets or capsules can be removed from the blister pack by manually applying pressure on the recesses whereby an opening is formed in the sheet at the place of the recess. The tablet or capsule can then be removed via said opening.


Another specific embodiment of a kit is a dispenser designed to dispense the daily doses one at a time in the order of their intended use. Preferably, the dispenser is equipped with a memory-aid, so as to further facilitate compliance with the regimen. An example of such a memory-aid is a mechanical counter that indicates the number of daily doses that has been dispensed. Another example of such a memory-aid is a battery-powered micro-chip memory coupled with a liquid crystal readout, or audible reminder signal which, for example, reads out the date that the last daily dose has been taken and/or reminds one when the next dose is to be taken.


EXAMPLES
Example 1
Preparation of 5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-n-(1-methylpiperidin-4-yl)-9h-pyrido[2,3-b]indole-7-carboxamide (Compound 1)









3-(6-chloro-3-methyl-2-nitro-4-(trifluoromethyl)phenyl)-2-fluoro-5-methylpyridine: 2-Fluoro-3-iodo-5-picoline (15.0 g, 63 mmol) was added drop wise during 2 h as a solution in NMP (20 mL) to a stirred suspension of 3,4-dichloro-2-nitro-6-(trifluoromethyl)-toluene (52.1 g, 190 mmol) and copper (12.1 g, 190 mmol) in NMP (115 mL) at 190° C. After completion of the reaction (2.5 h), the mixture was cooled to room temperature, filtered, rinsed with NMP (3×5 mL) followed by EtOAc (1×100 mL). The filtrate was diluted with EtOAc (400 mL) affording a turbid solution. The organic layer was partitioned with sat. NaHCO3 (150 mL) affording a suspension/emulsion. H2O (50 mL) and MeOH (50 mL) were added to aid solubility. The aqueous layer was washed with EtOAc (5×150 mL). The organic layers were combined, dried (MgSO4), and concentrated in vacuo. The crude product was purified by silica gel chromatography (98:2 Toluene:EtOAc) to provide the title compound as a tan solid (11.4 g, 52%). 1H NMR (400 MHz, DMSO-d6): δ 8.34 (s, 1H), 8.26 (s, 1H), 7.86-7.89 (m, 1H), 2.4 (s, 3H), 2.34 (s, 3H). MS (ES) [m+H] calc'd for C14H9ClF4N2O2, 349; found 349.2.


3-(3′-(ethylsulfonyl)-4-methyl-3-nitro-5-(trifluoromethyl)biphenyl-2-yl)-2-fluoro-5-methylpyridine: A mixture of Compound 83 (6.0 g, 17.2 mmol), 3-ethylsulfonylphenylboronic acid (4.79 g, 22.4 mmol), bis(dibenzylideneacetone)Pd(0) (1.48 g, 2.6 mmol), tricyclohexylphosphine (1.45 g, 5.2 mmol), Cs2CO3 (14.0 g, 43 mmol), and dioxane (60 mL) was heated at reflux for 4.5 hr. After completion the reaction was cooled to room temperature, filtered, rinsed with dioxane, and concentrated in vacuo. The resulting oil was reconstituted in EtOAc (75 mL) washed with H2O (1×30 mL) and brine (1×30 mL), dried (MgSO4), and concentrated in vacuo. The crude product was purified by silica gel chromatography (4:1 hexanes/EtOAc) to provide the title compound as a tan solid (6.5 g, 78%). 1H NMR (400 MHz, DMSO-d6): δ 8.15 (s, 1H), 8.04 (s, 1H), 7.90-7.93 (m, 1H), 7.80-7.82 (m, 1H), 7.60-7.70 (m, 3H), 3.1-3.2 (m, 2H), 2.49 (s, 3H), 2.25 (s, 3H), 0.85 (t, 3H). MS (ES) [m+H] calc'd for C22H18F4N2O4S, 483; found 483.3.


3′-(ethylsulfonyl)-2-(2-fluoro-5-methylpyridin-3-yl)-4-methyl-5-(trifluoromethyl)biphenyl-3-amine: A mixture of Compound 84 (6.4 g, 13.3 mmol), iron (3.7 g, 66.3 mmol), HOAc, (32 mL), and H2O (11 mL) was heated at 80° C. for 2 h. After completion the reaction was concentrated in vacuo. The residue was reconstituted in dichloromethane (100 mL), filtered, and rinsed with dichloromethane (3×30 mL). The organic phase was washed with sat. NaHCO3 (1×100 mL) and brine (1×50 mL), dried (MgSO4), filtered, and concentrated in vacuo. The crude product was purified by silica gel chromatography (1:1 hexanes/EtOAc) to provide the title compound as a tan solid (5.0 g, 83%). 1H NMR (400 MHz, DMSO-d6): δ 7.93 (s, 1H), 7.67-7.7.71 (m, 2H), 7.53 (t, 1H), 7.46-7.48 (m, 1H), 7.42 (s, 1H), 6.93 (s, 1H), 5.09 (s, 2H), 3.11 (q, 2H), 2.27 (s, 3H), 2.21 (s, 3H), 0.85 (t, 3H). MS (ES) [m+H] calc'd for C22H20F4N2O2S, 453; found 453.3.


5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-7-(trifluoromethyl)-9H-pyrido[2,3-b]indole acetate: Compound 85 (4.9 g, 10.8 mmol) was dissolved in HOAc (35 mL) and heated at reflux for 3 h. The reaction mixture was cooled to room temperature affording a crystalline product. The resulting suspension was filtered, rinsed with HOAc (3×5 mL) followed by H2O (3×10 mL) and the solids dried in vacuo to provide the title compound as a white solid (3.73 g, 70%). NMR analysis confirmed that the product was isolated as the mono-acetate salt. 1H NMR (400 MHz, DMSO-d6): δ 12.35 (s, 1H), 12.0 (s, 1H), 8.39 (s, 1H), 8.15 (s, 1H), 8.04-8.09 (m, 2H), 7.90 (t, 1H), 7.51 (s, 1H), 7.42 (s, 1H), 3.43 (q, 2H), 2.76 (s, 3H), 2.28 (s, 3H), 1.91 (s, 3H), 1.18 (t, 3H). MS (ES) [m+H] calc'd for C22H19F3N2O2S, 433; found 433.3.


5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-9H-pyrido[2,3-b]indole-7-carboxylic acid: Compound 86 (3.6 g, 7.3 mmol) was dissolved in concentrated H2SO4 (30 mL) and heated at 120° C. for 30 min. The reaction was cooled to room temperature and poured over ice affording a white precipitate. The resulting suspension was filtered, rinsed with H2O (3×30 mL) followed by IPA (3×10 mL) and dried in vacuo to provide the title compound as a white solid (3.2 g, quant.). 1H NMR (400 MHz, DMSO-d6): δ 12.20 (s, 1H), 8.36 (s, 1H), 8.12 (s, 1H), 8.02-8.07 (m, 2H), 7.89 (t, 1H), 7.61 (s, 1H), 7.54 (s, 1H), 3.43 (q, 2H), 2.85 (s, 3H), 2.28 (s, 3H), 1.18 (t, 3H). MS (ES) [m+H] calc'd for C22H20N2O4S, 409; found 409.3.


5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-N-(1-methylpiperidin-4-yl)-9H-pyrido[2,3-b]indole-7-carboxamide: A mixture of Compound 87 (11.3 g, 27.6 mmol), 1-methylpiperidin-4-amine (9.47 g, 82.9 mmol), HATU (13.66 g, 35.9 mmol), DIEA (17.88 g, 138 mmol), DMF (250 mL), and DCM (250 mL) was stirred at room temperature for 30 minutes. The resulting suspension was filtered, rinsed with DMF (10 mL×4) and concentrated in vacuo. The residue was dissolved in DMSO (77 mL), filtered, and the filtrate was purified by preparative HPLC (ACN/H2O with TFA). Following HPLC purification, the pure fractions were combined, basified with sodium bicarbonate and concentrated in vacuo to half volume. The resulting suspension was filtered, rinsed with H2O (200 mL×5) and dried in vacuo to provide Compound 88 as a white solid (11.41 g, 81.8%).


The hydrochloride salt of Compound 88 was prepared as follows. To a stirred suspension of Compound 88 (8.7 g) in ACN (175 mL) and H2O (175 mL) was added 1N HCl (18.1 mL, 1.05 eq) affording a yellow solution. After 15 minutes, the solution was frozen on dry ice/acetone and lyophilized to provide 5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-N-(1-methylpiperidin-4-yl)-9H-pyrido[2,3-b]indole-7-carboxamide hydrochloride as a yellow solid (9.02 g, 96.7%). The above process provided 5-(3-(ethylsulfonyl)phenyl)-3,8-dimethyl-N-(1-methylpiperidin-4-yl)-9H-pyrido[2,3-b]indole-7-carboxamide hydrochloride as the Amorphous Form, as determined by X-ray powder diffraction analysis (FIG. 1).


Example 2
Sample Characterization

The following analytical techniques and combination thereof were used determine the physical properties of the solid phases prepared.


