Forms and compositions of an ERK inhibitor

Information

  • Patent Grant
  • 10005760
  • Patent Number
    10,005,760
  • Date Filed
    Wednesday, August 12, 2015
    8 years ago
  • Date Issued
    Tuesday, June 26, 2018
    6 years ago
Abstract
The present invention provides compounds, compositions thereof, and methods of using the same for the inhibition of one or both of ERK1 and ERK2.
Description
FIELD OF THE INVENTION

The present invention provides various forms and compositions useful as inhibitors of ERK kinases, for example one or both of ERK1 and ERK2 kinases.


BACKGROUND OF THE INVENTION

The search for new therapeutic agents has been greatly aided in recent years by a better understanding of the structure of enzymes and other biomolecules associated with diseases. One important class of enzymes that has been the subject of extensive study is protein kinases.


Protein kinases constitute a large family of structurally related enzymes that are responsible for the control of a variety of signal transduction processes within the cell. 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 kinases may be categorized into families by the substrates they phosphorylate (e.g., protein-tyrosine, protein-serine/threonine, lipids, etc.).


The processes involved in tumor growth, progression, and metastasis are mediated by signaling pathways that are activated in cancer cells. The MAPK or Raf-Mek-ERK pathway plays a central role in regulating mammalian cell growth by relaying extracellular signals from ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase. Activation of the ERK occurs via a cascade of phosphorylation events that begins with activation of Ras. Activation of Ras leads to the recruitment and activation of Raf, a serine-threonine kinase. Activated Raf then phosphorylates and activates MEK1/2, which then phosphorylates and activates one or both of ERK1 and ERK2. When activated, one or both of ERK1 and ERK2 phosphorylates several downstream targets involved in a multitude of cellular events including cytoskeletal changes and transcriptional activation. The ERK/MAPK pathway is one of the most important for cell proliferation, and human tumor data suggest that the ERK/MAPK pathway is frequently activated in many tumors. Ras genes, which are upstream of one or both of ERK1 and ERK2, are mutated in several cancers including colorectal, melanoma, breast, lung, and pancreatic tumors. High Ras activity is accompanied by elevated ERK activity in many human tumors. In addition, activating mutations of BRAF, a serine-threonine kinase of the Raf family, are associated with increased RAF, MEK, and ERK kinase activity. Tumors types with the most frequent mutations in BRAF include melanomas (60%), thyroid cancers (greater than 40%) and colorectal cancers.


Many diseases are associated with abnormal cellular responses, proliferation and evasion of programmed cell-death, triggered by protein kinase-mediated events as described above. Accordingly, there remains a need to find protein kinase inhibitors useful as therapeutic agents.


SUMMARY OF THE INVENTION

It has now been found that novel forms of the present invention, and compositions thereof, are useful as inhibitors of one or more protein kinases and exhibit desirable characteristics for the same. In general, salt forms or freebase forms, and pharmaceutically acceptable compositions thereof, are useful for treating or lessening the severity of a variety of diseases or disorders as described in detail herein.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 depicts an XRPD pattern of Form A of compound 1.



FIG. 2 depicts a DSC thermogram and TGA trace of Form A of compound 1.



FIG. 3 depicts a DVS plot of Form A of compound 1.



FIG. 4 depicts an XRPD pattern of Form B of compound 1.



FIG. 5 depicts a DSC thermogram of Form B of compound 1.



FIG. 6 depicts a TGA trace of Form B of compound 1.



FIG. 7 depicts an XRPD pattern of Form C of compound 1.



FIG. 8 depicts a DSC thermogram of Form C of compound 1.



FIG. 9 depicts a TGA trace of Form C of compound 1.



FIG. 10 depicts an XRPD pattern of Form D of compound 1.



FIG. 11 depicts a DSC thermogram of Form D of compound 1.



FIG. 12 depicts a TGA trace of Form D of compound 1.



FIG. 13 depicts an XRPD pattern of Form A of compound 2.



FIG. 14 depicts a DSC thermogram of Form A of compound 2.



FIG. 15 depicts a TGA trace of Form A of compound 2.



FIG. 16 depicts a DVS plot of Form A of compound 2.



FIG. 17 depicts an XRPD pattern of Form B of compound 2.



FIG. 18 depicts a DSC thermogram of Form B of compound 2.



FIG. 19 depicts a TGA trace of Form B of compound 2.



FIG. 20 depicts a DVS plot of Form B of compound 2.



FIG. 21 depicts an XRPD pattern of Form C of compound 2.



FIG. 22 depicts a DSC thermogram of Form C of compound 2.



FIG. 23 depicts a TGA trace of Form C of compound 2.



FIG. 24 depicts a DVS plot of Form C of compound 2.



FIG. 25 depicts an XRPD pattern of Form D of compound 2.



FIG. 26 depicts a DSC thermogram of Form D of compound 2.



FIG. 27 depicts a TGA trace of Form D of compound 2.



FIG. 28 depicts an XRPD pattern of Form A of compound 3.



FIG. 29 depicts a DSC thermogram of Form A of compound 3.



FIG. 30 depicts a TGA trace of Form A of compound 3.



FIG. 31 depicts a DVS plot of Form A of compound 3.



FIG. 32 depicts an XRPD pattern of Form A of compound 4.



FIG. 33 depicts a DSC thermogram of Form A of compound 4.



FIG. 34 depicts a TGA trace of Form A of compound 4.



FIG. 35 depicts a DVS plot of Form A of compound 4.



FIG. 36 depicts an XRPD pattern of Form B of compound 4.



FIG. 37 depicts a DSC thermogram and TGA trace of Form B of compound 4.



FIG. 38 depicts an XRPD pattern of Form C of compound 4.



FIG. 39 depicts a DSC thermogram of Form C of compound 4.



FIG. 40 depicts an XRPD pattern of Form D of compound 4.



FIG. 41 depicts a DSC thermogram of Form D of compound 4.



FIG. 42 depicts a DSC thermogram and TGA trace of Form D of compound 4.



FIG. 43 depicts a DVS plot of Form D of compound 4.



FIG. 44 depicts an XRPD pattern of Form E of compound 4.



FIG. 45 depicts a DSC thermogram of Form E of compound 4.



FIG. 46 depicts an XRPD pattern of Form F of compound 4.



FIG. 47 depicts a DSC thermogram of Form F of compound 4.



FIG. 48 depicts an XRPD pattern of Form G of compound 4.



FIG. 49 depicts a DSC thermogram of Form G of compound 4.



FIG. 50 depicts an XRPD pattern of Form H of compound 4.



FIG. 51 depicts a DSC thermogram of Form H of compound 4.



FIG. 52 depicts an XRPD pattern of Form I of compound 4.



FIG. 53 depicts an XRPD pattern of Form A of compound 5.



FIG. 54 depicts a DSC thermogram of Form A of compound 5.



FIG. 55 depicts a TGA trace of Form A of compound 5.



FIG. 56 depicts a DVS plot of Form A of compound 5.



FIG. 57 depicts an XRPD pattern of Form B of compound 5.



FIG. 58 depicts a DSC thermogram and TGA trace of Form B of compound 5.



FIG. 59 depicts an XRPD pattern of Form C of compound 5.



FIG. 60 depicts a DSC thermogram and TGA trace of Form C of compound 5.



FIG. 61 depicts an XRPD pattern of Form D of compound 5.



FIG. 62 depicts a DSC thermogram and TGA trace of Form D of compound 5.



FIG. 63 depicts an XRPD pattern of Form E of compound 5.



FIG. 64 depicts a DSC thermogram of Form E of compound 5.



FIG. 65 depicts a DVS plot of Form E of compound 5.



FIG. 66 depicts an XRPD pattern of Form A of compound 6.



FIG. 67 depicts a DSC thermogram of Form A of compound 6.



FIG. 68 depicts a TGA trace of Form A of compound 6.



FIG. 69 depicts a DVS plot of Form A of compound 6.



FIG. 70 depicts an XRPD pattern of Form B of compound 6.



FIG. 71 depicts a DSC thermogram and TGA trace of Form B of compound 6.



FIG. 72 depicts an XRPD pattern of Form C of compound 6.



FIG. 73 depicts a DSC thermogram and TGA trace of Form C of compound 6.



FIG. 74 depicts an XRPD pattern of Form A of compound 7.



FIG. 75 depicts a DSC thermogram and TGA trace of Form A of compound 7.



FIG. 76 depicts a DVS plot of Form A of compound 7.



FIG. 77 depicts an XRPD pattern of Form A of compound 8.



FIG. 78 depicts a DSC thermogram and TGA trace of Form A of compound 8.



FIG. 79 depicts an XRPD pattern of Form B of compound 8.



FIG. 80 depicts a DSC thermogram and TGA trace of Form B of compound 8.



FIG. 81 depicts an XRPD pattern of Form C of compound 8.



FIG. 82 depicts a DSC thermogram and TGA trace of Form C of compound 8.



FIG. 83 depicts a DVS plot of Form C of compound 8.



FIG. 84 depicts a DSC thermogram and TGA trace of Form D of compound 8.



FIG. 85 depicts an XRPD pattern of Form A of compound 9.



FIG. 86 depicts a DSC thermogram and TGA trace of Form A of compound 9.



FIG. 87 depicts a DVS plot of Form A of compound 9.



FIG. 88 depicts a DSC thermogram and TGA trace of Form B of compound 9.



FIG. 89 depicts a DSC thermogram and TGA trace of Form A of compound 10.



FIG. 90 depicts an XRPD pattern of Form B of compound 10.



FIG. 91 depicts a DSC thermogram and TGA trace of Form B of compound 10.



FIG. 92 depicts an XRPD pattern of Form A of compound 11.



FIG. 93 depicts a DSC thermogram and TGA trace of Form A of compound 11.



FIG. 94 depicts an XRPD pattern of Form A of compound 12.



FIG. 95 depicts a DSC thermogram and TGA trace of Form A of compound 12.



FIG. 96 depicts an XRPD pattern of Form B of compound 12.



FIG. 97 depicts a DSC thermogram and TGA trace of Form B of compound 12.



FIG. 98 depicts an XRPD pattern of Form C of compound 12.



FIG. 99 depicts a DSC thermogram and TGA trace of Form C of compound 12.



FIG. 100A depicts PK data in dogs under fasted conditions.



FIG. 100B depicts PK data in dogs under fed conditions.





DETAILED DESCRIPTION OF THE INVENTION

General Description of Certain Aspects of the Invention:


PCT patent application serial number PCT/US14/15256, filed Feb. 7, 2014 and published as WO 2014/124230 on Aug. 14, 2014 (“the '230 publication,” the entirety of which is hereby incorporated herein by reference), describes certain ERK inhibitor compounds which covalently and irreversibly inhibit activity of one or both of ERK1 and ERK2 kinases. Such compounds include compound 1:




embedded image


Compound 1, N-(2-(2-((2-methoxy-5-methylpyridin-4-yl)amino)-5-(trifluoromethyl)pyrimidin-4-yl)amino)-5-methylphenyl)acrylamide, is designated as compound number 1-90 in the '230 publication and the synthesis of compound 1 is described in detail at Example 94 of the '230 publication, and is reproduced herein for ease of reference.


Compound 1 is active in a variety of assays and therapeutic models demonstrating covalent, irreversible inhibition of one or both of ERK1 and ERK2 kinases (see, e.g., Table A of the '230 publication). Accordingly, compound 1 is useful for treating one or more disorders associated with activity of one or both of ERK1 and ERK2.


It would be desirable to provide a solid form of compound 1 (e.g., as a freebase thereof or salt thereof) that imparts characteristics such as improved aqueous solubility, stability and ease of formulation. Accordingly, the present invention provides both free base forms and salt forms of compound 1.


Free Base Forms of Compound 1


It is contemplated that compound 1 can exist in a variety of physical forms. For example, compound 1 can be in solution, suspension, or in solid form. In certain embodiments, compound 1 is in solid form. When compound 1 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides a form of compound 1 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include different forms of compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 1. In certain embodiments, at least about 95% by weight of a form of compound 1 is present. In still other embodiments of the invention, at least about 99% by weight of a form of compound 1 is present.


According to one embodiment, a form of compound 1 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, a form of compound 1 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, a form of compound 1 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for a form of compound 1 is also meant to include all tautomeric forms of compound 1. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 1 can exist in a variety of solid forms. Exemplary such forms include polymorphs such as those described herein.


As used herein, the term “polymorph” refers to the different crystal structures into which a compound, or a salt or solvate thereof, can crystallize.


In certain embodiments, compound 1 is a crystalline solid. In other embodiments, compound 1 is a crystalline solid substantially free of amorphous compound 1. As used herein, the term “substantially free of amorphous compound 1” means that the compound contains no significant amount of amorphous compound 1. In certain embodiments, at least about 95% by weight of crystalline compound 1 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 1 is present.


It has been found that compound 1 can exist in at least four distinct polymorphic forms. In certain embodiments, the present invention provides a polymorphic form of compound 1 referred to herein as Form A. In certain embodiments, the present invention provides a polymorphic form of compound 1 referred to herein as Form B. In certain embodiments, the present invention provides a polymorphic form of compound 1 referred to herein as Form C. In certain embodiments, the present invention provides a polymorphic form of compound 1 referred to herein as Form D.


In some embodiments, compound 1 is amorphous. In some embodiments, compound 1 is amorphous, and is substantially free of crystalline compound 1.


Form A of Compound 1


In some embodiments, Form A of compound 1 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 1 below.









TABLE 1





XRPD Peak Positions for Form A of Compound 1


Position (°2 θ)















8.6


9.0


11.6


14.1


15.3


15.4


17.2


18.5


20.4


20.6


21.2


21.5


21.9


22.9


23.40


23.9


25.3


25.8


26.1


27.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 1 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.6, 17.2, and 18.5. In some embodiments, Form A of compound 1 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.6, 17.2, and 18.5. In some embodiments, Form A of compound 1 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.6, 17.2, and 18.5. As used herein, the term “about”, when used in reference to a degree 2-theta value refers to the stated value ±0.2 degree 2-theta.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 1.


Methods for preparing Form A of compound 1 are described infra.


Form B of Compound 1


In some embodiments, Form B of compound 1 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 2 below.









TABLE 2





XRPD Peak Positions for Form B of Compound 1


Position (°2 θ)















4.6


7.2


8.3


9.3


11.6


13.6


14.4


16.5


17.7


18.7


21.0


21.3


23.2


23.5


24.4


27.5






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 1 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.2, 9.3, and 17.7. In some embodiments, Form B of compound 1 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.2, 9.3, and 17.7. In some embodiments, Form B of compound 1 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.2, 9.3 and 17.7.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 4.


Methods for preparing Form B of compound 1 are described infra.


Form C of Compound 1


In some embodiments, Form C of compound 1 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 3 below.









TABLE 3





XRPD Peak Positions for Form C of Compound 1


Position (°2 θ)















7.6


8.8


9.6


11.7


12.3


14.5


15.3


15.9


17.5


18.0


20.1


21.0


22.8


23.4


23.8


24.7


25.2


25.5


26.4


27.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 1 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.6, 15.3 and 15.9. In some embodiments, Form C of compound 1 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.6, 15.3 and 15.9. In some embodiments, Form C of compound 1 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.6, 15.3 and 15.9.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 7.


Methods for preparing Form C of compound 1 are described infra.


Form D of Compound 1


In some embodiments, Form D of compound 1 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 4 below.









TABLE 4





XRPD Peak Positions for Form D of Compound 1


Position (°2 θ)















8.2


8.9


9.5


9.6


10.6


15.0


17.3


17.7


19.1


20.2


21.1


22.0


22.4


23.7


24.7


25.2


25.9


26.5


28.6


35.1






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form D of compound 1 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 10.6, 15.0, and 17.3. In some embodiments, Form D of compound 1 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 10.6, 15.0, and 17.3. In some embodiments, Form D of compound 1 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 10.6, 15.0, and 17.3.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 10.