1. Instrumentation














Instrument
Vendor/Model #
AMRI #







Differential Scanning
Mettler 822e DSC
10667


Calorimeter


Thermal Gravimetric
Mettler 851e SDTA/TGA
009111


Analyzer


X-Ray Powder Diffraction
Shimadzu XRD-6000
009856


System


Karl Fischer
Metrohm 756 KF Coulometer
005966


Nuclear Magnetic
500 MHz Bruker AVANCE
BH-003201


Resonance Spectrometer
with 5-mm BBO probe


Gas Chromatograph
Agilent HP 6890 Gas
GC #9


equipped with a Headspace
Chromatograph equipped with


Sampler
HP 7694 Headspace Sampler


Ion Chromatography
Dionex DX600 Ion
IC #1



Chromatograph


High-Performance Liquid
Varian ProStar
008956


Chromatography


Moisture-Sorption Analysis
Hiden IGAsorp Moisture
IGASA030



Sorption Instrument









2. Differential Scanning Calorimetry Analysis (DSC)


Differential scanning calorimetry (DSC) analyses were carried out on samples weighed in an aluminum pan, covered with a pierced lid, and then crimped. Analysis conditions were 30° C. to 350° C. ramped at 10° C./min.


3. Thermal Gravimetric Analysis (TGA)


Thermal gravimetric analysis (TGA) analyses were carried out on samples weighed in an alumina crucible and analyzed from 30° C. to 230 or 250° C. and at a ramp rate of 10° C./min.


4. X-Ray Powder Diffraction (XRPD)


Samples for X-ray powder diffraction (XRPD) were placed on Si zero-return ultra-micro sample holders and analyzed using the following conditions:


















X-ray tube:
Cu Kα, 40 kV, 40 mA



Slits



Divergence Slit
1.00 deg



Scatter Slit
1.00 deg



Receiving Slit
0.30 mm



Scanning



Scan Range
3.0-45.0 deg



Scan Mode
Continuous



Step Size
0.04°



Scan Rate
2°/min










5. Karl Fischer Analysis (KF)


Water content was determined by adding solid sample to the instrument with HYDRANAL-Coulomat AD. Micrograms of water were determined by coulometric titration.


6. Moisture-Sorption Analysis


Moisture-sorption experiments were carried out on three forms by first drying the sample at 0% RH and 25° C. until an equilibrium weight was reached or for a maximum of four hours. The sample was then subjected to an isothermal (25° C.) adsorption scan from 10 to 90% RH in steps of 10%. The sample was allowed to equilibrate to an asymptotic weight at each point for a maximum of four hours. Following adsorption, a desorption scan from 85 to 0% RH (at 25° C.) was run in steps of −10%, again allowing a maximum of four hours for equilibration to an asymptotic weight. The sample was then dried for one hour at 80° C. and the resulting solid analyzed by XRPD.


7. Nuclear Magnetic Resonance (NMR)


Samples (2 to 10 mg) were dissolved in DMSO-d6 with 0.05% tetramethylsilane (TMS) for internal reference. 1H-NMR spectra were acquired at 500 MHz using 5 mm broadband observe (1H—X) Z gradient probe. A 30 degree pulse with 20 ppm spectral width, 1.0 s repetition rate, and 16 to 64 transients were utilized in acquiring the spectra.


8. Organic Volatile Impurities (OVI)


Approximately 100 mg of sample was weighed into an individual 20-mL headspace vial and 5 mL of DMSO added. The vial was then sealed and gentle shaking/vortexing was used to ensure sample was entirely dissolved. Blank samples were prepared by transferring 5.0 mL of DMSO into a 20-mL headspace vial and then sealed. Standards were prepared using stock solutions in DMSO.


Instrument Parameters were as follows:















Column:
DB-1, 60 meter × 0.32 mm (inside diameter),



3-μm film thickness, P/N: 123-1064


Detector:
FID; hydrogen flow of 40 mL/min, air flow of



450 mL/min. Makeup gas (helium) flow of



30 mL/min.


Carrier Gas:
Helium


Carrier Flow:
2.2 mL/min


Oven Temperature:
40° C. isothermal held for 5 minutes; ramp at



5° C./minute to



105° C.; ramp at 10° C./minute to 165° C.; ramp



at 20° C./ minute to 245° C.; hold for 2 minutes


Injector Temperature:
140° C.


Detector Temperature:
260° C.


Injection Type:
Split


Split Flow:
25 mL/minute (includes flow contributed by the



headspace sampler)


Injection Volume:
1 mL (Headspace)


Analysis Time:
30 minutes









Headspace Sampler Conditions were as follows:
















Parameter
Setting




















Oven Temperature:
80°
C.



Loop Temperature:
100°
C.



Transfer Line Temperature:
110°
C.



GC Cycle Time:
40
minutes



Vial Equilibration Time:
20
minutes



Pressurization Time:
0.13
minutes



Loop Fill Time:
0.06
minutes



Loop Equilibration Time:
0.06
min



Injection Time:
0.20
minutes



Carrier Flow:
20
mL/min



Vial Pressurization:
10.0
psi



Shake:
2
(high)










9. Ion Chromatography (IC)


Sample solutions in DI water were prepared with a concentration of 0.1 mg/mL. IC was performed utilizing the following conditions:















Instrument:
Dionex DX600 Ion Chromatograph


AMRI System #:
1


Column:
Dionex IonPac AS17, 250 × 4 mm


Guard Column:
Dionex IonPac AS17, 50 × 4 mm


Column Temperature:
35 ± 2° C.


Detector Operating Mode:
Suppressed Conductivity


Suppressor Type:
Dionex ASRS Ultra 4 mm


Suppressor Current:
220 mA


Mobile Phase A:
Purified Water


Mobile Phase B:
Potassium Hydroxide, delivered using an



Eluent Generator


Gradient:
See table below.


Flow Rate:
1.5 mL/minute


Injection Volume:
10 μL


Needle Wash:
Purified Water


Diluent:
Purified Water









Gradient Conditions were as follows:














Time
Mobile
Concentration


(minutes)
Phase A
of KOH (mM)

















0.0
100%
5


3.0
100%
5


10.0
100%
15


20.0
100%
60


20.1
100%
5


30.0
100%
5









10. High Performance Liquid Chromatography (HPLC)


Equipment used was an HPLC system equipped with a UV detector, gradient capabilities, and electronic data collection and processing, or equivalent, an autosampler capable of 10 μL injection, an analytical Column: Waters X-Terra RP18, 4.6×150 mm, 3.5 μm, P/N 186000442, an analytical balance capable of weighing to ±0.01 mg, and class A volumetric pipettes and flasks


The instrument parameters were as follows:















Column:
Waters X-Terra RP18, 4.6 × 150 mm, 3.5 μm


Column
45 ± 2° C.


Temperature:


Auto-sampler
Ambient


Temperature:


Detection:
225 nm


Mobile Phase A:
0.05% TFA in Water


Mobile Phase B:
0.04% TFA in Acetonitrile


Gradient:
See table below


Flow Rate:
1.0 mL/minute


Injection Volume:
10 μL


Analysis Time:
38 minutes


Re-equilibration Time:
8 minutes


Data Collection time:
30 minutes


Needle Wash:
50:50 Acetonitrile/Water









Gradient Conditions were as follows:














Time (minutes)
% A
% B

















0.0
95
5


15.0
75
25


30.0
5
95


30.1
95
5


38.0
95
5









Example 3
Solvent Screen

A solubility study of Compound 1 in various solvents was executed to select appropriate solvents for the further crystallizations. The material was placed in vials and the solvent was added in 250 μL portions. Solvents were picked based on differences in polarity and functionality and on their classification according to the International Conference on Harmonization (ICH), with preferences given to class II and class III solvents. After each addition of solvent, the vials were visually inspected for residual solids, and further heating to 55° C. to ensure dissolution. Table 16 shows the solvents that were used and their ability to dissolve the material at room temperature.









TABLE 16







Solubility Screen of Compound 1














Material








Amt
Solvent Amt
Conc.


ICH


Solvent
(mg)
(mL)
(mg/mL)
Temp
Soluble
Class
















MeCN
2.4
5.25
0.5
55
No
II


Dioxane
1.1
1.50
0.7
55
Yes
II


Acetone
1.4
5.25
0.3
55
No
III


MTBE
1.2
4.75
0.3
55
No
III


EtOH
1.8
1.50
1.2
55
Yes
III


EtOAc
2.0
5.25
0.4
55
No
III


IPAC
2.2
4.75
0.5
55
No
III


IPA
2.3
4.75
0.5
55
No
III


THF
1.8
4.75
0.4
55
No
II


2-Me-THF
2.1
4.75
0.4
55
No
N/A


MEK
2.8
4.75
0.6
55
No
III


DMF
2.6
0.25
>10.4
RT
Yes
II


AcOH
2.3
0.25
>9.2
RT
Yes
III


MeOH
2.5
0.25
>10.0
RT
Yes
II


c-Hexane
3.1
6.00
0.5
55
No
II


Heptane
2.4
6.00
0.4
55
No
III


DCM
1.9
2.25
0.8
RT
No
II


Toluene
2.0
4.75
0.4
55
No
II


water
1.7
2.75
0.6
55
No
N/A


NMP
2.6
0.25
>10.4
RT
Yes
II


DMA
2.3
0.25
>9.2
RT
Yes
II


Chloroform
2.1
0.25
>8.4
RT
Yes
II









Example 4
Primary and Binary Solvent Efficiency Studies with Compound 1

Solvent efficiency experiments for Compound 1 were carried out by charging the free base version of Compound 1 (15-16 mg) to an 8-Dram clear vial equipped with magnetic stir bar. Seven primary solvents (MeCN, EtOH, THF, DMA, NMP, AcOH, and DMF) were chosen based on initial solubility data obtained during the solvent screen (Table 16) and added in 100 μL portions until complete dissolution was observed with heating to 50° C. Once complete dissolution was observed HCl was added as a 1M solution (1.05 equiv.) in the reaction solvents at elevated temperatures. The resulting mixtures were then allowed to stir at that temperature for approximately 15 minutes. Four anti-solvents (MtBE, EtOAc, IPAc, and heptane) were chosen based on solubility data and added in one vol. portions at elevated temperatures until a turbid mixture was observed. Each sample was then allowed to cool to ambient temperature at a rate of 20° C./h with further stirring for 16 hours. Solids were isolated by filtration and dried under vacuum at ambient temperature for 16 hours. All samples were analyzed by XRPD with the results outlined in Tables 17-18.