Methods for preparing Form D of compound 1 are described infra.


In some embodiments, the present invention provides compound 1:




embedded image



wherein said compound is crystalline.


In some embodiments, the present invention provides compound 1, wherein said compound is substantially free of amorphous compound 1.


In some embodiments, the present invention provides compound 1, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 1, wherein said compound has one or more peaks in its XRPD selected from those at about 8.6, about 17.2, and about 18.5 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound has at least two peaks in its XRPD selected from those at about 8.6, about 17.2, and about 18.5 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 1, wherein said compound has an XRPD substantially similar to that depicted in FIG. 1.


In some embodiments, the present invention provides compound 1, wherein said compound has one or more peaks in its XRPD selected from those at about 7.2, about 9.3, and about 17.7 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound has at least two peaks in its XRPD selected from those at about 7.2, about 9.3, and about 17.7 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 1, wherein said compound has an XRPD substantially similar to that depicted in FIG. 4.


In some embodiments, the present invention provides compound 1, wherein said compound has one or more peaks in its XRPD selected from those at about 7.6, about 15.3, and about 15.9 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound has at least two peaks in its XRPD selected from those at about 7.6, about 15.3, and about 15.9 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 1, wherein said compound has an XRPD substantially similar to that depicted in FIG. 7.


In some embodiments, the present invention provides compound 1, wherein said compound has one or more peaks in its XRPD selected from those at about 10.6, about 15.0, and about 17.3 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound has at least two peaks in its XRPD selected from those at about 10.6, about 15.0, and about 17.3 degrees 2-theta. In some such embodiments, the present invention provides compound 1, wherein said compound is of Form D.


In some embodiments, the present invention provides compound 1, wherein said has an XRPD substantially similar to that depicted in FIG. 10.


In some embodiments, the present invention provides a composition comprising compound 1 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 1 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 1 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Salt Forms of Compound 1


In some embodiments, an acid and compound 1 are ionically bonded to form one of compounds 2 through 12, described below. It is contemplated that compounds 2 through 12 can exist in a variety of physical forms. For example, compounds 2 through 12 can be in solution, suspension, or in solid form. In certain embodiments, compounds 2 through 12 are in solid form. When compounds 2 through 12 are in solid form, said compounds may be amorphous, crystalline, or a mixture thereof. Exemplary such solid forms of compounds 2 through 12 are described in more detail below.


Compound 2 (Phosphate Salts of Compound 1)


According to one embodiment, the present invention provides a phosphate salt of compound 1, represented by compound 2:




embedded image


It will be appreciated by one of ordinary skill in the art that the phosphoric acid and compound 1 are ionically bonded to form compound 2. It is contemplated that compound 2 can exist in a variety of physical forms. For example, compound 2 can be in solution, suspension, or in solid form. In certain embodiments, compound 2 is in solid form. When compound 2 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 2 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess phosphoric acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 2. In certain embodiments, at least about 95% by weight of compound 2 is present. In still other embodiments of the invention, at least about 99% by weight of compound 2 is present.


According to one embodiment, compound 2 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 2 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 2 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 2 is also meant to include all tautomeric forms of compound 2. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 2 can exist in a variety of solid forms. Exemplary such forms include polymorphs such as those described herein.


In certain embodiments, compound 2 is a crystalline solid. In other embodiments, compound 2 is a crystalline solid substantially free of amorphous compound 2. As used herein, the term “substantially free of amorphous compound 2” means that the compound contains no significant amount of amorphous compound 2. In certain embodiments, at least about 95% by weight of crystalline compound 2 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 2 is present.


It has been found that compound 2 can exist in at least four distinct polymorphic forms. In some embodiments, the present invention provides a polymorphic form of Compound 2 referred to herein as Form A. In certain embodiments, the present invention provides a polymorphic form of compound 2 referred to herein as Form B. In certain embodiments, the present invention provides a polymorphic form of compound 2 referred to herein as Form C. In certain embodiments, the present invention provides a polymorphic form of Compound 2 referred to herein as Form D.


In some embodiments, compound 2 is amorphous. In some embodiments, compound 2 is amorphous, and is substantially free of crystalline compound 2.


Form A of Compound 2


In some embodiments, Form A of compound 2 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 5 below.









TABLE 5





XRPD Peak Positions for Form A of Compound 2


Position (°2 θ)















5.9


6.3


6.8


9.8


10.1


11.1


13.8


14.4


15.4


16.0


16.6


17.3


17.9


18.9


19.2


19.7


20.3


20.8


21.3


22.2


23.0


23.3


23.6


24.0


24.7


25.5


26.0


26.8


27.4


27.9


28.4


29.2


30.5


31.3


31.8






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 2 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 6.8, 10.1, and 20.8. In some embodiments, Form A of compound 2 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 6.8, 10.1, and 20.8. In some embodiments, Form A of compound 2 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 6.8, 10.1, and 20.8.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 13.


Methods for preparing Form A of compound 2 are described infra.


Form B of Compound 2


In some embodiments, Form B of compound 2 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 6 below.









TABLE 6





XRPD Peak Positions for Form B of Compound 2


Position (°2 θ)















3.6


7.3


8.6


9.5


10.7


12.0


13.5


14.6


15.0


15.7


16.6


18.2


19.2


19.9


20.3


21.6


22.0


22.5


22.9


23.4


24.1


24.9


25.3


25.7


26.3


26.9


27.8


28.7


29.5


30.2


31.8


34.2


36.1


37.1


38.8


39.3






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 2 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 3.6, 7.3, and 15.0. In some embodiments, Form B of compound 2 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 3.6, 7.3, and 15.0. In some embodiments, Form B of compound 2 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 3.6, 7.3, and 15.0.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 17.


Methods for preparing Form B of compound 2 are described infra.


Form C of Compound 2


In some embodiments, Form C of compound 2 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 7 below.









TABLE 7





XRPD Peak Positions for Form C of Compound 2


Position (°2 θ)















4.2


6.8


8.4


9.3


11.6


12.5


12.7


13.7


15.3


15.8


16.5


18.7


19.4


20.5


22.0


22.7


24.5


25.2


26.2


32.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 2 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.4, 9.3, and 16.5. In some embodiments, Form C of compound 2 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.4, 9.3, and 16.5. In some embodiments, Form C of compound 2 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.4, 9.3, and 16.5.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 21.


Methods for preparing Form C of compound 2 are described infra.


Form D of Compound 2


In some embodiments, Form D of compound 2 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 8 below.









TABLE 8





XRPD Peak Positions for Form D of Compound 2


Position (°2 θ)















7.1


8.1


9.1


10.4


10.6


11.2


12.9


13.9


15.8


16.4


17.2


17.7


18.7


19.0


20.2


20.7


21.0


22.1


22.7


24.5


25.1


26.4


27.4


27.8


28.7


29.1


31.0


31.5


33.8


36.3


37.0


38.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form D of compound 2 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 9.1, 10.4, and 25.1. In some embodiments, Form D of compound 2 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 9.1, 10.4, and 25.1. In some embodiments, Form D of compound 2 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 9.1, 10.4, and 25.1.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 25.


Methods for preparing Form D of compound 2 are described infra.


In some embodiments, the present invention provides compound 2:




embedded image


In some embodiments, the present invention provides compound 2, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 2, wherein said compound is a crystalline solid substantially free of amorphous compound 2.


In some embodiments, the present invention provides compound 2, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 2, wherein said compound has one or more peaks in its XRPD selected from those at about 6.8, about 10.1, and about 20.8 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound has at least two peaks in its XRPD selected from those at about 6.8, about 10.1, and about 20.8 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 2, wherein said compound has an XRPD substantially similar to that depicted in FIG. 13.


In some embodiments, the present invention provides compound 2, wherein said compound has one or more peaks in its XRPD selected from those at about 3.6, about 7.3, and about 15.0 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound has at least two peaks in its XRPD selected from those at about 3.6, about 7.3, and about 15.0 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 2, wherein said compound has an XRPD substantially similar to that depicted in FIG. 17.


In some embodiments, the present invention provides compound 2, wherein said compound has one or more peaks in its XRPD selected from those at about 8.4, about 9.3, and about 16.5 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound has at least two peaks in its XRPD selected from those at about 8.4, about 9.3, and about 16.5 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 2, wherein said compound has an XRPD substantially similar to that depicted in FIG. 21.


In some embodiments, the present invention provides compound 2, wherein said compound has one or more peaks in its XRPD selected from those at about 9.1, about 10.4, and about 25.1 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound has at least two peaks in its XRPD selected from those at about 9.1, about 10.4, and about 25.1 degrees 2-theta. In some such embodiments, the present invention provides compound 2, wherein said compound is of Form D.


In some embodiments, the present invention provides compound 2, wherein said compound has an XRPD substantially similar to that depicted in FIG. 25.


In some embodiments, the present invention provides a composition comprising compound 2 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 2 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said compound 2 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder the disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 3 (Bisphosphate Complexes of Compound 1)


According to one embodiment, the present invention provides a bisphosphate complex of compound 1, represented by compound 3:




embedded image


It will be appreciated by one of ordinary skill in the art that one molecule of phosphoric acid and compound 1 are ionically bonded, while the second molecule of phosphoric acid and compound 1 are likely hydrogen bonded, to form compound 3. It is contemplated that compound 3 can exist in a variety of physical forms. For example, compound 3 can be in solution, suspension, or in solid form. In certain embodiments, compound 3 is in solid form. When compound 3 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below. For purposes of clarity, it will be understood that compound 3, a bisphosphate complex of compound 1, comprises two molecules of phosphoric acid per molecule of compound 1.


In some embodiments, the present invention provides compound 3 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess phosphoric acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 3. In certain embodiments, at least about 95% by weight of compound 3 is present. In still other embodiments of the invention, at least about 99% by weight of compound 3 is present.


According to one embodiment, compound 3 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 3 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 3 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 3 is also meant to include all tautomeric forms of compound 3. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


In certain embodiments, compound 3 is a crystalline solid. In other embodiments, compound 3 is a crystalline solid substantially free of amorphous compound 3. As used herein, the term “substantially free of amorphous compound 3” means that the compound contains no significant amount of amorphous compound 3. In certain embodiments, at least about 95% by weight of crystalline compound 3 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 3 is present.


It has been found that compound 3 can exist in at least one distinct crystalline form. In some embodiments, the present invention provides a crystalline form of Compound 3 referred to herein as Form A.


In some embodiments, compound 3 is amorphous. In some embodiments, compound 3 is amorphous, and is substantially free of crystalline compound 3.


Form A of Compound 3


In some embodiments, Form A of compound 3 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 9 below.









TABLE 9





XRPD Peak Positions for Form A of Compound 3


Position (°2 θ)















5.7


7.1


8.9


10.3


11.0


11.4


13.2


14.2


14.5


17.2


17.7


18.4


19.4


19.9


20.6


20.8


22.1


22.5


24.3


25.5






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 3 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 10.3, and 11.0. In some embodiments, Form A of compound 3 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 10.3, and 11.0. In some embodiments, Form A of compound 3 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 10.3, and 11.0.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 28.


Methods for preparing Form A of compound 3 are described infra.


In some embodiments, the present invention provides compound 3:




embedded image


In some embodiments, the present invention provides compound 3, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 3, wherein said compound is a crystalline solid substantially free of amorphous compound 3.


In some embodiments, the present invention provides compound 3, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 3, wherein said compound has one or more peaks in its XRPD selected from those at about 8.9, about 10.3, and about 11.0 degrees 2-theta. In some such embodiments, the present invention provides compound 3, wherein said compound has at least two peaks in its XRPD selected from those at about 8.9, about 10.3, and about 11.0 degrees 2-theta. In some such embodiments, the present invention provides compound 3, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 3, wherein said compound has an XRPD substantially similar to that depicted in FIG. 28.


In some embodiments, the present invention provides a composition comprising compound 3 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 3 or composition thereof. In some such embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 3 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 4 (HCl Salts of Compound 1)


According to one embodiment, the present invention provides a hydrochloride salt of compound 1, represented by compound 4:




embedded image


It will be appreciated by one of ordinary skill in the art that the hydrochloric acid and compound 1 are ionically bonded to form compound 4. It is contemplated that compound 4 can exist in a variety of physical forms. For example, compound 4 can be in solution, suspension, or in solid form. In certain embodiments, compound 4 is in solid form. When compound 4 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 4 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess hydrochloric acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 4. In certain embodiments, at least about 95% by weight of compound 4 is present. In still other embodiments of the invention, at least about 99% by weight of compound 4 is present.


According to one embodiment, compound 4 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 4 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 4 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 4 is also meant to include all tautomeric forms of compound 4. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 4 can exist in a variety of solid forms. Exemplary such forms include polymorphs such as those contemplated by the present invention.


In certain embodiments, compound 4 is a crystalline solid. In other embodiments, compound 4 is a crystalline solid substantially free of amorphous compound 4. As used herein, the term “substantially free of amorphous compound 4” means that the compound contains no significant amount of amorphous compound 4. In certain embodiments, at least about 95% by weight of crystalline compound 4 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 4 is present.


It has been found that compound 4 can exist in at least nine distinct polymorphic forms. In some embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form A. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form B. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form C. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form D. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form E. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form F. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form G. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form H. In certain embodiments, the present invention provides a polymorphic form of compound 4 referred to herein as Form I.


In some embodiments, compound 4 is amorphous. In some embodiments, compound 4 is amorphous, and is substantially free of crystalline compound 4.


Form A of Compound 4


In some embodiments, Form A of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 10 below.









TABLE 10





XRPD Peak Positions for Form A of Compound 4


Position (°2 θ)















7.5


9.3


11.2


11.9


14.2


15.0


15.3


15.7


21.4


21.9


22.6


22.8


23.4


24.6


24.8


25.2


26.4


26.8


30.5


34.1






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 9.3, 15.7, and 24.8. In some embodiments, Form A of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 9.3, 15.7, and 24.8. In some embodiments, Form A of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 9.3, 15.7, and 24.8.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 32.


Methods for preparing Form A of compound 4 are described infra.


Form B of Compound 4


In some embodiments, Form B of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 11 below.









TABLE 11





XRPD Peak Positions for Form B of Compound 4


Position (°2 θ)















8.0


8.4


9.1


11.0


11.8


12.7


16.2


17.2


17.8


18.9


20.4


20.9


23.7


23.9


24.7


25.4


25.6


25.9


29.2


30.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.4, 12.7, and 17.8. In some embodiments, Form B of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.4, 12.7, and 17.8. In some embodiments, Form B of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.4, 12.7, and 17.8.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 36.


Methods for preparing Form B of compound 4 are described infra.


Form C of Compound 4


In some embodiments, Form C of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 12 below.









TABLE 12





XRPD Peak Positions for Form C of Compound 4


Position (°2 θ)















7.7


8.2


9.0


9.5


10.2


12.1


12.4


12.6


13.5


15.3


16.4


18.4


21.0


21.3


23.1


23.4


23.7


24.2


25.5


25.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.7, 8.2, and 9.0. In some embodiments, Form C of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.7, 8.2, and 9.0. In some embodiments, Form C of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.7, 8.2, and 9.0.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 38.


Methods for preparing Form C of compound 4 are described infra.


Form D of Compound 4


In some embodiments, Form D of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 13 below.









TABLE 13





XRPD Peak Positions for Form D of Compound 4


Position (°2 θ)















7.1


9.1


11.2


12.7


14.0


14.4


14.6


17.0


17.2


21.7


22.0


22.4


22.8


23.7


24.8


25.3


25.5


28.0


34.2


35.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form D of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.1, 9.1, and 11.2. In some embodiments, Form D of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.1, 9.1, and 11.2. In some embodiments, Form D of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.1, 9.1, and 11.2.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 40.