TABLE 17







Solvent efficiency evaluation of Compound 1 using DMA















Amt
DMA Amt


Amt Anti-

Amt




Material
Solvent/Vol
Dissolution
Anti-
Solvent/Vol
Method of
Recovered
%
Form


(mg)
(mL)
Temp [° C.]
Solvent
(mL)
Isolation
(mg)
Yield
(XRPD)





50.73
0.300/6
n/a


n/a
n/a
n/a
n/a


50.06
0.400/8
41.5


filtration
15.70
29.2
B


51.36
0.500/10
29.9


filtration
6.24
11.3
A


50.91
0.600/12
29.8


filtration
7.25
13.2
C


50.39
0.700/14
30.8


evap.
n/a
n/a
mix


50.30
0.500/10
41.0
MTBE
0.500/10
filtration
28.46
52.8
B


50.78
0.500/10
41.0
MTBE
0.250/5
filtration
26.53
48.7
D


50.31
0.400/8
41.0
MTBE
0.200/4
filtration
10.02
18.6
mix


52.77
0.450/9
41.0
MTBE
0.100/2
filtration
12.66
22.4
B


49.60
0.400/8
41.0
MTBE
0.100/2
filtration
18.38
34.5
B
















TABLE 18







Solvent efficiency evaluation of Compound 1 using several organic solvents



















Amt







Amt
Dissolution

Anti-Solvent

Amt



Solvent Vol
Temp
Anti-
Vol
Method of
Recovered

Form


Solvent
(mL/vol.)
[° C.]
Solvent
(mL/Vol)
Isolation
(mg)
% Yield
(XRPD)





MeCN
2.25/150
n/a
n/a
n/a
n/a
n/a
n/a
n/a


EtOH
2.25/150
n/a
n/a
n/a
n/a
n/a
n/a
n/a


THF
2.25/150
n/a
n/a
n/a
n/a
n/a
n/a
n/a


DMAc
0.150/10
50


n/a
n/a
n/a
n/a


DMAc
0.150/10
50
MTBE
0.375/25
filtered
4.1
26
D


DMAc
0.150/10
50
EtOAc
0.150/10
filtered
2.8
17
C


DMAc
0.150/10
50
IPAc
0.150/10
filtered
1.9
12
B


DMAc
0.150/10
50


n/a
n/a
n/a
n/a


NMP
0.075/5
35


filtered
13.7 
78
E


NMP
0.075/5
35
MTBE
0.075/5 
filtered
2.0
11
E


NMP
0.075/5
35
EtOAc
0.075/5 
filtered
9.0
55
E


NMP
0.075/5
35
IPAc
0.075/5 
filtered
8.5
55
E


NMP
0.075/5
35
heptane
0.075/5 
filtered
2.3
13
E


AcOH
0.075/10
25


n/a
n/a
n/a
n/a


AcOH
0.075/5
25
MTBE
0.150/10
n/a
n/a
n/a
n/a


AcOH
0.075/5
25
EtOAc
0.150/10
n/a
n/a
n/a
n/a


AcOH
0.075/5
25
IPAc
0.150/10
n/a
n/a
n/a
n/a


AcOH
0.075/5
25
heptane
0.450/30
n/a
n/a
n/a
n/a


DMF
0.225/15
50


filtered
2.0
13
A


DMF
0.225/15
50


filtered
4.9
28
A


DMF
0.225/15
50
MTBE
0.375/25
filtered
3.1
18
A


DMF
0.225/15
50
EtOAc
0.200/13
filtered
4.7
30
A


DMF
0.225/15
50
IPAC
0.200/13
filtered
7.7
44
A









Example 5
Preparation of Compound 1 from DMF/IPAc

Preparation of Compound 1 was carried out in a 250 mL 3N-RBF equipped with magnetic stir bar and thermocouple. To this was added a free base version of Compound 1 starting material (5.05 g, 0.10 mol.) followed by the portion wise (approximately 5 mL) addition of DMF (50 mL, 10 vol.) with heating to 65° C. Once complete dissolution was observed the HCl counter ion was added as a 1M solution (10.49 mL, 1.05 equiv.) in DMF at 65° C., and the resultant mixture allowed to stir for 15 min. The reaction mixture was then allowed to cool to 55° C. at a rate of 20° C./h. Once an internal temperature of 55° C. had been achieved IPAc (50 mL, 10 vol.) was added as an anti-solvent in a dropwise fashion over a 30 minute period. The reaction mixture was then further cooled to ambient temperature at the same rate (20° C./h) followed by further cooling to 0° C. with an ice/water bath. A light precipitate was observed at 30° C., and the resultant slurry was allowed to continue stirring at 0° C. for an additional 4 hours. The solids were then isolated by filtration and the filter cake dried under vacuum at 40° C. for 16 hours to give Compound 1 (3.19 g, 59% yield of Form G) as a light tan crystalline solid.


Example 6
Single-Solvent Crystallizations

Using the initial solubility study (Table 16) and the methods outlined below, six solvents were selected for the single solvent crystallization: MeOH, EtOH, AcOH, DMF, DMA and NMP. All solids isolated were analyzed by XRPD to determine the physical form. Table 19 shows a list of the solvents that were used and the amount of solvent needed to dissolve the material in the fast cooling procedure and Table 20 shows the same information for the slow cooling procedure. Solutions of Compound 1 in acetic acid did not form a precipitate under either slow or fast cooling conditions. Both samples were evaporated to dryness and afforded amorphous materials. XRPD analysis of non-amorphous solids showed patterns consistent with Forms A, B, C, E, G, and mixtures of Forms C and P (originally designated as Form H) as shown in Tables 19 and 20.


1. Fast-Cooling Profile


Using the initial solvent screen six solvents were selected for the single solvent crystallization: MeOH, EtOH, AcOH, DMF, DMA and NMP. Compound 1 (˜20 mg) was weighed out into vials and enough solvent (starting with 0.25 mL) was added until the material completely dissolved at elevated temperature. After hot filtration the vials were placed in a refrigerator (4° C.) for 16 hours. The resultant solids were isolated by vacuum filtration. The samples without solids were evaporated to dryness using a gentle stream of nitrogen.


All resultant solids from filtration and evaporation were dried in vacuo at room temperature and 30 inches Hg for 16 hours. All solids were analyzed by XRPD to determine the physical form. Table 19 shows a list of the solvents that were used and the amount of solvent needed to dissolve the material in the fast cooling procedure.









TABLE 19







Single solvent crystallizations of Compound 1 using fast cooling procedure















Compound 1


Temp.







Amt (mg)
Solvent
Amt (mL)
(° C.)
Cooling
Precipitation
Recovery (mg)
Recovery (%)
Form





20.2
MeOH
0.75
60
Fast
Yes
 9.0
44.6
C + P


20.3
EtOH
4.00
75
Fast
Yes
12.6
62.1
C + P


20.4
AcOH
0.25
80
Fast
No
n/a
n/a
amorph


20.9
DMF
0.25
70
Fast
Yes
10.7
51.2
A


20.7
DMA
0.25
70
Fast
Yes
14.1
68.1
B


20.0
NMP
0.25
70
Fast
Yes
11.7
58.5
E





Amorph = amorphous













TABLE 20







Single solvent crystallizations of Compound 1 using slow cooling procedure















Compound 1


Temp







Amt (mg)
Solvent
Amt (mL)
(° C.)
Cooling
Precipitation
Recovery (mg)
Recovery (%)
Form


















19.9
MeOH
0.5
60
Slow
Yes
11.0
55.3
C


20.3
EtOH
3.00
75
Slow
Yes
13.8
68.0
C


20.9
AcOH
0.25
80
Slow
No/evap
n/a
n/a
amorph


19.9
DMF
0.25
80
Slow
Yes
 9.2
46.2
G


19.6
DMA
0.25
80
Slow
Yes
15.8
80.6
B


19.8
NMP
0.25
80
Slow
Yes
18.6
93.9
E





Amorph = amorphous






2. Slow-Cooling Profile


Using the initial solubility study six solvents were selected for the single solvent crystallization: MeOH, EtOH, AcOH, DMF, DMA and NMP. Compound 1 (approximately 20 mg) was weighed out into vials and enough solvent (starting with 0.25 mL) was added until the material completely dissolved at elevated temperature. After hot filtration the vials were slowly cooled to the room temperature at the rate of 20° C./h and stirred at this temperature for 16 hours. The resultant solids were isolated by vacuum filtration. The samples without solids were evaporated to dryness using a gentle stream of nitrogen. All resultant solids from filtration and evaporation were dried in vacuo at room temperature and 30 inches Hg for 16 hours. All solids were analyzed by XRPD to determine the physical form. Table 20 shows a list of the solvents that were used and the amount of solvent needed to dissolve the material.


Example 7
Binary-Solvent Crystallizations

Using the methods described below, binary solvent crystallizations were performed using MeOH, EtOH, AcOH, DMF, DMA and NMP as primary solvents. All obtained solids were analyzed by XRPD to determine the physical form. A summary of the experimental details for the fast and slow cooling experiments is presented in Tables 21 through 32.


All solids obtained from single and binary solvent crystallizations with fast and slow cooling procedures were analyzed by XRPD to determine the physical form. When either of the two known forms of the freebase (A or B) was observed, it was labeled as FB (A) or FB (B) respectively. For samples affording unique XRPD patterns, further analysis was performed on representative lots including: IC to determine counter-ion content, 1H NMR to determine residual solvent content and confirm degradation did not occur, and thermal analysis (DSC and TGA) to characterize thermal events. Tables 36a and 36b summarize characterization data for all forms discovered in this screen. Forms A, C, and L were found to be the most common non-solvated forms observed during the screen. These materials were used for slurry, moisture sorption, and humidity chamber studies.


Water was found to be a poor crystallization anti-solvent as it afforded a free base version of Compound 1 consistently except when acetic acid was used as the primary solvent. Acetic acid was found to be a poor crystallization solvent as it often afforded amorphous solids or sticky solids which were not analyzable. Non-amorphous solids from acetic acid still showed the presence of an amorphous halo suggesting the materials were semi-crystalline, which made definitive form assignment difficult.


1. Fast-Cooling Profile


Compound 1 (approximately 20 mg) was weighed out into vials and enough primary solvent was added until the material went into solution at elevated temperature. After hot filtration antisolvent was added portion wise until the solution became turbid or the vial became full. The vials were then placed in a refrigerator (4° C.) for 16 hours. Tables 21, 23, 25, 27, 29 and 31 show experimental details. After the cooling process precipitates were isolated by filtration, they were dried in vacuo at room temperature and 30 in Hg. The vials without solids were evaporated down to dryness using a gentle stream of nitrogen and also dried in vacuo at ambient temperature and 30 in Hg. All solids were analyzed by XRPD.


2. Slow-Cooling Profile


Compound 1 (approximately 20 mg) was weighed out into vials and enough primary solvent was added until the material went into solution at elevated temperature. After hot filtration antisolvent was added portion wise until the solution became turbid or the vial became full according to the data obtained from fast-cooling experiments. The vials were then slowly cooled to room temperature at the rate of 30° C./h. Tables 22, 24, 26, 28, 30 and 32 show experimental details. After the cooling process precipitates were isolated by filtration, they were dried in vacuo at room temperature and 30 in Hg. The vials without solids were evaporated down to dryness using a gentle stream of nitrogen and also dried in vacuo at ambient temperature and 30 in Hg. All solids were analyzed by XRPD.









TABLE 21







Binary solvent crystallizations of Compound 1 using fast cooling procedure and


MeOH as primary solvent
















Compound
MeOH
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















19.3
0.5
MeCN
6.00
60
Fast
clear/ppt
10.3
53.4
C + P


19.6
0.5
MTBE
0.47
60
Fast
turbid/ppt
10.6
54.1
C + P


20.5
0.5
EtOAc
2.00
60
Fast
turbid/ppt
13.8
67.3
C + P


20
0.5
IPAc
1.40
60
Fast
turbid/ppt
14.1
70.5
C + P


19.8
0.5
IPA
2.00
60
Fast
turbid/ppt
13.1
66.2
A


21.0
0.5
THF
6.00
60
Fast
clear/ppt
13.5
64.3
I


21.3
0.5
MEK
3.80
60
Fast
turbid/ppt
14.1
66.2
A + C + P


20.9
0.5
Heptane
1.00
60
Fast
2 layers/ppt
10.5
50.2
C + P


20.3
0.5
Water
6.00
60
Fast
clear/ppt
1.2
5.9
FB(A)










FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.









TABLE 22







Binary solvent crystallizations of Compound 1 using slow cooling procedure and


MeOH as primary solvent
















Compound
MeOH
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















21.5
0.5
MeCN
6.0
60
Slow
clear/ppt
14.2
66.0
C


20.5
0.5
MTBE
0.5
60
Slow
turbid/ppt
12.6
61.5
L


19.6
0.5
EtOAc
2.0
60
Slow
turbid/ppt
12.8
65.3
C + P


20.7
0.5
IPAc
1.4
60
Slow
turbid/ppt
14.2
68.6
L


20.3
0.5
IPA
2.0
60
Slow
turbid/ppt
13.3
65.5
A


19.8
0.5
THF
6.0
60
Slow
clear/ppt
8.4
42.4
A


20.8
0.5
MEK
4.0
60
Slow
turbid/ppt
14.5
69.7
A


19.7
0.5
Heptane
1.0
60
Slow
2 layers/ppt
9.0
45.7
L


20.5
0.5
Water
6.0
60
Slow
clear/ppt
2.0
9.8
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 23







Binary solvent crystallizations of Compound 1 using fast cooling procedure and


EtOH as primary solvent
















Compound
EtOH
Anti
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















21.9
4.0
MeCN
15
75
Fast
clear/no ppt
10.8
49.3
C + P


20.7
3.0
MTBE
7
70
Fast
turbid/ppt
17
82.1
J


21.5
3.0
EtOAc
15
70
Fast
clear/ppt
14.3
66.5
A + C + P


20.5
3.0
IPAc
15
70
Fast
clear/ppt
14.2
69.3
A + C + P


20.6
3.0
IPA
15
70
Fast
clear/ppt
15.4
74.8
J


20.7
3.0
THF
15
70
Fast
clear/no ppt
9.6
46.4
K


20.1
3.0
MEK
15
70
Fast
clear/ppt
10.5
52.2
A


20.3
3.0
Heptane
5
70
Fast
turbid/ppt
16.8
82.8
C + P


21.9
3.0
Water
15
70
Fast
clear/ppt
4.3
19.6
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 24







Binary solvent crystallizations of Compound 1 using slow cooling procedure and


EtOH as primary solvent
















Compound
EtOH
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















20.2
3.0
MeCN
15
70
Slow
clear/no ppt
10.8
53.5
C + P


19.6
3.0
MTBE
7
70
Slow
turbid/ppt
16.2
82.7
L


19.9
3.0
EtOAc
15
70
Slow
clear/ppt
14
70.4
C + P


21.3
3.0
IPAc
15
70
Slow
clear/ppt
17.1
80.3
A


20.8
3.0
IPA
15
70
Slow
clear/ppt
11.6
55.8
C + P


20.2
3.0
THF
15
70
Slow
clear/ppt
5.6
27.7
C + P


21.5
3.0
MEK
15
70
Slow
clear/ppt
14.1
65.6
A


20.4
3.0
Heptane
5
70
Slow
turbid/ppt
16.7
81.9
A + C + P


20.7
3.0
Water
15
70
Slow
clear/ppt
4.0
19.3
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 25







Binary solvent crystallizations of Compound 1 using fast cooling procedure and


AcOH as primary solvent
















Compound
AcOH
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















19
0.25
MeCN
7.00
70
Fast
Clear/No
n/a
n/a
amorph


19.8
0.25
MTBE
0.25
70
Fast
Turbid/Yes
4.5
22.7
amorph


19.5
0.25
EtOAc
6.00
70
Fast
Turbid/Yes
8.8
45.1
amorph


19.1
0.25
IPAc
1.35
70
Fast
Turbid/Yes
3.7
19.4
amorph


20.7
0.25
IPA
7.00
70
Fast
Clear/Yes
9.7
46.9
C + P + J


19.3
0.25
THF
7.00
70
Fast
Clear/Yes
12.3
63.7
I


19.8
0.25
MEK
7.00
70
Fast
Clear/Yes
8.9
44.9
L


20.6
0.25
Heptane
0.30
70
Fast
2 layers/No
n/a
n/a
amorph


19.8
0.25
Water
7.00
70
Fast
Clear/No
n/a
n/a
n/a





Amorph = amorphous


n/a Indicates the sample was not an isolatable sol













TABLE 26







Binary solvent crystallizations of Compound 1 using slow cooling procedure and


AcOH as primary solvent
















Compound
AcOH
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















19.7
0.25
MeCN
7.00
70
Slow
Clear/Yes
9.5
48.2
C


20.1
0.25
MTBE
0.30
70
Slow
Turbid/Yes
9.9
49.3
J


20.5
0.25
EtOAc
6.00
70
Slow
Turbid/Yes
13
63.4
J


21.2
0.25
IPAc
1.35
70
Slow
Turbid/Yes
12.5
59.0
J


21
0.25
IPA
7.00
70
Slow
Clear/Yes
11.2
53.3
J w/ add


20.0
0.25
THF
7.00
70
Slow
Clear/Yes
11.7
58.5
J w/ add


20.6
0.25
MEK
7.00
70
Slow
Clear/Yes
12.4
60.2
L


19.7
0.25
Heptane
0.20
70
Slow
2 layers/No
n/a
n/a
amorph


20.0
0.25
Water
7.00
70
Slow
Clear/No
n/a
n/a
n/a





Amorph = amorphous


J w/add = Form J with additional peaks present. Due to the semi-crystalline nature of the material definitive determination was not possible.