Methods for preparing Form D of compound 4 are described infra.


Form E of Compound 4


In some embodiments, Form E of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 14 below.









TABLE 14





XRPD Peak Positions for Form E of Compound 4


Position (°2 θ)















7.7


9.0


10.7


14.4


15.5


17.1


18.2


19.8


20.4


21.5


23.1


24.4


24.6


25.0


27.0


27.3


28.1


30.9


31.2


37.6






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form E of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.7, 10.7, and 17.1. In some embodiments, Form E of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.7, 10.7, and 17.1. In some embodiments, Form E of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.7, 10.7, and 17.1.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 44.


Methods for preparing Form E of compound 4 are described infra.


Form F of Compound 4


In some embodiments, Form F of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 15 below.









TABLE 15





XRPD Peak Positions for Form F of Compound 4


Position (°2 θ)















6.1


7.7


11.3


12.4


13.8


15.4


16.9


17.7


18.6


19.5


22.6


22.9


23.1


23.6


24.9


25.7


26.1


30.2


34.2


35.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form F of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 6.1, 11.3, and 18.6. In some embodiments, Form F of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 6.1, 11.3, and 18.6. In some embodiments, Form F of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 6.1, 11.3, and 18.6.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 46.


Methods for preparing Form F of compound 4 are described infra.


Form G of Compound 4


In some embodiments, Form G of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 16 below.









TABLE 16





XRPD Peak Positions for Form G of Compound 4


Position (°2 θ)















5.2


8.5


8.7


10.5


14.8


15.6


19.0


19.5


20.7


21.0


21.9


22.2


23.3


25.7


27.0


27.4


29.6


30.7


31.3






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form G of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 5.2, 8.7, and 19.5. In some embodiments, Form G of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 5.2, 8.7, and 19.5. In some embodiments, Form G of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 5.2, 8.7, and 19.5.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 48.


Methods for preparing Form G of compound 4 are described infra.


Form H of Compound 4


In some embodiments, Form H of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 17 below.









TABLE 17





XRPD Peak Positions for Form H of Compound 4


Position (°2 θ)















7.2


8.5


9.1


10.2


13.1


14.5


15.8


18.1


18.8


20.5


21.0


21.7


22.5


23.3


24.2


24.6


25.3


25.8


28.8


29.9






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form H of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.5, 9.1, and 10.2. In some embodiments, Form H of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.5, 9.1, and 10.2. In some embodiments, Form H of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.5, 9.1, and 10.2.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 50.


Methods for preparing Form H of compound 4 are described infra.


Form I of Compound 4


In some embodiments, Form I of compound 4 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 18 below.









TABLE 18





XRPD Peak Positions for Form I of Compound 4


Position (°2 θ)















4.7


7.1


9.3


10.8


11.9


14.1


15.0


16.7


18.8


20.3


21.1


22.2


23.1


23.5


24.2


25.4


26.0


29.1






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form I of compound 4 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 4.7, 7.1, and 9.3. In some embodiments, Form I of compound 4 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 4.7, 7.1, and 9.3. In some embodiments, Form I of compound 4 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 4.7, 7.1, and 9.3.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 52.


Methods for preparing Form I of compound 4 are described infra.


In some embodiments, the present invention provides compound 4:




embedded image


In some embodiments, the present invention provides compound 4, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 4, wherein said compound is a crystalline solid substantially free of amorphous compound 4.


In some embodiments, the present invention provides compound 4, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 9.3, about 15.7, and about 24.8 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 9.3, about 15.7, and about 24.8 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 32.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 8.4, about 12.7, and about 17.8 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 8.4, about 12.7, and about 17.8 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 36.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 7.7, about 8.2, and about 9.0 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 7.7, about 8.2, and about 9.0 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 38.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 7.1, about 9.1, and about 11.2 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 7.1, about 9.1, and about 11.2 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form D.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 40.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 7.7, about 10.7, and about 17.1 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 7.7, about 10.7, and about 17.1 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form E.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 44.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 6.1, about 11.3, and about 18.6 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 6.1, about 11.3, and about 18.6 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form F.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 46.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 5.2, about 8.7, and about 19.5 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 5.2, about 8.7, and about 19.5 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form G.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 48.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 8.5, about 9.1, and about 10.2 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 8.5, about 9.1, and about 10.2 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form H.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 50.


In some embodiments, the present invention provides compound 4, wherein said compound has one or more peaks in its XRPD selected from those at about 4.7, about 7.1, and about 9.3 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound has at least two peaks in its XRPD selected from those at about 4.7, about 7.1, and about 9.3 degrees 2-theta. In some such embodiments, the present invention provides compound 4, wherein said compound is of Form I.


In some embodiments, the present invention provides compound 4, wherein said compound has an XRPD substantially similar to that depicted in FIG. 52.


In some embodiments, the present invention provides a composition comprising compound 4 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 4 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 4 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 5 (HBr Salts of Compound 1)


According to one embodiment, the present invention provides a hydrobromide salt of compound 1, represented by compound 5:




embedded image


It will be appreciated by one of ordinary skill in the art that the hydrobromic acid and compound 1 are ionically bonded to form compound 5. It is contemplated that compound 5 can exist in a variety of physical forms. For example, compound 5 can be in solution, suspension, or in solid form. In certain embodiments, compound 5 is in solid form. When compound 5 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 5 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess hydrobromic acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 5. In certain embodiments, at least about 95% by weight of compound 5 is present. In still other embodiments of the invention, at least about 99% by weight of compound 5 is present.


According to one embodiment, compound 5 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 5 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 5 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 5 is also meant to include all tautomeric forms of compound 5. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 5 can exist in a variety of solid forms. Exemplary such forms include polymorphs such as those contemplated by the present invention.


In certain embodiments, compound 5 is a crystalline solid. In other embodiments, compound 5 is a crystalline solid substantially free of amorphous compound 5. As used herein, the term “substantially free of amorphous compound 5” means that the compound contains no significant amount of amorphous compound 5. In certain embodiments, at least about 95% by weight of crystalline compound 5 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 5 is present.


It has been found that compound 5 can exist in at least five distinct polymorphic forms. In some embodiments, the present invention provides a polymorphic form of compound 5 referred to herein as Form A. In certain embodiments, the present invention provides a polymorphic form of compound 5 referred to herein as Form B. In certain embodiments, the present invention provides a polymorphic form of compound 5 referred to herein as Form C. In certain embodiments, the present invention provides a polymorphic form of compound 5 referred to herein as Form D. In certain embodiments, the present invention provides a polymorphic form of compound 5 referred to herein as Form E.


In some embodiments, compound 5 is amorphous. In some embodiments, compound 5 is amorphous, and is substantially free of crystalline compound 5.


Form A of Compound 5


In some embodiments, Form A of compound 5 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 19 below.









TABLE 19





XRPD Peak Positions for Form A of Compound 5


Position (°2 θ)















7.5


9.5


11.3


13.0


14.3


14.7


15.0


15.7


17.3


20.3


20.9


21.6


22.6


23.2


23.7


24.8


26.0


28.8


30.5


33.8






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 5 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 9.5, 22.6, and 24.8. In some embodiments, Form A of compound 5 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 9.5, 22.6, and 24.8. In some embodiments, Form A of compound 5 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 9.5, 22.6, and 24.8.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 53.


Methods for preparing Form A of compound 5 are described infra.


Form B of Compound 5


In some embodiments, Form B of compound 5 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 20 below.









TABLE 20





XRPD Peak Positions for Form B of Compound 5


Position (°2 θ)















8.3


11.8


12.3


14.2


16.2


17.9


18.7


18.9


20.3


20.5


20.8


21.1


23.8


24.5


25.5


25.7


26.3


29.2


30.7


31.9






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 5 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.3, 17.9, and 25.5. In some embodiments, Form B of compound 5 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.3, 17.9, and 25.5. In some embodiments, Form B of compound 5 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.3, 17.9, and 25.5.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 57.


Methods for preparing Form B of compound 5 are described infra.


Form C of Compound 5


In some embodiments, Form C of compound 5 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 21 below.









TABLE 21





XRPD Peak Positions for Form C of Compound 5


Position (°2 θ)















7.4


8.4


10.5


15.4


15.8


17.4


20.1


20.8


22.1


22.3


22.6


23.2


24.2


24.5


25.9


26.6


29.0


31.8


33.8


37.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 5 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.4, 8.4, and 10.5. In some embodiments, Form C of compound 5 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.4, 8.4, and 10.5. In some embodiments, Form C of compound 5 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.4, 8.4, and 10.5.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 59.


Methods for preparing Form C of compound 5 are described infra.


Form D of Compound 5


In some embodiments, Form D of compound 5 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 22 below.









TABLE 22





XRPD Peak Positions for Form D of Compound 5


Position (°2 θ)















7.2


9.4


10.8


11.8


15.1


16.9


18.9


20.4


21.6


22.3


23.7


24.3


24.7


25.5


26.1


29.0


30.3


30.6


32.1


35.2






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form D of compound 5 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.2, 10.8, and 24.3. In some embodiments, Form D of compound 5 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.2, 10.8, and 24.3. In some embodiments, Form D of compound 5 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.2, 10.8, and 24.3.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 61.


Methods for preparing Form D of compound 5 are described infra.


Form E of Compound 5


In some embodiments, Form E of compound 5 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 23 below.









TABLE 23





XRPD Peak Positions for Form E of Compound 5


Position (°2 θ)















7.8


8.7


10.0


11.6


15.0


16.4


17.5


18.7


21.5


22.8


23.8


24.3


25.2


25.8


28.8


29.1


30.3


31.7


33.7


36.8






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form E of compound 5 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.8, 8.7, and 16.4. In some embodiments, Form E of compound 5 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.8, 8.7, and 16.4. In some embodiments, Form E of compound 5 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.8, 8.7, and 16.4.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 63.


Methods for preparing Form E of compound 5 are described infra.


In some embodiments, the present invention provides compound 5:




embedded image


In some embodiments, the present invention provides compound 5, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 5, wherein said compound is a crystalline solid substantially free of amorphous compound 5.


In some embodiments, the present invention provides compound 5, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 5, wherein said compound has one or more peaks in its XRPD selected from those at about 9.5 about 22.6, and about 24.8 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound has at least two peaks in its XRPD selected from those at about 9.5, about 22.6, and about 24.8 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 5, wherein said compound has an XRPD substantially similar to that depicted in FIG. 53.


In some embodiments, the present invention provides compound 5, wherein said compound has one or more peaks in its XRPD selected from those at about 8.3, about 17.9, and about 25.5 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound has at least two peaks in its XRPD selected from those at about 8.3, about 17.9, and about 25.5 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 5, wherein said compound has an XRPD substantially similar to that depicted in FIG. 57.


In some embodiments, the present invention provides compound 5, wherein said compound has one or more peaks in its XRPD selected from those at about 7.4, about 8.4, and about 10.5 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound has at least two peaks in its XRPD selected from those at about 7.4, about 8.4, and about 10.5 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 5, wherein said compound has an XRPD substantially similar to that depicted in FIG. 59.


In some embodiments, the present invention provides compound 5, wherein said compound has one or more peaks in its XRPD selected from those at about 7.2, about 10.8, and about 24.3 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound has at least two peaks in its XRPD selected from those at about 7.2, about 10.8, and about 24.3 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound is of Form D.


In some embodiments, the present invention provides compound 5, wherein said compound has an XRPD substantially similar to that depicted in FIG. 61.


In some embodiments, the present invention provides compound 5, wherein said compound has one or more peaks in its XRPD selected from those at about 7.8, about 8.7, and about 16.4 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound has at least two peaks in its XRPD selected from those at about 7.8, about 8.7, and about 16.4 degrees 2-theta. In some such embodiments, the present invention provides compound 5, wherein said compound is of Form E.


In some embodiments, the present invention provides compound 5, wherein said compound has an XRPD substantially similar to that depicted in FIG. 63.


In some embodiments, the present invention provides a composition comprising compound 5 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 5 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 5 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 6 (Sulfate Salts of Compound 1)


According to one embodiment, the present invention provides a sulfate salt of compound 1, represented by compound 6:




embedded image


It will be appreciated by one of ordinary skill in the art that the sulfuric acid and compound 1 are ionically bonded to form compound 6. It is contemplated that compound 6 can exist in a variety of physical forms. For example, compound 6 can be in solution, suspension, or in solid form. In certain embodiments, compound 6 is in solid form. When compound 6 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 5 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess sulfuric acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 6. In certain embodiments, at least about 95% by weight of compound 6 is present. In still other embodiments of the invention, at least about 99% by weight of compound 6 is present.


According to one embodiment, compound 6 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 6 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 6 contains no more than about a percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 6 is also meant to include all tautomeric forms of compound 6. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 6 can exist in a variety of solid forms. Exemplary such forms include polymorphs such as those contemplated by the present invention.


In certain embodiments, compound 6 is a crystalline solid. In other embodiments, compound 6 is a crystalline solid substantially free of amorphous compound 6. As used herein, the term “substantially free of amorphous compound 6” means that the compound contains no significant amount of amorphous compound 6. In certain embodiments, at least about 95% by weight of crystalline compound 6 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 6 is present.


It has been found that compound 6 can exist in at least three distinct polymorphic forms. In some embodiments, the present invention provides a polymorphic form of compound 6 referred to herein as Form A. In certain embodiments, the present invention provides a polymorphic form of compound 6 referred to herein as Form B. In certain embodiments, the present invention provides a polymorphic form of compound 6 referred to herein as Form C.


In some embodiments, compound 6 is amorphous. In some embodiments, compound 6 is amorphous, and is substantially free of crystalline compound 6.


Form A of Compound 6


In some embodiments, Form A of compound 6 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 24 below.









TABLE 24





XRPD Peak Positions for Form A of Compound 6


Position (°2 θ)















6.2


7.1


9.9


14.2


14.7


19.4


19.5


20.1


20.2


20.7


21.4


23.4


23.9


24.3


24.8


25.3


26.0


26.9


28.7


29.8






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 6 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 6.2, 7.1, and 21.4. In some embodiments, Form A of compound 6 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 6.2, 7.1, and 21.4. In some embodiments, Form A of compound 6 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 6.2, 7.1, and 21.4.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 66.


Methods for preparing Form A of compound 6 are described infra.


Form B of Compound 6


In some embodiments, Form B of compound 6 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 25 below.









TABLE 25





XRPD Peak Positions for Form B of Compound 6


Position (°2 θ)















7.1


7.6


10.1


11.4


11.6


12.4


13.7


15.2


17.3


17.8


18.7


18.7


20.3


21.7


22.9


24.1


25.4


26.0


29.7


34.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 6 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.6, 15.2, and 22.9. In some embodiments, Form B of compound 6 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.6, 15.2, and 22.9. In some embodiments, Form B of compound 6 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.6, 15.2, and 22.9.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 70.


Methods for preparing Form B of compound 6 are described infra.


Form C of Compound 6


In some embodiments, Form C of compound 6 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 26 below.









TABLE 26





XRPD Peak Positions for Form C of Compound 6


Position (°2 θ)















7.1


7.6


8.3


9.3


12.6


13.5


14.2


17.4


18.1


19.6


20.2


20.5


20.9


21.2


23.1


24.4


25.1


26.2


27.9


29.9






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 6 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.1, 7.6, and 23.1. In some embodiments, Form C of compound 6 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.1, 7.6, and 23.1. In some embodiments, Form C of compound 6 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.1, 7.6, and 23.1.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 72.


Methods for preparing Form C of compound 6 are described infra.


In some embodiments, the present invention provides compound 6:




embedded image


In some embodiments, the present invention provides compound 6, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 6, wherein said compound is a crystalline solid substantially free of amorphous compound 6.