n/a Indicates the sample was not an isolatable solid













TABLE 27







Binary solvent crystallizations of Compound 1 using fast cooling procedure and


DMF as primary solvent
















Compound
DMF
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















20.2
0.25
MeCN
0.77
70
Fast
Turbid/Yes
16.1
79.7
A


21.2
0.25
MTBE
0.40
70
Fast
Turbid/Yes
12.8
60.4
A


19.2
0.25
EtOAc
0.38
70
Fast
Turbid/Yes
13.0
67.7
A


19.8
0.25
IPAc
0.20
70
Fast
Turbid/Yes
15.7
79.3
G + A


19.7
0.25
IPA
0.92
70
Fast
Turbid/Yes
13.8
70.1
A


20.9
0.25
THF
0.97
70
Fast
Turbid/Yes
19.1
91.4
I


20.6
0.25
MEK
0.63
70
Fast
Turbid/Yes
17.2
83.5
G + A


19.6
0.25
Heptane
1.00
70
Fast
2 layers/yes
11.0
56.1
G


20.6
0.25
Water
6.00
70
Fast
Turbid/Small
1.4
6.8
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 28







Binary solvent crystallizations of Compound 1 using slow cooling procedure and


DMF as primary solvent
















Compound
DMF
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















20.9
0.25
MeCN
0.60
70
Slow
Turbid/Yes
16.1
77.0
C


19.7
0.25
MTBE
0.39
70
Slow
Turbid/Yes
12.8
65.0
A + G


19.1
0.25
EtOAc
0.50
70
Slow
Turbid/Yes
13.0
68.1
A


20.5
0.25
IPAc
0.15
70
Slow
Turbid/Yes
15.7
76.6
A + G


20.4
0.25
IPA
0.96
70
Slow
Turbid/Yes
13.8
67.6
C + P


19.6
0.25
THF
1.00
70
Slow
Turbid/Yes
19.1
97.4
I


20.3
0.25
MEK
0.82
70
Slow
Turbid/Yes
17.2
84.7
G


20.9
0.25
Heptane
1.00
70
Slow
2 layers/Yes
11.0
52.6
G


20.0
0.25
Water
6.00
70
Slow
Turbid/Small
1.4
7.0
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 29







Binary solvent crystallizations of Compound 1 using fast cooling procedure and


DMA as primary solvent
















Compound
DMA
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















19.5
0.25
MeCN
0.60
70
Fast
Turbid/Yes
15.7
80.5
A


20.7
0.25
MTBE
0.30
70
Fast
Turbid/Yes
16
77.3
B


20.2
0.25
EtOAc
0.45
70
Fast
Turbid/Yes
17.1
84.7
B


19.9
0.25
IPAc
0.32
70
Fast
Turbid/Yes
17.8
89.4
B


19.9
0.25
IPA
0.60
70
Fast
Turbid/Yes
15.6
78.4
A


19.5
0.25
THF
0.60
70
Fast
Turbid/Yes
16.8
86.2
B + A


20.8
0.25
MEK
0.65
70
Fast
Turbid/Yes
17.8
85.6
B + A


20.1
0.25
Heptane
0.22
70
Fast
2 layers/Yes
14.8
73.6
B


20.0
0.25
Water
6.00
70
Fast
Clear/Small
3.6
18.0
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 30







Binary solvent crystallizations of Compound 1 using slow cooling procedure and


DMA as primary solvent
















Compound
DMA
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















19.7
0.25
MeCN
0.51
70
Slow
Turbid/Yes
10.0
50.8
C


19.2
0.25
MTBE
0.27
70
Slow
Turbid/Yes
16.5
85.9
B + A


20.7
0.25
EtOAc
0.45
70
Slow
Turbid/Yes
18.9
91.3
B + A


20.5
0.25
IPAc
0.35
70
Slow
Turbid/Yes
17.9
87.3
B + A


20.4
0.25
IPA
0.72
70
Slow
Turbid/Yes
16.1
78.9
A


20.4
0.25
THF
0.75
70
Slow
Turbid/Yes
19.2
94.1
B + A


20.5
0.25
MEK
0.73
70
Slow
Turbid/Yes
18.9
92.2
B + A


20.5
0.25
Heptane
0.25
70
Slow
2 layers/Yes
14.8
72.2
B


20.6
0.25
Water
6.00
70
Slow
Clear/Small
1.0
4.9
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 31







Binary solvent crystallizations of Compound 1 using fast cooling procedure and


NMP as primary solvent
















Compound
NMP
Anti-
Amt
Temp.


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















19.4
0.25
MeCN
1.00
70
Fast
Turbid/Yes
17.7
91.2
E


20
0.25
MTBE
0.43
70
Fast
Turbid/Yes
16.1
80.5
E


20.6
0.25
EtOAc
0.65
70
Fast
Turbid/Yes
18.3
88.8
E


20.5
0.25
IPAc
0.50
70
Fast
Turbid/Yes
19.4
94.6
E


19.8
0.25
IPA
6.00
70
Fast
Clear/Yes
15.1
76.3
A


19.2
0.25
THF
2.00
70
Fast
Turbid/Yes
14.9
77.6
E


20.8
0.25
MEK
1.00
70
Fast
Turbid/Yes
19.4
93.3
E


20.7
0.25
Heptane
0.70
70
Fast
2 layers/Yes
18.0
87.0
E


20.9
0.25
Water
6.00
70
Fast
Clear/Small
<1
n/a
n/a





N/A - sample was not analyzable.













TABLE 32







Binary solvent crystallizations of Compound 1 using slow cooling procedure and


NMP as primary solvent
















Compound
NMP
Anti-
Amt
Temp


Recovery
Recovery



1 (mg)
(mL)
Solvent
(mL)
(° C.)
Cooling
Precipitation
(mg)
(%)
Form



















20.3
0.25
MeCN
1.00
70
Slow
Turbid/Yes
18.8
92.6
E


19.8
0.25
MTBE
0.42
70
Slow
Turbid/Yes
18.6
93.9
E


19.2
0.25
EtOAc
0.67
70
Slow
Turbid/Yes
18.7
97.4
E


20.3
0.25
IPAc
0.50
70
Slow
Turbid/Yes
18.5
91.1
E


20.2
0.25
IPA
6.00
70
Slow
Clear/Yes
16.2
80.2
A


19.2
0.25
THF
2.00
70
Slow
Turbid/Yes
19.6
102.1
I


19.4
0.25
MEK
1.00
70
Slow
Turbid/Yes
18.8
96.9
E


20.9
0.25
Heptane
0.70
70
Slow
2 layers/Yes
15.8
75.6
E


20.7
0.25
Water
6.00
70
Slow
Clear/Small
1.7
8.2
FB(A)





FB(A) indicates the pattern is consistent with free base a free base versions of Compound 1.













TABLE 33







Details of scale-up experiments of selected forms
















Targeted
Compound

Amount
Anti-
Amount
Temp

Recovery
Obtained


Form
1 (mg)
Solvent
(mL)
Solvent
(mL)
(° C.)
Cooling
(%)
Form



















A
300
DMA
3.60
IPA
10.50
70
Slow
95.3
N


C
301
DMF
3.60
MeCN
10.00
70
Slow
91.7
L


H*
301
MeOH
9.50
EtOAc
30.00
60
Slow
75.7
N


A
400
MeOH
10
IPA
40
60
Slow
84.00
C + P*


C
400
MeOH
10
MeCN
112
60
Slow
76.05
C


H*
400
EtOH
60
EtOAc
300
70
Slow
80.33
C + P*





*A mixture of Forms C and P.













TABLE 34







Details and results of 1 week slurry experiments
















Starting










material
Amt



Recovery
Recovery
Form


Entry
(Form)
(mg)
Solvent
Amt (mL)
Temp (° C.)
(mg)
(%)
(1 wk)


















1
Form N
34.1
water
1.0
RT
23.0
67.4
A


2

28.8
IPA
1.0
RT
21.2
73.6
C + P


3

27.9
EtOAc
1.0
RT
23.3
83.5
C + P


4

27.5
MeCN
1.0
RT
23.2
84.4
C


5

27.6
EtOH
1.0
RT
21.7
78.6
C


6

27.1
Dioxane
1.0
RT
24.1
88.9
C + P


7
Form L
30.3
water
1.0
RT
16.4
54.1
A


8

26.8
IPA
1.0
RT
23.9
89.2
L


9

27.7
EtOAc
1.0
RT
25.3
91.3
L


10

27.5
MeCN
1.0
RT
18.6
67.6
C


11

27.2
EtOH
1.0
RT
20.8
76.5
C


12

28.2
Dioxane
1.0
RT
25.0
88.7
L


13
Form N
30.3
water
1.0
RT
21.1
69.6
A


14

30.1
IPA
1.0
RT
24.7
82.1
C


15

27.7
EtOAc
1.0
RT
23.1
83.4
C


16

27.5
MeCN
1.0
RT
18.1
65.8
C


17

28.7
EtOH
1.0
RT
24.1
84.0
C


18

26.4
Dioxane
1.0
RT
19.8
75.0
C


19
Form C
27.1
water
1.0
RT
22.3
82.3
A


20

27.3
IPA
1.0
RT
23.6
86.4
C


21

28.4
EtOAc
1.0
RT
24.5
86.3
C


22

27.9
MeCN
1.0
RT
25.6
91.8
C


23

28.0
EtOH
1.0
RT
23.9
85.4
C


24

27.5
Dioxane
1.0
RT
22.5
81.8
C


25
Form H
26.8
water
1.0
RT
18.8
70.1
A


26

26.8
IPA
1.0
RT
23.5
87.7
C


27

26.8
EtOAc
1.0
RT
24.8
92.5
C


28

27.6
MeCN
1.0
RT
24.5
88.8
C


29

26.5
EtOH
1.0
RT
21.8
82.3
C


30

26.5
Dioxane
1.0
RT
22.2
83.8
C


31
Form A
26.6
water
1.0
RT
19.2
72.2
A


32

29.7
IPA
1.0
RT
20.9
70.3
A


33

29.2
EtOAc
1.0
RT
10.7
36.6
A


34

27.4
MeCN
1.0
RT
12.9
47.1
C


35

26.8
EtOH
1.0
RT
17.0
63.4
C


36

28.2
Dioxane
1.0
RT
17.6
62.4
A
















TABLE 35







Details and results of MeCN/water slurry experiments of Forms A and C at


ambient temperature.
