In some embodiments, the present invention provides compound 6, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 6, wherein said compound has one or more peaks in its XRPD selected from those at about 6.2, 7.1, about and about 21.4 degrees 2-theta. In some such embodiments, the present invention provides compound 6, wherein said compound has at least two peaks in its XRPD selected from those at about 6.2, 7.1, about and about 21.4 degrees 2-theta. In some such embodiments, the present invention provides compound 6, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 6, wherein said compound has an XRPD substantially similar to that depicted in FIG. 66.


In some embodiments, the present invention provides compound 6, wherein said compound has one or more peaks in its XRPD selected from those at about 7.6, about 15.2, and about 22.9 degrees 2-theta. In some such embodiments, the present invention provides compound 6, wherein said compound has at least two peaks in its XRPD selected from those at about 7.6, about 15.2, and about 22.9 degrees 2-theta. In some such embodiments, the present invention provides compound 6, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 6, wherein said compound has an XRPD substantially similar to that depicted in FIG. 70.


In some embodiments, the present invention provides compound 6, wherein said compound has one or more peaks in its XRPD selected from those at about 7.1, about 7.6, and about 23.1 degrees 2-theta. In some such embodiments, the present invention provides compound 6, wherein said compound has at least two peaks in its XRPD selected from those at about 7.1, about 7.6, and about 23.1 degrees 2-theta. In some such embodiments, the present invention provides compound 6, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 6, wherein said compound has an XRPD substantially similar to that depicted in FIG. 72.


In some embodiments, the present invention provides a composition comprising compound 6 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 6 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 6 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 7 (Bis-Sulfate Salts of Compound 1)


According to one embodiment, the present invention provides a bis-sulfate salt of compound 1, represented by compound 7:




embedded image


It will be appreciated by one of ordinary skill in the art that sulfuric acid and compound 1 are ionically bonded to form compound 7. It is contemplated that compound 7 can exist in a variety of physical forms. For example, compound 7 can be in solution, suspension, or in solid form. In certain embodiments, compound 7 is in solid form. When compound 7 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 7 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess sulfuric acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 7. In certain embodiments, at least about 95% by weight of compound 7 is present. In still other embodiments of the invention, at least about 99% by weight of compound 7 is present.


According to one embodiment, compound 7 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 7 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 7 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 7 is also meant to include all tautomeric forms of compound 7. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


In certain embodiments, compound 7 is a crystalline solid. In other embodiments, compound 7 is a crystalline solid substantially free of amorphous compound 7. As used herein, the term “substantially free of amorphous compound 7” means that the compound contains no significant amount of amorphous compound 7. In certain embodiments, at least about 95% by weight of crystalline compound 7 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 7 is present.


It has been found that compound 7 can exist in at least one distinct crystalline form. In some embodiments, the present invention provides a crystalline form of compound 7 referred to herein as Form A.


In some embodiments, compound 7 is amorphous. In some embodiments, compound 7 is amorphous, and is substantially free of crystalline compound 7.


Form A of Compound 7


In some embodiments, Form A of compound 7 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 27 below.









TABLE 27





XRPD Peak Positions for Form A of Compound 7


Position (°2 θ)















7.3


8.7


9.6


11.4


11.5


14.1


17.9


18.2


19.0


19.2


20.5


20.7


21.0


22.2


23.5


24.3


24.3


27.5


28.6


31.1






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 7 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 7.3, 8.7, and 23.5. In some embodiments, Form A of compound 7 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 7.3, 8.7, and 23.5. In some embodiments, Form A of compound 7 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 7.3, 8.7, and 23.5.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 74.


Methods for preparing Form A of compound 7 are described infra.


In some embodiments, the present invention provides compound 7:




embedded image


In some embodiments, the present invention provides compound 7, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 7, wherein said compound is a crystalline solid substantially free of amorphous compound 7.


In some embodiments, the present invention provides compound 7, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 7, wherein said compound has one or more peaks in its XRPD selected from those at about 7.3, about 8.7, and about 23.5 degrees 2-theta. In some such embodiments, the present invention provides compound 7, wherein said compound has at least two peaks in its XRPD selected from those at about 7.3, about 8.7, and about 23.5 degrees 2-theta. In some such embodiments, the present invention provides compound 7, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 7, wherein said compound has an XRPD substantially similar to that depicted in FIG. 74.


In some embodiments, the present invention provides a composition comprising the compound 7 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 7 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 7 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 8 (Tosylate Salts of Compound 1)


According to one embodiment, the present invention provides a tosylate salt of compound 1, represented by compound 8:




embedded image


By “tosylate” is meant p-toluene sulfonate, i.e., the ionic form of p-toluenesulfonic acid. It will be appreciated by one of ordinary skill in the art that p-toluenesulfonic acid and compound 1 are ionically bonded to form compound 8. It is contemplated that compound 8 can exist in a variety of physical forms. For example, compound 8 can be in solution, suspension, or in solid form. In certain embodiments, compound 8 is in solid form. When compound 8 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 8 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess p-toluenesulfonic acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 8. In certain embodiments, at least about 95% by weight of compound 8 is present. In still other embodiments of the invention, at least about 99% by weight of compound 8 is present.


According to one embodiment, compound 8 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 8 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 8 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 8 is also meant to include all tautomeric forms of compound 8. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 8 can exist in a variety of solid forms. Exemplary such forms include polymorphs and amorphous forms such as those contemplated by the present invention.


In certain embodiments, compound 8 is a crystalline solid. In other embodiments, compound 8 is a crystalline solid substantially free of amorphous compound 8. As used herein, the term “substantially free of amorphous compound 8” means that the compound contains no significant amount of amorphous compound 8. In certain embodiments, at least about 95% by weight of crystalline compound 8 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 8 is present.


In certain embodiments, compound 8 is an amorphous solid. In certain embodiments, at least about 95% by weight of amorphous compound 8 is present. In still other embodiments of the invention, at least about 99% by weight of amorphous compound 8 is present.


It has been found that compound 8 can exist in at least four distinct forms, three of which are polymorphic and one of which is amorphous. In some embodiments, the present invention provides a polymorphic form of compound 8 referred to herein as Form A. In some embodiments, the present invention provides a polymorphic form of compound 8 referred to herein as Form B. In some embodiments, the present invention provides a polymorphic form of compound 8 referred to herein as Form C. In some embodiments, the present invention provides an amorphous form of compound 8 referred to herein as Form D.


In some embodiments, compound 8 is amorphous. In some embodiments, compound 8 is amorphous, and is substantially free of crystalline compound 8.


Form A of Compound 8


In some embodiments, Form A of compound 8 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 28 below.









TABLE 28





XRPD Peak Positions for Form A of Compound 8


Position (°2 θ)















8.6


9.0


12.4


12.8


13.4


16.1


16.4


16.6


17.0


17.9


19.1


20.0


20.5


22.9


23.5


23.7


23.8


24.8


27.8


30.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 8 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 9.0, 23.8, and 24.8. In some embodiments, Form A of compound 8 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 9.0, 23.8, and 24.8. In some embodiments, Form A of compound 8 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 9.0, 23.8, and 24.8.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 77.


Methods for preparing Form A of compound 8 are described infra.


Form B of Compound 8


In some embodiments, Form B of compound 8 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 29 below.









TABLE 29





XRPD Peak Positions for Form B of Compound 8


Position (°2 θ)















8.6


8.9


12.2


16.2


16.5


16.9


17.2


17.9


19.0


19.1


20.1


23.1


23.4


23.6


24.6


24.7


25.1


26.3


27.6


28.2






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 8 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 23.4, and 27.6. In some embodiments, Form B of compound 8 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 23.4, and 27.6. In some embodiments, Form B of compound 8 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 23.4, and 27.6.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 79.


Methods for preparing Form B of compound 8 are described infra.


Form C of Compound 8


In some embodiments, Form C of compound 8 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 30 below.









TABLE 30







XRPD Peak Positions for Form C of Compound 8










Peak No.
Position (°2 θ)














8.8
23.1



9.5
23.8



12.5
24.2



13.3
24.6



14.2
25.2



16.9
26.3



17.4
30.3



17.8
31.1



18.9
31.8



20.1
38.0








1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 8 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.8, 16.9, and 24.2. In some embodiments, Form C of compound 8 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.8, 16.9, and 24.2. In some embodiments, Form C of compound 8 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.8, 16.9, and 24.2.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 81.


Methods for preparing Form C of compound 8 are described infra.


Form D of Compound 8


In some embodiments, compound 8 is an amorphous solid. For instance, in some embodiments, compound 8 is of Form D.


In certain embodiments, Form D of compound 8 is characterized by having a DSC thermogram substantially similar to that of FIG. 84.


In certain embodiments, Form D of compound 8 is characterized by having a TGA trace substantially similar to that of FIG. 84.


Methods for preparing Form D of compound 8 are described infra.


In some embodiments, the present invention provides compound 8:




embedded image


In some embodiments, the present invention provides compound 8, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 8, wherein said compound is a crystalline solid substantially free of amorphous compound 8.


In some embodiments, the present invention provides compound 8, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 8, wherein said compound has one or more peaks in its XRPD selected from those at about 9.0, about 23.8, and about 24.8 degrees 2-theta. In some such embodiments, the present invention provides compound 8, wherein said compound has at least two peaks in its XRPD selected from those at about 9.0, about 23.8, and about 24.8 degrees 2-theta. In some such embodiments, the present invention provides compound 8, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 8, wherein said compound has an XRPD substantially similar to that depicted in FIG. 77.


In some embodiments, the present invention provides compound 8, wherein said compound has one or more peaks in its XRPD selected from those at about 8.9, about 23.4, and about 27.6 degrees 2-theta. In some such embodiments, the present invention provides compound 8, wherein said compound has at least two peaks in its XRPD selected from those at about 8.9, about 23.4, and about 27.6 degrees 2-theta. In some such embodiments, the present invention provides compound 8, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 8, wherein said compound has an XRPD substantially similar to that depicted in FIG. 79.


In some embodiments, the present invention provides compound 8, wherein said compound has one or more peaks in its XRPD selected from those at about 8.8, about 16.9, and about 24.2 degrees 2-theta. In some such embodiments, the present invention provides compound 8, wherein said compound has at least two peaks in its XRPD selected from those at about 8.8, about 16.9, and about 24.2 degrees 2-theta. In some such embodiments, the present invention provides compound 8, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 8, wherein said compound has a XRPD substantially similar to that depicted in FIG. 81.


In some embodiments, the present invention provides compound 8, wherein said compound has a DSC thermogram substantially similar to that depicted in FIG. 84. In some such embodiments, the present invention provides compound 8, wherein said compound is of Form D.


In some embodiments, the present invention provides a composition comprising compound 8 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 8 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 8 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 9 (Besylate Salts of Compound 1)


According to one embodiment, the present invention provides a besylate salt of compound 1, represented by compound 9:




embedded image


It will be appreciated by one of ordinary skill in the art that benzenesulfonic acid and compound 1 are ionically bonded to form compound 9. It is contemplated that compound 9 can exist in a variety of physical forms. For example, compound 9 can be in solution, suspension, or in solid form. In certain embodiments, compound 9 is in solid form. When compound 9 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 9 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess benzenesulfonic acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 9. In certain embodiments, at least about 95% by weight of compound 9 is present. In still other embodiments of the invention, at least about 99% by weight of compound 9 is present.


According to one embodiment, compound 9 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 9 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 9 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 9 is also meant to include all tautomeric forms of compound 9. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 9 can exist in a variety of solid forms. Exemplary such forms include polymorphs and amorphous forms such as those contemplated by the present invention.


In certain embodiments, compound 9 is a crystalline solid. In other embodiments, compound 9 is a crystalline solid substantially free of amorphous compound 9. As used herein, the term “substantially free of amorphous compound 9” means that the compound contains no significant amount of amorphous compound 9. In certain embodiments, at least about 95% by weight of crystalline compound 9 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 9 is present.


In certain embodiments, compound 9 is an amorphous solid. In certain embodiments, at least about 95% by weight of amorphous compound 9 is present. In still other embodiments of the invention, at least about 99% by weight of amorphous compound 9 is present.


It has been found that compound 9 can exist in at least two distinct forms, one of which is crystalline and one of which is amorphous. In some embodiments, the present invention provides a crystalline form of compound 9 referred to herein as Form A. In some embodiments, the present invention provides an amorphous form of compound 9 referred to herein as Form B.


In some embodiments, compound 9 is amorphous. In some embodiments, compound 9 is amorphous, and is substantially free of crystalline compound 9.


Form A of Compound 9


In some embodiments, Form A of compound 9 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 31 below.









TABLE 31







XRPD Peak Positions for Form A of Compound 9










Peak No.
Position (°2 θ)














8.9
23.6



12.5
24.0



13.2
25.2



15.6
25.5



17.6
26.1



18.0
27.0



18.5
27.9



19.1
29.6



20.6
30.3



21.7
39.5








1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 9 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 18.5, and 25.2. In some embodiments, Form A of compound 9 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 18.5, and 25.2. In some embodiments, Form A of compound 9 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 18.5, and 25.2.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 85.


Methods for preparing Form A of compound 9 are described infra.


Form B of Compound 9


In some embodiments, compound 9 is an amorphous solid. For instance, in some embodiments, compound 9 is of Form B.


In certain embodiments, Form B of compound 9 is characterized by having a DSC thermogram substantially similar to that of FIG. 88.


In certain embodiments, Form B of compound 9 is characterized by having a TGA trace substantially similar to that of FIG. 88.


Methods for preparing Form B of compound 9 are described infra.


In some embodiments, the present invention provides compound 9:




embedded image


In some embodiments, the present invention provides compound 9, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 9, wherein said compound is a crystalline solid substantially free of amorphous compound 9.


In some embodiments, the present invention provides compound 9, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 9, wherein said compound has one or more peaks in its XRPD selected from those at about 8.9, about 18.5, and about 25.2 degrees 2-theta. In some such embodiments, the present invention provides compound 9, wherein said compound has at least two peaks in its XRPD selected from those at about 8.9, about 18.5, and about 25.2 degrees 2-theta. In some such embodiments, the present invention provides compound 9, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 9, wherein said compound has a XRPD substantially similar to that depicted in FIG. 85.


In some embodiments, the present invention provides compound 9, wherein said compound has a DSC thermogram substantially similar to that depicted in FIG. 88. In some such embodiments, the present invention provides compound 9, wherein said compound is of Form B.


In some embodiments, the present invention provides a composition comprising compound 9 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 9 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 9 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 10 (Mesylate Salts of Compound 1)


According to one embodiment, the present invention provides a mesylate salt of compound 1, represented by compound 10:




embedded image


It will be appreciated by one of ordinary skill in the art that methanesulfonic acid and compound 1 are ionically bonded to form compound 10. It is contemplated that compound 10 can exist in a variety of physical forms. For example, compound 10 can be in solution, suspension, or in solid form. In certain embodiments, compound 10 is in solid form. When compound 10 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 10 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess methanesulfonic acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 10. In certain embodiments, at least about 95% by weight of compound 10 is present. In still other embodiments of the invention, at least about 99% by weight of compound 10 is present.


According to one embodiment, compound 10 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 10 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 10 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 10 is also meant to include all tautomeric forms of compound 10. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 10 can exist in a variety of solid forms. Exemplary such forms include partially crystalline forms and amorphous forms such as those contemplated by the present invention.


In certain embodiments, compound 10 is a crystalline solid. In other embodiments, compound 10 is a crystalline solid substantially free of amorphous compound 10. As used herein, the term “substantially free of amorphous compound 10” means that the compound contains no significant amount of amorphous compound 10. In certain embodiments, at least about 95% by weight of crystalline compound 10 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 10 is present.


In some embodiments, compound 10 is a partially crystalline solid.


In certain embodiments, compound 10 is an amorphous solid. In certain embodiments, at least about 95% by weight of amorphous compound 10 is present. In still other embodiments of the invention, at least about 99% by weight of amorphous compound 10 is present.


It has been found that compound 10 can exist in at least two forms, one of which is amorphous and the other of which is partially crystalline. In some embodiments, the present invention provides an amorphous form of compound 10 referred to herein as Form A. In some embodiments, the present invention provides a partially crystalline form of compound 10 referred to herein as Form B.


In some embodiments, compound 10 is amorphous. In some embodiments, compound 10 is amorphous, and is substantially free of crystalline compound 10.


Form A of Compound 10


In some embodiments, compound 10 is an amorphous solid. For instance, in some embodiments, compound 10 is of Form A.


In certain embodiments, Form A of compound 10 is characterized by having a DSC thermogram substantially similar to that of FIG. 89.


In certain embodiments, Form A of compound 10 is characterized by having a TGA trace substantially similar to that of FIG. 89.


Methods for preparing Form A of compound 10 are described infra.


Form B of Compound 10


In some embodiments, Form B of compound 10 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 32 below.









TABLE 32





XRPD Peak Positions for Form B of Compound 10


Position (°2 θ)















6.1


7.9


8.3


16.3


22.6






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 10 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 6.1, 8.3, and 22.6. In some embodiments, Form B of compound 10 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 6.1, 8.3, and 22.6. In some embodiments, Form B of compound 10 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 6.1, 8.3, and 22.6.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 90.


Methods for preparing Form B of compound 10 are described infra.


In some embodiments, the present invention provides compound 10:




embedded image


In some embodiments, the present invention provides compound 10, wherein said compound is crystalline. In some embodiments, the present invention provides compound 10, wherein said compound is partially crystalline.


In some embodiments, the present invention provides compound 10, wherein said compound is a crystalline solid substantially free of amorphous compound 10.


In some embodiments, the present invention provides compound 10, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 10, wherein said compound has a DSC thermogram substantially similar to that depicted in FIG. 89. In some such embodiments, the present invention provides compound 10, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 10, wherein said compound has one or more peaks in its XRPD selected from those at about 6.1, about 8.3, and about 22.6 degrees 2-theta. In some such embodiments, the present invention provides compound 10, wherein said compound has at least two peaks in its XRPD selected from those at about 6.1, about 8.3, and about 22.6 degrees 2-theta. In some such embodiments, the present invention provides compound 10, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 10, wherein said compound has an XRPD substantially similar to that depicted in FIG. 90.


In some embodiments, the present invention provides compound 10, wherein said compound has a DSC thermogram substantially similar to that depicted in FIG. 91.


In some embodiments, the present invention provides a composition comprising compound 10 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 10 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 10 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 11 (Maleate Salts of Compound 1)


According to one embodiment, the present invention provides a maleate salt of compound 1, represented by compound 11:




embedded image


It will be appreciated by one of ordinary skill in the art that maleic acid and compound 1 are ionically bonded to form compound 11. It is contemplated that compound 11 can exist in a variety of physical forms. For example, compound 11 can be in solution, suspension, or in solid form. In certain embodiments, compound 11 is in solid form. When compound 11 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 11 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess maleic acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 11. In certain embodiments, at least about 95% by weight of compound 11 is present. In still other embodiments of the invention, at least about 99% by weight of compound 11 is present.


According to one embodiment, compound 11 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 11 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 11 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 11 is also meant to include all tautomeric forms of compound 11. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


In certain embodiments, compound 11 is a crystalline solid. In other embodiments, compound 11 is a crystalline solid substantially free of amorphous compound 11. As used herein, the term “substantially free of amorphous compound 11” means that the compound contains no significant amount of amorphous compound 11. In certain embodiments, at least about 95% by weight of crystalline compound 11 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 11 is present.


It has been found that compound 11 can exist in at least one distinct crystalline form. In some embodiments, the present invention provides a crystalline form of compound 11 referred to herein as Form A.


In some embodiments, compound 11 is amorphous. In some embodiments, compound 11 is amorphous, and is substantially free of crystalline compound 11.


Form A of Compound 11


In some embodiments, Form A of compound 11 has at least 1, 2, 3, 4 or 5 spectral peak(s) is or are selected from the peaks listed in Table 33 below.









TABLE 33





XRPD Peak Positions for Form A of Compound 11


Position (°2 θ)















5.2


8.9


9.2


10.4


12.0


13.3


13.4


15.3


16.1


18.7


20.9


23.0


23.2


23.9


24.6


25.6


26.1


26.3


26.7


27.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 11 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 9.2, and 16.1. In some embodiments, Form A of compound 11 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 9.2, and 16.1. In some embodiments, Form A of compound 11 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 8.9, 9.2, and 16.1.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 92.


Methods for preparing Form A of compound 11 are described infra.


In some embodiments, the present invention provides compound 11:




embedded image


In some embodiments, the present invention provides compound 11, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 11, wherein said compound is a crystalline solid substantially free of amorphous compound 11.


In some embodiments, the present invention provides compound 11, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 11, wherein said compound has one or more peaks in its XRPD selected from those at about 8.9, about 9.2, and about 16.1 degrees 2-theta. In some such embodiments, the present invention provides compound 11, wherein said compound has at least two peaks in its XRPD selected from those at about 8.9, about 9.2, and about 16.1 degrees 2-theta. In some such embodiments, the present invention provides compound 11, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 11, wherein said compound has an XRPD substantially similar to that depicted in FIG. 92.


In some embodiments, the present invention provides a composition comprising compound 11 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 11 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 11 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


Compound 12 (Oxalate Salts of Compound 1)


According to one embodiment, the present invention provides an oxalate salt of compound 1, represented by compound 12:




embedded image


It will be appreciated by one of ordinary skill in the art that oxalic acid and compound 1 are ionically bonded to form compound 12. It is contemplated that compound 12 can exist in a variety of physical forms. For example, compound 12 can be in solution, suspension, or in solid form. In certain embodiments, compound 12 is in solid form. When compound 12 is in solid form, said compound may be amorphous, crystalline, or a mixture thereof. Exemplary solid forms are described in more detail below.


In some embodiments, the present invention provides compound 12 substantially free of impurities. As used herein, the term “substantially free of impurities” means that the compound contains no significant amount of extraneous matter. Such extraneous matter may include excess oxalic acid, excess compound 1, residual solvents, or any other impurities that may result from the preparation of, and/or isolation of, compound 12. In certain embodiments, at least about 95% by weight of compound 12 is present. In still other embodiments of the invention, at least about 99% by weight of compound 12 is present.


According to one embodiment, compound 12 is present in an amount of at least about 97, 97.5, 98.0, 98.5, 99, 99.5, 99.8 weight percent where the percentages are based on the total weight of the composition. According to another embodiment, compound 12 contains no more than about 3.0 area percent HPLC of total organic impurities and, in certain embodiments, no more than about 1.5 area percent HPLC total organic impurities relative to the total area of the HPLC chromatogram. In other embodiments, compound 12 contains no more than about 1.0% area percent HPLC of any single impurity; no more than about 0.6 area percent HPLC of any single impurity, and, in certain embodiments, no more than about 0.5 area percent HPLC of any single impurity, relative to the total area of the HPLC chromatogram.


The structure depicted for compound 12 is also meant to include all tautomeric forms of compound 12. Additionally, structures depicted here are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structure except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention.


It has been found that compound 12 can exist in a variety of solid forms. Exemplary such forms include polymorphs forms such as those contemplated by the present invention.


In certain embodiments, compound 12 is a crystalline solid. In other embodiments, compound 12 is a crystalline solid substantially free of amorphous compound 12. As used herein, the term “substantially free of amorphous compound 12” means that the compound contains no significant amount of amorphous compound 12. In certain embodiments, at least about 95% by weight of crystalline compound 12 is present. In still other embodiments of the invention, at least about 99% by weight of crystalline compound 12 is present.


It has been found that compound 12 can exist in at least three distinct polymorphic forms. In some embodiments, the present invention provides a polymorphic form of compound 12 referred to herein as Form A. In some embodiments, the present invention provides a polymorphic form of compound 12 referred to herein as Form B. In some embodiments, the present invention provides a polymorphic form of compound 12 referred to herein as Form C.


In some embodiments, compound 12 is amorphous. In some embodiments, compound 12 is amorphous, and is substantially free of crystalline compound 12.


Form A of Compound 12


In some embodiments, Form A of compound 12 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 34 below.









TABLE 34





XRPD Peak Positions for Form A of Compound 12


Position (°2 θ)















5.4


5.8


6.8


9.7


10.3


12.3


13.4


14.4


16.4


17.4


17.7


20.3


22.0


23.4


23.7


24.9


25.2


26.9


30.2


35.4






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form A of compound 12 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 5.4, 5.8, and 22.0. In some embodiments, Form A of compound 12 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 5.4, 5.8, and 22.0. In some embodiments, Form A of compound 12 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 5.4, 5.8, and 22.0.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 94.


Methods for preparing Form A of compound 12 are described infra.


Form B of Compound 12


In some embodiments, Form B of compound 12 has at least 1, 2, 3, 4 or 5 spectral peak(s) selected from the peaks listed in Table 35 below.









TABLE 35





XRPD Peak Positions for Form B of Compound 12


Position (°2 θ)















5.0


5.8


8.3


9.1


9.9


12.5


13.4


14.8


15.2


16.7


17.4


20.3


21.7


24.8


25.3


26.3


26.8


28.1


29.2


30.6






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form B of compound 12 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 5.0, 9.9, and 26.3. In some embodiments, Form B of compound 12 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 5.0, 9.9, and 26.3. In some embodiments, Form B of compound 12 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 5.0, 9.9, and 26.3.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 96.


Methods for preparing Form B of compound 12 are described infra.


Form C of Compound 12


In some embodiments, Form C of compound 12 has at least 1, 2, 3, 4 or 5 spectral peak(s) is or are selected from the peaks listed in Table 36 below.









TABLE 36





XRPD Peak Positions for Form C of Compound 12


Position (°2 θ)















5.6


5.8


8.4


9.3


10.1


12.4


13.4


14.9


16.2


16.6


17.5


18.5


21.8


22.2


23.4


25.2


25.9


26.3


26.9


36.5






1 In this and all subsequent tables, the position 2 θ is within ±0.2.







In some embodiments, Form C of compound 12 is characterized in that it has one or more peaks in its X-ray powder diffraction pattern selected from those at about 5.6, 5.8, and 8.4. In some embodiments, Form C of compound 12 is characterized in that it has two or more peaks in its X-ray powder diffraction pattern selected from those at about 5.6, 5.8, and 8.4. In some embodiments, Form C of compound 12 is characterized in that it has all three peaks in its X-ray powder diffraction pattern selected from those at about 5.6, 5.8, and 8.4.


In certain embodiments, the X-ray powder diffraction pattern is substantially similar to the XRPD provided in FIG. 98.


Methods for preparing Form C of compound 12 are described infra.


In some embodiments, the present invention provides compound 12:




embedded image


In some embodiments, the present invention provides compound 12, wherein said compound is crystalline.


In some embodiments, the present invention provides compound 12, wherein said compound is a crystalline solid substantially free of amorphous compound 12.


In some embodiments, the present invention provides compound 12, wherein said compound is substantially free of impurities.


In some embodiments, the present invention provides compound 12, wherein said compound has one or more peaks in its XRPD selected from those at about 5.4, about 5.8, and about 22.0 degrees 2-theta. In some such embodiments, the present invention provides compound 12, wherein said compound has at least two peaks in its XRPD selected from those at about 5.4, about 5.8, and about 22.0 degrees 2-theta. In some such embodiments, the present invention provides compound 12, wherein said compound is of Form A.


In some embodiments, the present invention provides compound 12, wherein said compound has an XRPD substantially similar to that depicted in FIG. 94.


In some embodiments, the present invention provides compound 12, wherein said compound has one or more peaks in its XRPD selected from those at about 5.0, about 9.9, and about 26.3 degrees 2-theta. In some such embodiments, the present invention provides compound 12, wherein said compound has at least two peaks in its XRPD selected from those at about 5.0, about 9.9, and about 26.3 degrees 2-theta. In some such embodiments, the present invention provides compound 12, wherein said compound is of Form B.


In some embodiments, the present invention provides compound 12, wherein said compound has a XRPD substantially similar to that depicted in FIG. 96.


In some embodiments, the present invention provides compound 12, wherein said compound has one or more peaks in its XRPD selected from those at about 5.6, about 5.8, and about 8.4 degrees 2-theta. In some such embodiments, the present invention provides compound 12, wherein said compound has at least two peaks in its XRPD selected from those at about 5.6, about 5.8, and about 8.4 degrees 2-theta. In some such embodiments, the present invention provides compound 12, wherein said compound is of Form C.


In some embodiments, the present invention provides compound 12, wherein said compound has an XRPD substantially similar to that depicted in FIG. 98.


In some embodiments, the present invention provides a composition comprising compound 12 and a pharmaceutically acceptable carrier or excipient.


In some embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient compound 12 or composition thereof.


In some embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient compound 12 or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


In some embodiments, the present invention provides a compound selected from: compound 1, Form A, compound 1, Form B, compound 1, Form C, compound 1, Form D, compound 2, Form A, compound 2, Form B, compound 2, Form C, compound 2, Form D, compound 3, Form A, compound 4, Form A, compound 4, Form B, compound 4, Form C, compound 4, Form D, compound 4, Form E, compound 4, Form F, compound 4, Form G, compound 4, Form H, compound 4, Form I, compound 5, Form A, compound 5, Form B, compound 5, Form C, compound 5, Form D, compound 5, Form E, compound 6, Form A, compound 6, Form B, compound 6, Form C, compound 7, Form A, compound 8, Form A, compound 8, Form B, compound 8, Form C, compound 8, Form D, compound 9, Form A, compound 9, Form B, compound 10, Form A, compound 10, Form B, compound 11, Form A, compound 12, Form A, compound 12, Form B, and compound 12, Form C. In some such embodiments, the present invention provides a composition comprising one of the above compound forms and a pharmaceutically acceptable carrier or excipient. In some such embodiments, the present invention provides a method of inhibiting one or both of ERK1 and ERK2 in a patient comprising administering to said patient one of the above compound forms or composition thereof. In some such embodiments, the present invention provides a method of treating an ERK1- or ERK2-mediated disorder in a patient, comprising administering to said patient one of the above compound forms or composition thereof. In some such embodiments, the ERK1- or ERK2-mediated disorder is selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases.


General Methods of Providing a Salt Compound


Compound 1 is prepared according to the methods described in detail in the '230 publication, the entirety of which is hereby incorporated herein by reference. Salt compounds of general formula A, which formula encompasses, inter alia, salt compounds 2 through 12, and/or particular forms thereof, are prepared from compound 1, according to the general Scheme below.




embedded image


For instance, each of compounds 2 through 12, and forms thereof, are prepared from compound 1 by combining compound 1 with an appropriate acid to form a salt of that acid. Thus, another aspect of the present invention provides a method for preparing compounds 2 through 12, and forms thereof.


As described generally above, in some embodiments, the present invention provides a method for preparing a salt compound of the general formula A:




embedded image



comprising steps of:


combining compound 1:




embedded image


with a suitable acid and optionally a suitable solvent under conditions suitable for forming a salt compound of general formula A.


In some embodiments, a suitable acid is phosphoric acid. In some embodiments, the present invention provides a method of making a phosphate salt of compound 1. In some embodiments, the phosphate salt of compound 1 is a monophosphate salt. In certain embodiments, the phosphate salt of compound 1 is compound 2. In certain embodiments, the phosphate salt of compound 1 is Form A of compound 2. In certain embodiments, the phosphate salt of compound 1 is Form B of compound 2. In certain embodiments, the phosphate salt of compound 1 is Form C of compound 2. In certain embodiments, the phosphate salt of compound 1 is Form D of compound 2. In some embodiments, the phosphate salt of compound 1 is a bisphosphate complex In certain embodiments, the phosphate complex of compound 1 is compound 3. In certain embodiments, the phosphate complex of compound 1 is Form A of compound 3.