Amt
Amt
MeCN/water


Filtration

Filtration


Starting
Form A
Form C
(v/v
Amt
Form
after
Form
after


material
(mg)
(mg)
ratio)
(mL)
1 day
1 day
1 wk
1 week


















50:50 A/C
20.2
20.0
1:9
2.0
A
slow
A
slow



20.8
20.4
 1:1*
2.0
*
*
*
*



20.5
20.8
9:1
2.0
A
fast
A
fast


Form A
39.4
0.0
1:9
2.0
A
slow
A
slow



37.2
0.0
 1:1*
2.0
*
*
*
*



38.0
0.0
9:1
2.0
A
fast
A
fast


Form C
0.0
40.2
1:9
2.0
A
slow
A
slow



0.0
37.8
 1:1*
2.0
*
*
*
*



0.0
39.2
9:1
2.0
A
fast
A
fast





*All solids were dissolved in 50/50 mixtures of MeCN/water, therefore characterization was not possible.













TABLE 36a







Analytical results summary for non-solvated forms of Compound 1.

















TGA




Form
State
Solvents/Conditions
DSC (° C.)
(wt %)
IC
NMR





A
Monohydrate
Slurry in water
68, 198{circumflex over ( )}, 327
2.4
1.0:1
Consistent;








Protonated








species observed


C
Anhydrate
MeOH/MeCN
335
0.0
1.0:1
Consistent;








Protonated








species observed


D
Anhydrate
DMA/MTBE
249, 264{circumflex over ( )}, 318
1.8
0.6:1
Consistent;




(observed twice)



Protonated








species observed


F
De-solvate
Heat solvate in
328

1.0:1
Consistent;




TGA



Protonated








species observed


H
Anhydrate
Fast cooling and
331
0.5
1.0:1
Consistent;


(C + P)

slow cooling with



Protonated




multiple solvents



species observed


J
Anhydrate
Fast cooling and
219, 225{circumflex over ( )}, 236{circumflex over ( )},
0.4
0.9:1
Consistent;




slow cooling with
323, 328, 338


Protonated




multiple solvents



species observed


K
Anhydrate
EtOH/THF fast
322
0.0
0.9:1
Consistent;




cooling



Protonated








species observed


L
Channel
EtOH/MTBE,
333
1.7
1.0:1
Consistent;



hydrate
DMF/MeCN



Protonated








species observed


M
unknown hydrate
DMF/IPAc
215, 332
6.0

Consistent;








Protonated








species observed


N
unknown hydrate
MeOH/EtOAc
 335**
2.4
 1.0:1*
Consistent;








Protonated








species observed


O
De-hydrate
Heat hydrate in
332







TGA


P
Unknown form
Found during in-







(metastable)
process checks that




yielded Form H





{circumflex over ( )}Indicates exothermic event;


*Takes into account one-half mole of water present;


**Baseline shift observed at ~175° C.;


— indicates test was not performed or does not apply













TABLE 36b







Analytical results summary for solvate forms of Compound 1.

















TGA




Form
State
Solvents/Conditions
DSC (° C.)
(wt %)
IC
NMR
















B
DMA Solvate
DMA as primary
189, 238{circumflex over ( )}, 330
11.7
0.8:1*
Consistent;




solvent



Protonated species observed


E
NMP Solvate
NMP as primary
220, 228{circumflex over ( )}, 236
11.9
0.8:1*
Consistent;




solvent



Protonated species observed


G
DMF Solvate
DMF as primary
201, 336
11.6
1.2:1*
Consistent;




solvent



Protonated species observed


I
THF Solvate
THF used as anti-
206, 242{circumflex over ( )}, 336
7.9
 0.9:1**
Consistent;




solvent



Protonated species observed





{circumflex over ( )}Indicates exothermic event.


*Takes one mole of solvent into account.


**Takes 6.4 wt % THF present into account - form may be a hemi-solvate or incomplete solvate.






Example 8
Scale-Up Experiments of Compound 1, Forms A, C and L

Preparation of Forms A, C, and H of Compound 1 were carried out on 300 mg scale in DMA/IPA, DMF/MeCN, and MeOH/EtOAc respectively, by charging Compound 1 to a 25 mL equipped with magnetic stir bar and thermocouple. To this was added the appropriate solvent in one portion followed by heating to 60-70° C. with stirring until complete dissolution was observed. Each reaction solution was polish filtered followed by the addition of anti-solvents at elevated temperatures with additional stirring for 5-10 minutes. Each reaction was then allowed to slowly cool to ambient temperature at a rate of 20° C./h, followed by further stirring for 16 hours. Solids were isolated by filtration and dried under vacuum at ambient temperature for 16 hours to give Compound 1 (286 mg, 95% yield; 276 mg, 91% yield; and 228 mg, 75% yield) as Forms N, L, and N respectively. A summary of the experimental details are outlined in Table 33.


Preparation of Forms A, C and H of Compound 1 were also carried out on 400 mg scale in MeOH/IPA, MeOH/MeCN and EtOH/EtOAc, respectively. Form C was produced by using MeOH instead of DMF as the primary solvent. The crystallization targeting Form A was found to produce a mixture of Forms C and P (originally designated as Form H) upon isolation. In-process checks presented a pattern consistent with Form P. Since the mixture of forms was isolated, the material was seeded with Form A in an effort to generate Form A. However, seeding did not produce Form A. Therefore the resulting material was isolated as a mixture and then was further slurried in water to afford Form A.


Example 9
Slurry Experiments

Slurry experiments were performed using six solvents (water, IPA, EtOAc, MeCN, EtOH and dioxane) and six lots of Compound 1. Each vial was charged with approximately 25-30 mg of API and 1 mL of the corresponding solvent, followed by stirring at room temperature for one week. Table 34 shows experimental details. The obtained solids were isolated by filtration, dried under vacuum at room temperature and analyzed by XRPD.


Slurry experiments of Compound 1, Forms A and C, were conducted in mixtures of MeCN and water. Each vial was charged with approximately 40 mg of Compound 1 (pure Form A, pure Form C, or 50:50 mixtures) and 2 mL of MeCN/water mixture in a 1:9, 1:1, and 9:1 ratio. Table 35 provides the experimental details. Some samples (indicated in table) afforded clear solutions in the 1:1 mixture of MeCN/water, therefore characterization of these samples could not be performed, but the experiment was continued to determine if a more stable form would precipitate. All samples were stirred at room temperature for approximately 24 hours. An aliquot (1 mL) of the suspension was taken from the slurries (samples #1, 3, 4, 6, 7 and 9) for determination of form by XRPD. All aliquots were filtered and dried in vacuo at 30 mm Hg and ambient temperature. All samples were stirred at ambient temperature for an additional six days, filtered, dried, and again the solid form and filterability were determined.


It was also found during the concurrent process control screen that Form A was isolated when Form B or Form G was slurried in DI water for several hours (2-24 hours).


Example 10
Further Preparation of Compound 1, Forms A and C

Preparation of the Compound 1 Form A was carried out by charging a free base version of Compound 1 (18 g, 0.036 mol.) to a 1 L-3N RBF equipped with magnetic stir bar and thermocouple. To this was added a 1:1 (v/v) mixture of MeCN and water in one portion at ambient temperature. Once complete dissolution was observed, concentrated HCl (3.062 mL, 1.05 equiv.) was added to the reaction solution, followed by additional stirring for 10 minutes, followed by a polish filtration. Additional MeCN (720 mL, 40 vol.) was then added as an anti-solvent to facilitate precipitation. The resultant slurry was allowed to stir at ambient temperature for approximately 19 h, before the solids were isolated by filtration. The filter cake was rinsed with two portions of MeCN (2×100 mL, 5.6 vol.), and dried under vacuum at ambient temperature for 16 hours to give Compound 1 (16.19 g, 84% yield) as a light orange crystalline (Form A) powder.