In some embodiments, a suitable acid is hydrochloric acid. In some embodiments, the present invention provides a method of making a hydrochloride salt of compound 1. In certain embodiments, the hydrochloride salt of compound 1 is compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form A of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form B of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form C of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form D of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form E of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form F of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form G of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form H of compound 4. In certain embodiments, the hydrochloride salt of compound 1 is Form I of compound 4.


In some embodiments, a suitable acid is hydrobromic acid. In some embodiments, the present invention provides a method of making a hydrobromide salt of compound 1. In certain embodiments, the hydrobromide salt of compound 1 is compound 5. In certain embodiments, the hydrobromide salt of compound 1 is Form A of compound 5. In certain embodiments, the hydrobromide salt of compound 1 is Form B of compound 5. In certain embodiments, the hydrobromide salt of compound 1 is Form C of compound 5. In certain embodiments, the hydrobromide salt of compound 1 is Form D of compound 5. In certain embodiments, the hydrobromide salt of compound 1 is Form E of compound 5.


In some embodiments, a suitable acid is sulfuric acid. In some embodiments, the present invention provides a method of making a sulfate salt of compound 1. In some embodiments, the sulfate salt of compound 1 is a monosulfate salt. In certain embodiments, the sulfate salt of compound 1 is compound 6. In certain embodiments, the sulfate salt of compound 1 is Form A of compound 6. In certain embodiments, the sulfate salt of compound 1 is Form B of compound 6. In certain embodiments, the sulfate salt of compound 1 is Form C of compound 6. In some embodiments, the sulfate salt of compound 1 is a bis-sulfate salt. In certain embodiments, the sulfate salt of compound 1 is compound 7. In certain embodiments, the phosphate salt of compound 1 is Form A of compound 7.


In some embodiments, a suitable acid is p-toluenesulfonic acid. In some embodiments, the present invention provides a method of making a tosylate salt of compound 1. In certain embodiments, the tosylate salt of compound 1 is compound 8. In certain embodiments, the tosylate salt of compound 1 is Form A of compound 8. In certain embodiments, the tosylate salt of compound 1 is Form B of compound 8. In certain embodiments, the tosylate salt of compound 1 is Form C of compound 8. In certain embodiments, the tosylate salt of compound 1 is Form D of compound 8.


In some embodiments, a suitable acid is benzenesulfonic acid. In some embodiments, the present invention provides a method of making a besylate salt of compound 1. In certain embodiments, the besylate salt of compound 1 is compound 9. In certain embodiments, the besylate salt of compound 1 is Form A of compound 9. In certain embodiments, the besylate salt of compound 1 is Form B of compound 9.


In some embodiments, a suitable acid is methanesulfonic acid. In some embodiments, the present invention provides a method of making a mesylate salt of compound 1. In certain embodiments, the mesylate salt of compound 1 is compound 10. In certain embodiments, the mesylate salt of compound 1 is Form A of compound 10. In certain embodiments, the mesylate salt of compound 1 is Form B of compound 10.


In some embodiments, a suitable acid is maleic acid. In some embodiments, the present invention provides a method of making a maleate salt of compound 1. In certain embodiments, the maleate salt of compound 1 is compound 11. In certain embodiments, the maleate salt of compound 1 is Form A of compound 11.


In some embodiments, a suitable acid is oxalic acid. In some embodiments, the present invention provides a method of making an oxalate salt of compound 1. In certain embodiments, the oxalate salt of compound 1 is compound 12. In certain embodiments, the oxalate salt of compound 1 is Form A of compound 12. In certain embodiments, the oxalate salt of compound 1 is Form B of compound 12. In certain embodiments, the oxalate salt of compound 1 is Form C of compound 12.


A suitable solvent may be any solvent system (e.g., one solvent or a mixture of solvents) in which compound 1 and/or an acid are soluble, or are at least partially soluble.


Examples of suitable solvents useful in the present invention include, but are not limited to protic solvents, aprotic solvents, polar aprotic solvent, or mixtures thereof. In certain embodiments, suitable solvents include an ether, an ester, an alcohol, a ketone, or a mixture thereof. In some embodiments, the solvent is one or more organic alcohols. In some embodiments, the solvent is chlorinated. In some embodiments, the solvent is an aromatic solvent.


In certain embodiments, a suitable solvent is methanol, ethanol, isopropanol, or acetone wherein said solvent is anhydrous or in combination with water or heptane. In some embodiments, suitable solvents include tetrahydrofuran, dimethylformamide, dimethylsulfoxide, glyme, diglyme, methyl t-butyl ether, t-butanol, n-butanol, and acetonitrile. In some embodiments, a suitable solvent is ethanol. In some embodiments, a suitable solvent is anhydrous ethanol. In some embodiments, the suitable solvent is MTBE.


In some embodiments, a suitable solvent is ethyl acetate. In some embodiments, a suitable solvent is a mixture of methanol and methylene chloride. In some embodiments, a suitable solvent is a mixture of acetonitrile and water. In certain embodiments, a suitable solvent is methyl acetate, isopropyl acetate, acetone, or tetrahydrofuran. In certain embodiments, a suitable solvent is diethylether. In certain embodiments, a suitable solvent is water. In certain embodiments, a suitable solvent is methyl ethyl ketone. In certain embodiments, a suitable solvent is toluene.


In some embodiments, the present invention provides a method for preparing a salt compound of the general formula A, comprising one or more steps of removing a solvent and adding a solvent. In some embodiments, an added solvent is the same as the solvent removed. In some embodiments, an added solvent is different from the solvent removed. Means of solvent removal are known in the synthetic and chemical arts and include, but are not limited to, any of those described herein and in the Exemplification.


In some embodiments, a method for preparing a salt compound of the general formula A comprises one or more steps of heating or cooling a preparation.


In some embodiments, a method for preparing a salt compound of the general formula A comprises one or more steps of agitating or stirring a preparation.


In some embodiments, a method for preparing a salt compound of the general formula A comprises a step of adding a suitable acid to a solution or slurry of compound 1.


In some embodiments, a method for preparing a salt compound of the general formula A comprises a step of heating.


In certain embodiments, a salt compound of formula A precipitates from the mixture. In another embodiment, a salt compound of formula A crystallizes from the mixture. In other embodiments, a salt compound of formula A crystallizes from solution following seeding of the solution (i.e., adding crystals of a salt compound of formula A to the solution).


A salt compound of formula A can precipitate out of the reaction mixture, or be generated by removal of part or all of the solvent through methods such as evaporation, distillation, filtration (ex. nanofiltration, ultrafiltration), reverse osmosis, absorption and reaction, by adding an anti-solvent such as heptane, by cooling or by different combinations of these methods.


As described generally above, a salt compound of formula A is optionally isolated. It will be appreciated that a salt compound of formula A may be isolated by any suitable physical means known to one of ordinary skill in the art. In certain embodiments, precipitated solid salt compound of formula A is separated from the supernatant by filtration. In other embodiments, precipitated solid salt compound of formula A is separated from the supernatant by decanting the supernatant.


In certain embodiments, a salt compound of formula A is separated from the supernatant by filtration.


In certain embodiments, an isolated salt compound of formula A is dried in air. In other embodiments isolated Compound 2 is dried under reduced pressure, optionally at elevated temperature.


Uses of Compounds and Pharmaceutically Acceptable Compositions


As described generally above, compound 1, and pharmaceutically acceptable salts thereof described herein, is an inhibitor of one or both of ERK1 and ERK2. One of ordinary skill in the art will recognize that ERK is one of the key components in the RAS-RAF-MEK-ERK MAPK pathway and that ERK1 and ERK2 are downstream nodes within the MAPK pathway. Without wishing to be bound by theory, because of the downstream location of ERK1 and ERK1 in the MAPK pathway, an ERK inhibitor can treat disease or disorders in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role, including one or both of ERK1 and ERK2 as well as other nodes in the MAPK pathway upstream from ERK (such as Ras, Raf and Mek). Furthermore, because ERK is a downstream target, ERK inhibitors are believed to be able to overcome, in some instances, drug resistance induced by inhibitors of targets upstream of ERK within the MAPK pathway. For example, small molecule inhibitors of RAF or MEK utilized in the treatment of K-RAS and B-RAF mutant tumors have resulted in such drug resistance. Similarly, drug resistance has been associated with other tumors driven by hyperactivation of the MAPK pathway (such as NF 1 mutant tumors). Kinase selectivity was achieved through silencing the selective Cys in a combination of the interactions between the covalent inhibitors of the invention and unique amino acids in the ATP binding pocket. Targeting the selective Cys provides for prolonged pharmacodynamics in silencing ERK activity, as well as potential lower doses in cancer treatment, compared to reversible inhibitors.


The activity of compound 1, and pharmaceutically acceptable salts thereof, as an inhibitor of one or both of an ERK1 and ERK2 kinase, or a mutant thereof, may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of downstream phosphorylation, changes in gene expression, subsequent functional markers and consequences, and/or kinase activity of one or both of activated ERK1 and ERK2 kinase, or a mutant thereof. Alternate in vitro assays quantitate the ability of the test compound to bind to one or both of ERK1 and ERK2. Test compound binding may be measured by radiolabeling the test compound prior to binding, isolating one or both of the compound/ERK1 complex and the compound/ERK2 complex, and determining the amount of radiolabel bound. Alternatively, test compound binding may be determined by running a competition experiment where test compounds are incubated with one or both of ERK1 and ERK2 kinase bound to known radioligands. Test compound binding may be determined by competition with an ERK covalent probe that is amenable to further functionalization with a detection probe, such as, for example, a fluorophore, biotin conjugate, radiolabel, or any other probe that facilitates its quantification. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of one or both of ERK1 and ERK2, or a mutant thereof, are also set forth below and/or in the Examples of the '230 publication.


The term “measurably inhibit”, as used herein means a measurable change in one or both of ERK1 and ERK2 protein kinase activity between a sample comprising a provided composition, and one or both of an ERK1 and ERK2 protein kinase and an equivalent sample comprising one or both of ERK1 and ERK2 protein kinase in the absence of a provided composition. Such measurements of protein kinase activity are known to one of ordinary skill in the art and include those methods set forth herein below and/or in the Examples of the '230 publication.


As described above, in some embodiments, compound 1, and pharmaceutically acceptable salts thereof, is an inhibitor of one or both of ERK1 and ERK2 protein kinases, and ERK1 and ERK2 are downstream targets within the MAPK pathway. Without wishing to be bound by any particular theory, such compounds and compositions are particularly useful for treating or lessening the severity of a disease, condition, or disorder in which activation of the MAPK pathway at any level (Ras-Raf-Mek-ERK) is known or suspected to play a role. Such disease, condition, or disorder may be referred to herein as associated with the MAPK pathway or alternatively as associated with one or both of ERK1 and ERK2. Such diseases, conditions, or disorders may also be referred to herein as an “ERK1- or ERK2-mediated disease, condition, or disorder.”


In some embodiments, the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where activation of the MAPK pathway (at any level in Ras-Raf-Mek-ERK), including one or both of ERK1 and ERK2 protein kinases, is implicated in said disease, condition, or disorder, wherein said method comprises administering to a patient in need thereof a compound of the present invention.


In some embodiments, the present invention relates to a method of inhibiting one or both of ERK1 and ERK2 protein kinase activity in a patient comprising the step of administering to said patient a composition comprising a compound of the present invention.


In other embodiments, the present invention provides a method for treating a disease, condition, or disorder mediated by one or both of ERK1 and ERK2 kinase, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound of the present invention.


In certain embodiments, the present invention provides a method for overcoming drug resistance to Raf or MEK inhibitors, comprising the step of administering to a patient an inhibitor compound of one or both of ERK1 and ERK2, such as a compound of the present invention. In certain embodiments, the mechanism of drug resistance is through mutation of a target protein or reactivation of the MAPK pathway.


As used herein, the term “resistance” may refer to changes in a wild-type nucleic acid sequence coding a target protein, and/or to the amino acid sequence of the target protein and/or to the amino acid sequence of another protein, which changes, decreases or abolishes the inhibitory effect of the inhibitor on the target protein. The term “resistance” may also refer to overexpression or silencing of a protein differing from a target protein that can reactivate the MAPK pathway or other survival pathways.


As used herein, the terms “treatment,” “treat,” and “treating” refer to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein. In some embodiments, treatment is administered after one or more symptoms have developed. In other embodiments, treatment is administered in the absence of symptoms. For example, treatment is administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment is also continued after symptoms have resolved, for example to prevent, delay or lessen the severity of their recurrence.


In some embodiments, the present invention provides a method of treating, stabilizing or lessening the severity or progression of one or more diseases or disorders associated with one or both of ERK1 and ERK2 comprising administering to a patient in need thereof a composition comprising a compound of the present invention.


General diseases, conditions, or disorders treated by a compound of the present invention include cancer, an autoimmune disorder, a neurodegenerative or neurological disorder, liver disease, a cardiac disorder, schizophrenia, or a bone-related disorder.


In some embodiments, the present invention relates to a method of treating or lessening the severity of a disease, condition, or disorder selected from cancer, stroke, diabetes, hepatomegaly, cardiovascular disease including cardiomegaly, Alzheimer's disease, cystic fibrosis, viral disease, autoimmune diseases, atherosclerosis, restenosis, psoriasis, allergic disorders including asthma, inflammation, neurological disorders and hormone-related diseases, wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention. In some embodiments, the cancer is recurring. In certain embodiments, the cancer is refractory. In some embodiments, the cancer is metastatic. In some embodiments, the cancer is locally advanced.


In certain embodiments, the cancer is a RAF inhibitor-resistant cancer. In some such embodiments, the RAF inhibitor-resistant cancer is a BRAF inhibitor-resistant cancer.


In certain embodiments, the cancer is a MEK inhibitor-resistant cancer.


In certain embodiments, the cancer is a MAPK pathway-mediated cancer.


In some embodiments, the cancer is a BRAF-mutated cancer. In certain embodiments, the BRAF-mutated cancer is a BRAFV600-mutated cancer, such as BRAFV600E BRAFV600K, BRAFV600R, and BRAFV600D.


In some embodiments, the cancer is a RAS-mutated cancer. In certain embodiments, the RAS-mutated involves codons 12, 13, or 61. In certain embodiments, the RAS-mutated cancer is a KRAS-mutated cancer, including, but not limited to, KRASG12C/D/V, KRASG13C/D, or KRASQ61L/H/R. In certain embodiments, the RAS-mutated cancer is an NRAS-mutated cancer, including, but not limited to, NRASQ61R, NRASQ61K, NRASQ61L, or NRASQ61H. In certain embodiments, the RAS-mutated cancer is an HRAS-mutated cancer, including, but not limited to, HRASG12V, HRASQ61R, and HRASG12S.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is selected from multiple myeloma, breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach (gastric), skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung, bone, colon, thyroid, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma (including uveal melanoma) sarcoma, bladder carcinoma, liver carcinoma (e.g., hepatocellular carcinoma (HCC)) and biliary passage carcinoma), kidney carcinoma, myeloid disorders, lymphoid disorders, Hodgkin's, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colorectal carcinoma, large intestine, rectum, brain and central nervous system, endometrial, multiple myeloma (MM), prostate, AML, and leukemia. In some such embodiments, the cancer is relapsed. In some embodiments, the cancer is refractory. In some embodiments, the cancer is locally advanced. In some embodiments, the cancer is metastatic.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is selected from carcinoma, lymphoma, blastoma, sarcoma, and leukemia. In some embodiments, a sarcoma is a soft tissue sarcoma. In some embodiments, a lymphoma is non-hodgkins lymphoma. In some embodiments, a lymphoma is large cell immunoblastic lymphoma. In some embodiments, the cancer is selected from adenocarcinoma; adenoma; adrenocortical cancer; bladder cancer; bone cancer; brain cancer; breast cancer; cancer of the buccal cavity; cervical cancer; colon cancer; colorectal cancer; endometrial or uterine carcinoma; epidermoid carcinoma; esophogeal cancer; eye cancer; follicular carcinoma; gallbladder cancer; prostate, AML, multiple myeloma (MM), gastrointestinal cancer, such as, for example, gastrointestinal stromal tumor; cancer of the genitourinary tract; glioblastoma; hairy cell carcinoma; various types of head and neck cancer; hepatic carcinoma; hepatocellular cancer; Hodgkin's disease; keratoacanthoma; kidney cancer; large cell carcinoma; cancer of the large intestine; laryngeal cancer; liver cancer; lung cancer, such as, for example, adenocarcinoma of the lung, anaplastic carcinoma of the lung, papillary lung adenocarcinoma, small-cell lung cancer, squamous carcinoma of the lung, non-small cell lung cancer; melanoma and nonmelanoma skin cancer; lymphoid disorders; myeloproliferative disorders, such as, for example, polycythemia vera, essential thrombocythemia, chronic idiopathic myelofibrosis, myeloid metaplasia with myelofibrosis, chronic myeloid leukemia (CML), chronic myelomonocytic leukemia, chronic eosinophilic leukemia, chronic lymphocytic leukemia (CLL), hypereosinophilic syndrome, systematic mast cell disease, atypical CML, AML, or juvenile myelomonocytic leukemia; plasmacytoma; multiple myeloma; neuroblastoma; ovarian cancer; papillary carcinoma; pancreatic cancer; cancer of the peritoneum; prostate cancer, including benign prostatic hyperplasia; rectal cancer; salivary gland carcinoma; sarcoma; seminoma; squamous cell cancer; small cell carcinoma; cancer of the small intestine; stomach cancer; testicular cancer; thyroid cancer; undifferentiated carcinoma; and vulval cancer. In some such embodiments, the cancer is relapsed. In some embodiments, the cancer is refractory. In some embodiments, the cancer is locally advanced. In some embodiments, the cancer is metastatic.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is selected from melanoma, pancreatic cancer, thyroid cancer, colorectal cancer, lung cancer (e.g., non-small cell lung cancer), breast cancer, endometrial cancer, prostate cancer, ovarian cancer, hepatocellular carcinoma (HCC), multiple myeloma (MM), and leukemia. In some embodiments, a leukemia is an acute leukemia. In certain embodiments, a leukemia is acute myeloid leukemia. In certain embodiments, a leukemia is acute lymphoblastic leukemia.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is selected from melanoma, colorectal cancer, lung cancer, or pancreatic.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is melanoma. In certain embodiments, the melanoma is uveal melanoma. In some embodiments, the melanoma is a melanoma of the skin. In certain embodiments, the melanoma is locally advanced. In some embodiments, the melanoma is metastatic. In some embodiments, the melanoma is recurring. In some embodiments, the melanoma is BRAFv600-mutated melanoma. In certain embodiments, the melanoma is a RAS-mutated melanoma. In some embodiments, the melanoma is NRAS-mutated melanoma. In certain embodiments, the melanoma is wild type for KRAS, NRAS or BRAF. In certain embodiments, the melanoma is a BRAF inhibitor-resistant (e.g., Vemurfenib-resistant, dabrafenib-resistant, etc.) melanoma. In certain embodiments, the cancer is a VemR (i.e., Vemurfenib-resistant) BRAF-mutated melanoma. In some embodiments, the melanoma is relapsed. In some embodiments, the melanoma is refractory.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is colorectal cancer. In certain embodiments, the colorectal cancer is locally advanced. In certain embodiments, the colorectal cancer is metastatic. In certain embodiments, the colorectal cancer is a BRAF-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a BRAFv600-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a RAS-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a KRAS-mutated colorectal cancer. In certain embodiments, the colorectal cancer is a NRAS-mutated colorectal cancer. In some embodiments, the colorectal cancer is relapsed. In some embodiments, the colorectal cancer is refractory.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is pancreatic cancer. In certain embodiments, the pancreatic cancer is locally advanced. In certain embodiments, the pancreatic cancer is metastatic. In certain embodiments, the pancreatic cancer is a pancreatic ductal adenocarcinoma (PDAC). In certain embodiments, the pancreatic cancer is a RAS-mutated pancreatic cancer. In certain embodiments, the pancreatic cancer is a KRAS-mutated pancreatic cancer. In certain embodiments, the pancreatic cancer is KRAS-mutated pancreatic cancer, including, but not limited to, KRASG12C/D/V, KRASG13C/D, or KRASQ61L/H/R. In some embodiments, the pancreatic cancer is relapsed. In some embodiments, the pancreatic cancer is refractory.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is a papillary thyroid cancer. In certain embodiments, the papillary thyroid cancer is locally advanced. In some embodiments, the papillary thyroid cancer is metastatic. In some embodiments, the papillary thyroid cancer is recurring. In some embodiments, the papillary thyroid cancer is BRAF-mutated papillary thyroid cancer. In some embodiments, the papillary thyroid cancer is BRAFv600-mutated papillary thyroid cancer. In some embodiments, the papillary thyroid cancer is relapsed. In some embodiments, the papillary thyroid cancer is refractory. In some embodiments, the papillary thyroid cancer includes undifferentiated or dedifferentiated histology.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is lung cancer. In certain embodiments, the lung cancer is non-small cell lung cancer (NSCLC). In certain embodiments, the lung cancer is locally advanced. In certain embodiments, the lung cancer is metastatic. In certain embodiments, the lung cancer is a RAS-mutated lung cancer. In certain embodiments, the lung cancer is KRAS-mutated lung cancer. In certain embodiments, the lung cancer is a KRAS-mutated lung cancer, including, but not limited to, KRASG12C/D/V, KRASG13C/D, KRASQ61L/H/R. In some embodiments, the lung cancer is relapsed. In some embodiments, the lung cancer is refractory.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is a leukemia. In some embodiments, a leukemia is a chronic leukemia. In certain embodiments, a leukemia is chronic myeloid leukemia. In some embodiments, a leukemia is an acute leukemia. In certain embodiments, a leukemia is acute myeloid leukemia (AML). In certain embodiments, a leukemia is acute monocytic leukemia (AMoL, or AML-M5). In certain embodiments, a leukemia is acute lymphoblastic leukemia (ALL). In certain embodiments, a leukemia is acute T cell leukemia. In certain embodiments, a leukemia is myelomonoblastic leukemia. In certain embodiments, a leukemia is human B cell precursor leukemia. In certain embodiments, a leukemia has a Flt3 mutation or rearrangement. In some embodiments, the leukemia is relapsed. In some embodiments, the leukemia is refractory.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is a CNS cancer, for instance CNS tumors. In certain embodiments, a CNS tumor is a glioblastoma or glioblastoma multiforme (GBM). In some embodiments, the present invention relates to a method of treating stomach (gastric) and esophageal tumors and cancers.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is multiple myeloma (MM). In certain embodiments, the multiple myeloma is locally advanced. In certain embodiments, the multiple myeloma is metastatic. In certain embodiments, the multiple myeloma is a RAS-mutated multiple myeloma. In certain embodiments, the multiple myeloma is KRAS-mutated multiple myeloma. In certain embodiments, the multiple myeloma is a KRAS-mutated multiple myeloma, including, but not limited to, KRASG12C/D/V, KRASG13C/D, or KRASQ61L/H/R. In some embodiments, the multiple myeloma is relapsed. In some embodiments, the multiple myeloma is refractory.


In some embodiments, the present invention relates to a method of treating a cancer, wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is hepatocellular carcinoma (HCC). In certain embodiments, the HCC is locally advanced. In certain embodiments, the HCC is metastatic. In certain embodiments, the HCC is a RAS-mutated HCC. In certain embodiments, the HCC is KRAS-mutated HCC. In certain embodiments, the HCC is a KRAS-mutated HCC, including, but not limited to, KRASG12C/D/V, KRASG13C/D, or KRASQ61L/H/R. In some embodiments, the hepatocellular carcinoma is relapsed. In some embodiments, the hepatocellular carcinoma is refractory.


In some embodiments, the present invention relates to a method of treating a cancer wherein the method comprises administering to a patient in need thereof a composition comprising a compound of the present invention, wherein the cancer is selected from breast, colorectal, endometrial, hematological, leukemia (e.g., AML), liver, lung, melanoma, ovarian, pancreatic, prostate, or thyroid.


Pharmaceutically Acceptable Compositions


The compounds and compositions, according to the method of the present invention, are administered using any amount and any route of administration effective for treating or lessening the severity of a disorder provided above. The exact amount required will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. Compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression “dosage unit form” as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.


Pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention are administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.


Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.


Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.


Injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.


In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.


Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.


Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.


Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.


The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.


Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.


According to one embodiment, the invention relates to a method of inhibiting protein kinase activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.


According to another embodiment, the invention relates to a method of inhibiting one or both of ERK 1 and ERK2 kinase, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound. In certain embodiments, the invention relates to a method of irreversibly inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a biological sample comprising the step of contacting said biological sample with a compound of this invention, or a composition comprising said compound.


The term “biological sample”, as used herein, includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.


Inhibition of one or both of ERK1 and ERK2, or a mutant thereof, activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, biological specimen storage, and biological assays.


Another embodiment of the present invention relates to a method of inhibiting protein kinase activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound.


According to another embodiment, the invention relates to a method of inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. According to certain embodiments, the invention relates to a method of irreversibly inhibiting one or both of ERK1 and ERK2 kinase, or a mutant thereof, activity in a patient comprising the step of administering to said patient a compound of the present invention, or a composition comprising said compound. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 183 of ERK1. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 166 of ERK2. In certain embodiments, the activity is inhibited irreversibly by covalently modifying Cys 183 of ERK1 and Cys 166 of ERK2. In other embodiments, the present invention provides a method for treating a disease, disorder, or condition mediated by one or both of ERK1 and ERK2 kinase, or a mutant thereof, in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof. Such disorders are described in detail herein.


All features of each of the aspects of the invention apply to all other aspects mutatis mutandis. Each of the references referred to herein, including but not limited to patents, patent applications and journal articles, is incorporated by reference herein as though fully set forth in its entirety.


In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting this invention in any manner.


EXEMPLIFICATION

As depicted in the Examples below, in certain exemplary embodiments, compounds are prepared according to the following general procedures. It will be appreciated that, although the general methods depict the synthesis of certain compounds of the present invention, the following general methods, and other methods known to one of ordinary skill in the art, can be applied to all compounds and subclasses and species of each of these compounds, as described herein.


General Procedures


X-ray powder diffraction patterns were collected on one of two instruments, a PANalytical Empyrean X-ray powder diffractometer or a Rigaku XRD, both with CuKα radiation. The PANalytical Empyrean instrument was equipped with a fine focus X-ray tube. The voltage and amperage of the X-ray generator were set at 45 kV and 40 mA, respectively. The divergence slits were set at 1/16° and ⅛°, and the receiving slits were set at 1/16°. Diffracted radiation was measured using a Pixel 2D detector. A theta-two theta continuous scan was set at step size 0.013 or 0.026 from 3° to 40° 2θ with sample spinning rate of 4. A sintered alumina standard was used to check the peak positions. The diffraction patterns measured on the Rigaku system were obtained on a Rigaku XRD; SmartLab with Cu-Kα radiation and D/teX Ultra detector.


The powder samples were deposited on a zero-background polished silicon sample holder and were spun during measurement. Measurements were performed as follows: 40 kV/44 mA tube power, 0.02° 2θ step size, 4 or 5° 2θ/min, and 3-40° 2θ scanning range.


Proton Nuclear Magnetic Resonance (1H NMR) spectra were obtained on a Bruker AVANCE-300 MHz NMR spectrometer. Deuterated DMSO was used as solvent.


DSC data was obtained on a TA Instruments Q1000, Q2000, or Discover Series DSC. Samples were weighed into aluminum pans, crimped with pin hole aluminum lids, and heated at a rate of 10° C./min. Indium was used as the calibration standard.


TGA analyses were performed on a TA instruments Q500 or Discovery Series TGA. About 2 to 10 mg were added to an aluminum sample pan and heated at a rate of 10° C./min.


TGA/DSC data was obtained on a Mettler Toledo TGA/DSC 1 Star System. The samples were loaded on a aluminum sample pan and heated at a rate of 10° C./min.


Hygroscopicity was determined on a Surface Measurement Systems DVS-1 or DVS-Advantage Dynamic Vapor Sorption analyzer. A typical sample size of 5 to 20 mg was loaded into the DVS instrument sample pan and the sample was analyzed on a DVS automated sorption analyzer at room temperature. The relative humidity (RH) was increased from 0 or 5% to 90 or 95% RH at 5 or 10% RH steps. The RH was then decreased in a similar manner to accomplish a full absorption/desorption cycle. In some cases a second absorption/desorption cycle was performed.


Example A
General Preparation of Compound 1



embedded image


The title compound was prepared according to the steps and intermediates described below and in the '230 publication, the entirety of which is incorporated herein by reference.


Step 1: N-(2-(2-Chloro-5-(trifluoromethyl)pyrimidin-4-ylamino)5-methylphenyl)acrylamide (Intermediate 1)



embedded image


To a stirred solution of N-(2-amino-5-methylphenyl)acrylamide (22.2 mmol) in dimethyl acetamide (25 mL) was added potassium carbonate (46.0 mmol) at rt, and the mixture was stirred for 15 minutes. To this reaction mixture, 2,4-dichloro-5-trifluoromethylpyrimidine (22.2 mmol) was added, and the stirring continued at 60° C. for 1 h. Upon completion, the reaction mixture was diluted with water (2×50 mL) and extracted with EtOAc (2×100 mL). The organic layer was dried over sodium sulfate and concentrated to get the crude product. This crude was purified by silica gel column chromatography and subsequently purified by prep-HPLC to get desired intermediate 1.


Step 2: Acid Catalyzed Coupling Method

To a solution of Intermediate 1 (2.923 mmol) in 0.04 M PTSA solution in 1,4-dioxane (20 mL) was added 2-methoxy-5-methylpyridin-4-amine (3.5076 mmol), and the mixture was stirred at 95° C. for 16 h. Upon completion, the reaction mixture was directly absorbed on silica gel and purified by column chromatography. The resulting product was stirred in a mixture of DCM:EtOAc:diethyl ether (10 mL:10 mL:30 mL) for 10 min, then filtered and dried under vacuum to obtain the desired compound.


MS m/z 459.2 (ES+, M+H). 1HNMR (DMSO-d6) δ 2.10 (s, 3H), 2.32 (s, 3H), 3.75 (s, 3H), 5.78 (dd, 1H, J=2.0, 10.0 Hz), 6.28 (dd, 1H, J=2.0, 16.8 Hz), 6.45 (dd, 1H, J=10.6, 16.8 Hz), 7.09 (br t, 3H, J=8.0 Hz), 7.50 (d, 1H, J=8.4 Hz), 7.79 (s, 1H), 8.36 (s, 2H), 8.72 (s, 1H), 10.25 (s, 1H).


Alternative Step 2: Pd-Catalyzed Coupling Method

Alternatively, Step 2 can be carried out by adding Intermediate 1 to a suitable coupling partner in the presence of Na2CO3, a degassed solvent (e.g., tert-amyl alcohol), a suitable palladium catalyst (e.g., tris-dibenzylamino dipalladium) and a suitable phosphine ligand (e.g., Dave Phos) under conditions suitable to effect coupling.