Compound 1 Form A (5.05 g) was slurried in 50 mL (10 vol.) of anhydrous MeCN at room temperature under nitrogen. In-process samples were taken after 24, 72, and 96 hours and XRPD showed the material to still be Form A. To help promote conversion to Form C, the reaction mixture was diluted with anhydrous MeCN (150 mL, 30 vol.) and allowed to stir at room temperature for three days, as previous experiments to convert Form A to Form C were successful using 30 or more volumes of MeCN. After stirring for an additional three days the material was still found to be Form A.


Based on a previous observation that Form C could also be obtained from a cooling crystallization employing acetonitrile and MeOH, it was decided to add anhydrous MeOH (50 mL, 10 volumes) and to heat the reaction mixture to 60° C. in order to improve the solubility of the material and possibly promote solution-mediated polymorph conversion. After stirring with the addition of MeOH, the slurry visibly thinned out then thickened after approximately 30 minutes at 60° C. Analysis of a small aliquot from the batch by XRPD showed the material to be consistent with Form C. After stirring the slurry at 60° C. for a total of 1.5 hours, the mixture was naturally cooled to room temperature. The solids were isolated by filtration, washed with MeCN (2×50 mL) and dried under vacuum at 40° C. to afford 4.26 g of Compound 1 Form C material (84% recovery).


Example 11
Solubility Study

The solubility measurement of Compound 1 Form A was performed in DI water (Milli-Q) and in 20 mM phosphate buffer (pH 3.2), see Table 37. Each vial was charged with 50-55 mg of Compound 1 and 1 mL of the corresponding solvent.









TABLE 37







Solubility of Form A of Compound 1.















Solu-








bility

Solubility
pH



pH
mg/mL
XRPD
mg/mL
(final,
XRPD


Media
(initial)
(1 day)
(1 day)
(1 week)
1 week)
(1 week)





DI water
8.1
3.1
Form A
4.3
5.0
Form A


Phosphate
3.2
3.3
Form A
4.1
3.3
Form A


buffer









Both slurries were stirred at room temperature for approximately 19 hours. An aliquot (0.2 mL) was taken from each sample for the solubility determination and XRPD test. Table 36 shows the results of these tests. Both slurries were allowed to stir for an additional 6 days at room temperature. The pH of the supernatant liquid and solubility were determined following the 1 week slurry.


Example 12
Humidity Chamber Study

Humidity chamber studies of Compound 1 Forms A and C were set up at ambient temperature as shown in Table 38. The 0% RH chamber was prepared with drierite and the 95% RH chamber was made with a saturated solution of Na2HPO4.12H2O in DI water. The chambers were equilibrated for >1 week prior to introducing samples. Samples were placed in Teflon-lined caps for scintillation vials and allowed to equilibrate for one week then analyzed by XRPD and KF to determine form and water content.









TABLE 38







Humidity chamber studies of Compound 1 Forms A and C.













Form
KF

Result Form
Result



(initial)
(initial)
% RH
(1 week)
KF (1 week)

















C
0.20
0%
C
0.51



A
3.3
0%
A
3.6



C
0.20
95%
A
3.5



A
3.3
95%
A
3.3










Further stability studies in humid environments were performed and showed Form C converted to Form A after equilibrating at 95% RH for one week at ambient temperature. FIG. 60 summarizes the conversion of the forms of Compound 1 observed from slurry and humidity chamber studies.


Example 13
Drying Study

Compound 1 (4.08 g, Form A) was dried under vacuum at 40° C. overnight. A small sample (˜200 mg) was taken for XRPD and OVI tests. The rest of the material was further dried under vacuum at 55° C. overnight. A second sample (˜200 mg) was taken for XRPD and OVI tests. The oven temperature was increased to 70° C. and the material was further dried under vacuum at 70° C. overnight. A third sample (˜200 mg) was taken for XRPD and OVI tests. The oven temperature was then increased to 85° C. and the material was further dried under vacuum over weekend. Analysis by XRPD and OVI was completed as shown in Table 39 along with experimental details. A summary of moisture sorption data for Forms A, C, and L is shown in Table 40.









TABLE 39







Drying Study of Compound 1 Form A










Temperature
Time at Temperature
Form by
Residual Amt of


(° C.)
(total)
XRPD
MeCN by OVI (ppm)













40
16
A
1353


55
24 (40)
A
1303


70
24 (64)
A
1113


85
 72 (136)
A
670
















TABLE 40







Summary of Moisture Data for Forms A, C, and L












Form
State
KF
Moisture Sorption

















A
Monohydrate
3.1%*
60% RH:
3.7%






90% RH:
4.2%










XRPD consistent




before and after



analysis













C
Anhydrate
0.2%
60% RH:
1.4%






90% RH:
1.9%










XRPD consistent




before and after



analysis













L
Channel
2.1%
60% RH:
2.9%




Hydrate

90% RH:
3.9%







The theoretical weight % water for a monohydrate is 3.2%.





Claims
  • 1. A polymorphic form of Compound 1 having the formula:
  • 2. The polymorphic form of claim 1, wherein the polymorphic form is Form B which is a dimethylacetamide (DMA) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 13.8, 17.1 and 19.7 degrees 2-theta (°2θ).
  • 3. The polymorphic form of claim 2, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 16.5, 20.1 and 25.0°2θ.
  • 4. The polymorphic form of claim 2, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 7.
  • 5. The polymorphic form of claim 2, wherein said Form B further having a differential scanning calorimetry (DSC) curve comprising a first and a second endotherms and an exotherm, wherein said first endotherm is centered at about 211° C., said second endotherm is forked and having peaks centered at about 331° C. and at about 338° C., and said exotherm is centered at about 245° C.
  • 6. The polymorphic form of claim 2, wherein said Form B further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 8.
  • 7. The polymorphic form of claim 2, wherein said Form B is prepared by treating Compound 1 with DMA.
  • 8. The polymorphic form of claim 1, wherein the polymorphic form is Form C which is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.1, 19.8 and 26.4°2θ
  • 9. The polymorphic form of claim 8, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 17.7 and 22.0°2θ.
  • 10. The polymorphic form of claim 8, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 11.
  • 11. The polymorphic form of claim 8, wherein Form C further having a differential scanning calorimetry (DSC) curve comprising an endotherm which onset at about 314° C.
  • 12. The polymorphic form of claim 11, wherein the endotherm is centered at about 335° C.
  • 13. The polymorphic form of claim 8, wherein said Form C having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 12.
  • 14. The polymorphic form of claim 8, wherein said Form C is prepared by drying Compound 1.
  • 15. The polymorphic form of claim 8, wherein said Form C is prepared by dissolving Compound 1 in an anhydrous solvent.
  • 16. The polymorphic form of claim 1, wherein the polymorphic form is Form D which is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.8, 17.6, and 20.9°2θ
  • 17. The polymorphic form of claim 16, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 5.9 and 25.2°2θ
  • 18. The polymorphic form of claim 16, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 16.
  • 19. The polymorphic form of claim 16, wherein said Form D further having a differential scanning calorimetry (DSC) curve comprising an endotherm centered at about 249° C. and an exotherm centered at about 264° C.
  • 20. The polymorphic form of claim 16, wherein said Form D further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 17.
  • 21. The polymorphic form of claim 16, wherein said Form D is prepared by: dissolving Compound 1 in DMA; andadding an antisolvent to the Compound 1 and DMA solution, wherein the antisolvent is methyl tert-butylether (MTBE).
  • 22. The polymorphic form of claim 1, wherein said polymorphic form is Form E which is an N-methylpyrrolidinone (NMP) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 17.0, 19.6 and 20.2°2θ.
  • 23. The polymorphic form of claim 22, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 13.9, 25.1 and 26.2°2θ.
  • 24. The polymorphic form of claim 22, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 20.
  • 25. The polymorphic form of claim 22, wherein said Form E further having a differential scanning calorimetry (DSC) curve comprising a first and a second endotherm and an exotherm, wherein said first endotherm centered at about 220° C., said second endotherm centered at about 336° C., and said exotherm centered at about 228° C.
  • 26. The polymorphic form of claim 22, wherein said Form E further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 21.
  • 27. The polymorphic form of claim 22, wherein said Form E is prepared by treating Compound 1 with NMP.
  • 28. The polymorphic form of claim 1, wherein the polymorphic form is Form F which is a desolvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.0, 17.2, and 25.9°2θ.
  • 29. The polymorphic form of claim 28, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 5.2, 10.3 and 20.2°2θ.
  • 30. The polymorphic form of claim 28, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 24.
  • 31. The polymorphic form of claim 28, wherein said Form F further having a differential scanning calorimetry (DSC) curve comprising an endotherm which onset about 304° C.
  • 32. The polymorphic form of claim 31, wherein the endotherm is centered at about 328° C.
  • 33. The polymorphic form of claim 28, wherein said Form F further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 25.
  • 34. The polymorphic form of claim 28, wherein said Form F is prepared by treating Compound 1 with DMA or DMF, and heating the treated Compound 1.
  • 35. The polymorphic form of claim 1, wherein the polymorphic form is Form G which is a dimethylformamide (DMF) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.5, 10.9 and 22.0°2θ.
  • 36. The polymorphic form of claim 35, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 16.5, 18.4 and 19.5°2θ.
  • 37. The polymorphic form of claim 35, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 27.
  • 38. The polymorphic form of claim 35, wherein Form G further having a differential scanning calorimetry (DSC) curve comprising a broad endotherm at approximately 201° C. and a second endotherm which onsets at approximately 314° C.
  • 39. The polymorphic form of claim 38, wherein the second endotherm is centered at about 336° C.
  • 40. The polymorphic form of claim 35, wherein Form G further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 28.
  • 41. The polymorphic form of claim 35, wherein Form G is prepared by treating Compound 1 with DMF.
  • 42. The polymorphic form of claim 1, wherein the polymorphic form is Form I which is a tetrahydrofuran (THF) solvate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 7.0, 16.7 and 17.4 degrees 2-theta (°2θ).
  • 43. The polymorphic form of claim 42, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 19.6, 20.2 and 24.6°2θ.
  • 44. The polymorphic form of claim 42, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 31.
  • 45. The polymorphic form of claim 42, wherein said Form I further having a differential scanning calorimetry (DSC) curve comprising a first endotherm centered at about 206° C., an exotherm centered at about 242° C., and a second endotherm onset at about 314° C. and centered at about 336° C.
  • 46. The polymorphic form of claim 42, wherein said Form I further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 32.
  • 47. The polymorphic form of claim 42, wherein said Form I is prepared by treating Compound 1 with THF.
  • 48. The polymorphic form of claim 1, wherein the polymorphic form is Form J which is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 4.9, 17.5 and 20.0 degrees 2-theta (°2θ).
  • 49. The polymorphic form of claim 48, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 9.2, 22.1 and 25.2°2θ.
  • 50. The polymorphic form of claim 48, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 35.
  • 51. The polymorphic form of claim 48, wherein said Form J further having a differential scanning calorimetry (DSC) curve comprising a first endotherm centered at about 219° C., a forked exotherm having peaks centered at about 223° C. and 236° C., and a forked endotherm which onsets at about 302° C.
  • 52. The polymorphic form of claim 51, wherein the forked endotherm having peaks centered at approximately 323° C., 328° C. and 338° C.
  • 53. The polymorphic form of claim 48, wherein said Form J further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 36.
  • 54. The polymorphic form of claim 48, wherein said Form J is prepared by treating Compound 1 with isopropyl alcohol.
  • 55. The polymorphic form of claim 1, wherein the polymorphic form is Form K which is an anhydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.3, 8.5 and 10.5°2θ.
  • 56. The polymorphic form of claim 55, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 13.3, 18.6 and 21.3°2θ.
  • 57. The polymorphic form of claim 55, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 39.
  • 58. The polymorphic form of claim 55, wherein said Form K further having a differential scanning calorimetry (DSC) curve comprising an endotherm onset at about 306° C.
  • 59. The polymorphic form of claim 58, wherein the endotherm is centered at about 322° C.
  • 60. The polymorphic form of claim 55, wherein said Form K further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 40.
  • 61. The polymorphic form of claim 55, wherein said Form K is prepared by: dissolving Compound 1 in EtOH; andadding THF to the solution.
  • 62. The polymorphic form of claim 1, wherein said polymorphic form is Form L which is a channel hydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 10.4 and 20.7 degrees 2-theta (°2θ).
  • 63. The polymorphic form of claim 62, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 15.5, 16.9 and 24.4°2θ.
  • 64. The polymorphic form of claim 62, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 43.
  • 65. The polymorphic form of claim 62, wherein said Form L having a differential scanning calorimetry (DSC) curve comprising an endotherm which onsets at about 303° C.
  • 66. The polymorphic form of claim 65, wherein the endotherm is centered at about 333° C.
  • 67. The polymorphic form of claim 62, wherein said Form L having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 44.
  • 68. The polymorphic form of claim 62, wherein said Form L is prepared by dissolving Compound 1 in methanol; andadding an antisolvent to Compound 1 dissolved in the solvent, wherein the antisolvent is selected from the group consisting of methyl tert-butylether, isopropyl acetate and heptane.
  • 69. The polymorphic form of Compound 1, wherein the polymorphic form is Form M having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.1, 8.2 and 10.2 degrees 2-theta (°2θ).
  • 70. The polymorphic form of claim 69, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 18.1 and 20.6°2θ.
  • 71. The polymorphic form of claim 69, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 48.
  • 72. The polymorphic form of claim 69, wherein Form M further having a differential scanning calorimetry (DSC) curve comprising an endotherm centered at about 332° C.
  • 73. The polymorphic form of claim 69, wherein said Form M further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 49.
  • 74. The polymorphic form of claim 69, wherein said Form M is prepared by treating Compound 1 with water.
  • 75. The polymorphic form of Compound 1, wherein the polymorphic form is Form N having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.2, 8.4 and 10.3°2θ.
  • 76. The polymorphic form of claim 75, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 18.6, 20.0 and 21.0°2θ.
  • 77. The polymorphic form of claim 75, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 52.
  • 78. The polymorphic form of claim 75, wherein said Form N further having a differential scanning calorimetry (DSC) curve comprising an endotherm centered at about 333° C.
  • 79. The polymorphic form of claim 75, wherein said Form N further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 53.
  • 80. The polymorphic form of claim 75, wherein said Form N is prepared by treating Compound 1 with water.
  • 81. The polymorphic form of claim 1, wherein said polymorphic form is Form O which is a dehydrate having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 6.3, 12.6 and 25.3°2θ.
  • 82. The polymorphic form of claim 81, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 10.5 and 21.0°2θ.
  • 83. The polymorphic form of claim 81, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 56.
  • 84. The polymorphic form of claim 81, wherein said Form O further having a differential scanning calorimetry (DSC) curve comprising an endotherm centered at about 327° C.
  • 85. The polymorphic form of claim 81, wherein said Form O further having substantially a differential scanning calorimetry (DSC) curve as shown in FIG. 57.
  • 86. The polymorphic form of claim 81, wherein said Form O is prepared by treating Compound 1 with water; andheating the treated Compound 1.
  • 87. The polymorphic form of claim 1, wherein the polymorphic form is Form P which having an X-ray powder diffraction pattern (CuKα) comprising significant diffraction peaks at about 5.0, 9.4 and 10.0 degrees 2-theta (°2θ).
  • 88. The polymorphic form of claim 87, wherein the X-ray powder diffraction pattern (CuKα) further comprises significant diffraction peaks at about 17.2 and 25.7°2θ.
  • 89. The polymorphic form of claim 87, wherein the X-ray diffraction pattern (CuKα) is substantially as shown in FIG. 59.
  • 90. A pharmaceutical composition comprising, as an active ingredient, Compound 1 of the formula:
  • 91. The pharmaceutical composition of claim 82, wherein the portion of polymorphic form is between about 0.1% to about 100%.
  • 92. The pharmaceutical composition of claim 82, wherein said portion is greater than 1%.
  • 93. The pharmaceutical composition of claim 82, wherein said portion is greater than 10%.
  • 94. The pharmaceutical composition of claim 82, wherein said portion is greater than 90%.
  • 95. A therapeutic method comprising: administering Compound 1, wherein at least a portion of Compound 1 is present as a polymorphic form selected from the group consisting of Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P
  • 96. A method of treating a disease state for which a kinase possesses activity that contributes to the pathology and/or symptomology of the disease state, the method comprising: administering Compound 1 to a subject in need thereof, wherein at least a portion of Compound 1 is present as a polymorphic form selected from the group consisting of Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P.
  • 97. A method of treating cancer comprising administering a therapeutically effective amount of Compound 1 to a mammalian species in need thereof, wherein at least a portion of Compound 1 is present as a polymorphic form selected from the group consisting of Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P.
  • 98. The method according to claim 97, wherein the cancer is selected from the group consisting of squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, small-cell lung cancer, non small-cell lung cancers, bladder cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, glioma, colorectal cancer, genitourinary cancer, gastrointestinal cancer, thyroid cancer, skin cancer, kidney cancer, rectal cancer, colonic cancer, cervical cancer, mesothelioma, pancreatic cancer, liver cancer, uterus cancer, cerebral tumor cancer, urinary bladder cancer and blood cancers including multiple myeloma, chronic myelogenous leukemia and acute lymphocytic leukemia.
  • 99. A method for preventing or treating dementia related diseases, Alzheimer's Disease and conditions associated with kinases, comprising administration to a mammalian species in need thereof of a therapeutically effective amount of Compound 1, wherein at least a portion of Compound 1 is present as a polymorphic form selected from the group consisting of Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P.
  • 100. The method according to claim 99, wherein the dementia related diseases are selected from the group consisting of Frontotemporal dementia Parkinson's Type, Parkinson dementia complex of Guam, HIV dementia, diseases with associated neurofibrillar tangle pathologies, predemented states, vascular dementia, dementia with Lewy bodies, Frontotemporal dementia and dementia pugilistica.
  • 101. A method for treating arthritis comprising administration to a mammal in need thereof a therapeutically effective amount of Compound 1, wherein at least a portion of Compound 1 is present as a polymorphic form selected from the group consisting of Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P.
  • 102. A method of inhibiting cell proliferation in a patient comprising administering to the patient a therapeutically effective amount of Compound 1, wherein at least a portion of Compound 1 is present as a polymorphic form selected from the group consisting of Form B, Form C, Form D, Form E, Form F, Form G, Form I, Form J, Form K, Form L, Form M, Form N, Form O and Form P.
RELATED APPLICATION

This application claims benefit of U.S. Provisional Application No. 61/045,523 filed on Apr. 16, 2008, which is incorporated herein by reference.

Provisional Applications (1)
Number Date Country
61045523 Apr 2008 US