Example 1
Preparation of Free Base Forms A-D of Compound 1



embedded image


Compound 1 is prepared according to the method described in detail in Example 94 of the '230 publication, the entirety of which is hereby incorporated herein by reference.


Form A of Compound 1


Form A of compound 1 was prepared as follows.


Procedure A: Form B of compound 1 was slurried in ethyl acetate at 30° C. After overnight vigorous stirring, the reaction was filtered and dried.


Procedure B: Form B of compound 1 was heated to a temperature of 170° C., then cooled to room temperature. Solids converted to Form A at a temperature of 145° C.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A.


Table 1, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 1.









TABLE 1





XRPD Peak Positions for Form A of Compound 1


Position (°2 θ)















8.6


9.0


11.6


14.1


15.3


15.4


17.2


18.5


20.4


20.6


21.2


21.5


21.9


22.9


23.4


23.9


25.3


25.8


26.1


27.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 1 depicts an XRPD pattern of Form A of compound 1.



FIG. 2 depicts a DSC thermogram and TGA trace of Form A of compound 1.



FIG. 3 depicts a DVS plot of Form A of compound 1.


Form B of Compound 1


Form B of compound 1 was prepared as follows.


Procedure A: compound 1 was slurried in 10 volumes of methanol for 8 hours at 20° C., filtered, and dried.


Procedure B: compound 1 was dissolved in MeOH/CH2Cl2 (11.20 mg/mL), dried under nitrogen purge, then slurried in MeOH overnight, filtered and dried in a vacuum oven at ambient temperature until analysis.


Characterization of the resulting material demonstrated a crystalline Form B.


Table 2, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 1.









TABLE 2





XRPD Peak Positions for Form B of Compound 1


Position (°2 θ)















4.6


7.2


8.3


9.3


11.6


13.6


14.4


16.5


17.7


18.7


21.0


21.3


23.2


23.5


24.4


27.5






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 4 depicts an XRPD pattern of Form B of compound 1.



FIG. 5 depicts a DSC thermogram of Form B of compound 1.



FIG. 6 depicts a TGA trace of Form B of compound 1.


Elemental analysis—Calculated: C, 57.64; H, 4.62; N, 18.33; Found: C, 57.46; H, 4.58; N, 18.36.


Karl Fisher titration: 0%


Form C of Compound 1


Form C of Compound 1 was prepared as follows.


Procedure A: Form A of compound 1 was slurried in methyl acetate, acetone, or tetrahydrofuran for seven days at room temperature.


Procedure B: 6.8 g of free base Form B were charged to 41 mL isopropylacetate and slurried at 20° C. for about 1 hour. The batch was filtered, washed with 7 mL of isopropyl acetate and dried under reduced pressure to yield 6.4 g.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form C.


Table 3, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 1.









TABLE 3





XRPD Peak Positions for Form C of Compound 1


Position (°2 θ)















7.6


8.8


9.6


11.7


12.3


14.5


15.3


15.9


17.5


18.0


20.1


21.0


22.8


23.4


23.8


24.7


25.2


25.5


26.4


27.7






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 7 depicts an XRPD pattern of Form C of compound 1.



FIG. 8 depicts a DSC thermogram of Form C of compound 1.



FIG. 9 depicts a TGA trace of Form C of compound 1.


Form D of Compound 1


Form D of compound 1 was prepared as follows.


Procedure: Form B of Compound 1 (400 mg) was charged to a vial with 1.7 mL tetrahydrofuran and 0.3 mL water and vigorously stirred. After 24 hours, the slurry was filtered and dried in a vacuum oven at 45° C.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form D.


Table 4, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form D of compound 1.









TABLE 4





XRPD Peak Positions for Form D of Compound 1


Position (°2 θ)















8.2


8.9


9.5


9.6


10.6


15.0


17.3


17.7


19.1


20.2


21.1


22.0


22.4


23.7


24.7


25.2


25.9


26.5


28.6


35.1






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 10 depicts an XRPD pattern of Form D of compound 1.



FIG. 11 depicts a DSC thermogram of Form D of compound 1.



FIG. 12 depicts a TGA trace of Form D of compound 1.


Free Base Competitive Slurries:


Three competitive slurry experiments were performed:


1: Form A (20.0 mg)+Form C (20.4 mg) in 0.4 ml ethyl acetate


2: Form B (15.7 mg)+Form C (15.6 mg) in 0.3 ml ethyl acetate


3: Form A (13.3 mg)+Form B (11.5 mg) in 0.3 ml ethyl acetate


The three slurries were stirred vigorously for three days. Solids were then filtered and dried. XRPD and DSC showed conversion to form C for all three experiments.


About 20 mg Form C was stirred in methanol (˜2 mL) at room temperature for 24 hours. Solids were filtered and dried. XRPD and DSC showed solids converted to Form B.


Form B of compound 1 was heated at 5° C./min in an XRD-DSC instrument. Both XRPD and DSC were recorded simultaneously, indicating Form B experienced solid-solid transition between 153° C. and 160° C., resulting in Form A, which melted around 222° C. and recrystallized to become Form C, which melted around 235° C.


Example 2
Preparation of Forms A-D of Compound 2



embedded image



Form A of Compound 2


Form A of compound 2 was prepared as follows.


Procedure A: Compound 1 was dissolved in 15× tetrahydrofuran. One molar equivalent of 2 molar phosphoric acid in acetonitrile was charged. The batch was slurried at 20° C. for 1 to 2 hours. The solvent was removed under reduced pressure. The resulting solids were slurried in acetone for about 16 hours at 20° C., filtered and dried.


Procedure B: Compound 1 was dissolved in THF. Equal molar equivalent of 1.08 M phosphoric acid in acetonitrile was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetone with a stirring bar at ambient temperature overnight, then filtered and dried in vacuum oven at 30° C. overnight.


Procedure C: Compound 1 was dissolved in THF (20× vol) at 20° C. Seeds of compound 2 Form A (5% wt) were charged. A 1 M solution of phosphoric acid (1 mol eq.) in ethanol was charged. The batch was left under vigorous agitation for two hours. Solvent exchange to isopropyl acetate was carried out with a constant volume distillation under reduced pressure, with temperature not exceeding 40° C. The batch was cooled to 20° C. The solvent was removed under nitrogen purge. The batch was filtered, washed two times with isopropyl acetate and dried in a vacuum oven at ˜40° C. overnight, under vacuum with nitrogen bleed.


Procedure D: Compound 1 was dissolved in 9× vol THF/H2O (95:5 vol). A solution of H3PO4 (1.2 mol eq.) in ethanol was charged to a second flask, seeds of compound 2 form A (5%) were charged and vigorous agitation was started. The solution of compound 1 was charged to the H3PO4 solution (reverse addition) over one hour. The slurry was aged for one hour. Solvent exchange to ethanol was started (constant volume vacuum distillation with continuous addition of ethanol, final THF NMT 0.5%). The batch was cooled to 20° C., filtered and dried in a vacuum oven at ˜40° C. overnight, under vacuum with nitrogen bleed.


Procedure E: Compound 1 was dissolved in 10× vol THF/H2O (95:5 vol). Isopropyl alcohol (5× vol) was charged. Constant volume distillation, with continuous addition of isopropyl alcohol was started at atmospheric pressure. Solvent exchange was carried out until THF content was below 5%. Compound 1 recrystallized during the solvent exchange. The batch was cooled to 30° C. A 1 M solution of H3PO4 in IPA was charged over 2 hours. Seeds of compound 2 Form A (1%) were then charged. The batch was stirred vigorously overnight. The batch was filtered and dried in a vacuum oven at ˜40° C. overnight, under vacuum with nitrogen bleed.


Procedure F: Compound 1 was dissolved in 9× vol THF/H2O (95:5 vol). After polish filtration, distillation to reduce volume from 9× to 5× was performed, followed by addition of 8× ethyl acetate to bring the total volume to 13×. Solvent exchange to ethyl acetate, with constant volume distillation was carried out (final THF NMT 2%). The temperature was then reduced to 30° C. Seeds of compound 2 (1% wt) were charged. A solution of H3PO4 (1.2 eq.) in ethanol (5×) was then dosed in over 2 hours. The temperature was reduced to 20° C., the batch was aged for 12 hours under vigorous stirring, then filtered, washed two times with ethyl acetate and dried in a vacuum oven at ˜40° C. overnight, under vacuum with nitrogen bleed.


Procedure G: Compound 1 was charged to a reactor, then ethanol (4× vol) and ethyl acetate (6×), were charged. The batch was agitated at 30° C. A solution of H3PO4 (1.2 mol eq.) in ethanol (2× vol) was charged over 2 hours. Seeds of compound 2 Form A (1%) were charged. The batch was filtered, washed two times with ethyl acetate, dried overnight at ˜40° C., under vacuum with nitrogen bleed.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 2. Up to 3.8% water uptake was observed for this Form at 95% relative humidity.


Table 5, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 2.









TABLE 5





XRPD Peak Positions for Form A of Compound 2


Position (°2 θ)















5.9


6.3


6.8


9.8


10.1


11.1


13.8


14.4


15.4


16.0


16.6


17.3


17.9


18.9


19.2


19.7


20.3


20.8


21.3


22.2


23.0


23.3


23.6


24.0


24.7


25.5


26.0


26.8


27.4


27.9


28.4


29.2


30.5


31.3


31.8






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 13 depicts an XRPD pattern of Form A of compound 2.



FIG. 14 depicts a DSC thermogram of Form A of compound 2.



FIG. 15 depicts a TGA trace of Form A of compound 2.



FIG. 16 depicts a DVS plot of Form A of compound 2.


Elemental analysis—Calculated: C, 47.49; H, 4.35; N, 15.10; P, 5.57; Found: C, 47.09; H, 4.33; N, 14.90; P, 5.57.


Karl Fisher titration: 0.22%


Form B of Compound 2


Form B of compound 2 was prepared as follows.


Procedure: The phosphate salt of compound 1 was dissolved in tetrahydrofuran or ethanol and filtered using a syringe filter. The solution was evaporated at room temperature under a nitrogen stream.


Characterization of the resulting material demonstrated a crystalline Form B of compound 2 in the form of a hydrate.


Table 6, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 2.









TABLE 6





XRPD Peak Positions for Form B of Compound 2


Position (°2 θ)















3.6


7.3


8.6


9.5


10.7


12.0


13.5


14.6


15.0


15.7


16.6


18.2


19.2


19.9


20.3


21.6


22.0


22.5


22.9


23.4


24.1


24.9


25.3


25.7


26.3


26.9


27.8


28.7


29.5


30.2


31.8


34.2


36.1


37.1


38.8


39.3






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 17 depicts an XRPD pattern of Form B of compound 2.



FIG. 18 depicts a DSC thermogram of Form B of compound 2.



FIG. 19 depicts a TGA trace of Form B of compound 2.



FIG. 20 depicts a DVS plot of Form B of compound 2.


Phosphate content: 17.8 wt %


Karl Fisher titration: 3.1%


Form C of Compound 2


Form C of compound 2 was prepared as follows.


Procedure: The phosphate salt of compound 1 was dissolved in ethanol at room temperature and filtered using a syringe filter. Three to five volumes of acetonitrile was added as anti-solvent. The mixture was cooled in a refrigerator. Precipitated solids were isolated by filtration and air dried.


Characterization of the resulting material demonstrated a crystalline Form C of compound 2.


Table 7, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 2.









TABLE 7





XRPD Peak Positions for Form C of Compound 2


Position (°2 θ)















4.2


6.8


8.4


9.3


11.6


12.5


12.7


13.7


15.3


15.8


16.5


18.7


19.4


20.5


22.0


22.7


24.5


25.2


26.2


32.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 21 depicts an XRPD pattern of Form C of compound 2.



FIG. 22 depicts a DSC thermogram of Form C of compound 2.



FIG. 23 depicts a TGA trace of Form C of compound 2.



FIG. 24 depicts a DVS plot of Form C of compound 2.


Phosphate content: 16.8 wt %


Karl Fisher titration: 0.14%


Form D of Compound 2


Form D of compound 2 was prepared as follows.


Procedure: The phosphate salt of compound 1 (i.e., compound 2, Form C) was heated to 140° C. to obtain Form D of compound 2. Alternatively, compound 1 free base (3 g) was dissolved in about 60 ml of THF. Phosphoric acid (1 molar equiv., 1M in water) was charged. The solution was stirred for one hour followed by removal of the solvent under reduced pressure. Acetone (10 volumes) was charged to the isolated solid and the slurry was agitated overnight after which the solids were collected and dried under reduced pressure at about 40° C.


Characterization of the resulting material demonstrated a crystalline Form D of compound 2.


Table 8, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form D of compound 2.









TABLE 8





XRPD Peak Positions for Form D of Compound 2


Position (°2 θ)















7.1


8.1


9.1


10.4


10.6


11.2


12.9


13.9


15.8


16.4


17.2


17.7


18.7


19.0


20.2


20.7


21.0


22.1


22.7


24.5


25.1


26.4


27.4


27.8


28.7


29.1


31.0


31.5


33.8


36.3


37.0


38.0






1 In this and all subsequent tables, the position 2 θ is within ±0.2.








FIG. 25 depicts an XRPD pattern of Form D of compound 2.



FIG. 26 depicts a DSC thermogram of Form D of compound 2.



FIG. 27 depicts a TGA trace of Form D of compound 2.


Phosphate Interconversions


From polymorph screen:















Starting

Temperature/



Form(s)
Solvent/Condition
Condition
XRPD Result







Form B
slurry in MeOH
RT, 5 days
free base (Form B)


Form B
slurry in EtOH
RT, 5 days
Form A + free base





(Form C)


Form B
slurry in acetone
RT, 5 days
Form A


Form B
slurry in MeCN
RT, 5 days
Form A


Form B
slurry in
RT, 5 days
free base (Form D)



MeCN/water (1:1)









Example 3
Preparation of Form A of Compound 3



embedded image



Form A of Compound 3


Form A of compound 3 was prepared as follows.


Procedure: Compound 1 was dissolved in 20 volumes of isopropanol. Eight molar equivalents of phosphoric acid were charged. The reaction was heated to 60° C. for 24 hours, then cooled to 20° C., filtered, washed with 10 volumes of isopropanol, and dried under reduced pressure at 40° C.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 3.


Table 9, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 3.









TABLE 9





XRPD Peak Positions for Form A of Compound 3


Position (°2θ)















5.7


7.1


8.9


10.3


11.0


11.4


13.2


14.2


14.5


17.2


17.7


18.4


19.4


19.9


20.6


20.8


22.1


22.5


24.3


25.5






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 28 depicts an XRPD pattern of Form A of compound 3.



FIG. 29 depicts a DSC thermogram of Form A of compound 3.



FIG. 30 depicts a TGA trace of Form A of compound 3.



FIG. 31 depicts a DVS plot of Form A of compound 3.


Elemental analysis—Calculated: C, 40.38; H, 4.16; N, 12.84; P, 9.47; Found: C, 40.01; H, 4.15; N, 12.61; P, 9.69.


Example 4
Preparation of Forms A-I of Compound 4



embedded image



Form A of Compound 4


Form A of compound 4 was prepared as follows.


Procedure A: Two grams of the hydrochloride salt of compound 1 were dissolved in 10 mL of methanol and passed through a 0.2 uM filter. The solution was reduced in volume to about 3 mL and stirred for about 16 hours at 23° C. The resulting solids were collected by suction filtration and dried under a nitrogen stream.


Procedure B: 507 mg of compound 1 were mixed with 8 mL of MeOH/CH2Cl2 (1/1 v/v, premixed), followed by addition of 1.1 mL 1N HCl (in diethylether). The solution was stirred for 1 hour to become a clear solution. The solution was dried in a fume hood under nitrogen purge. Next, 1 mL of methanol was added to the vial, which was vortex agitated until precipitation was observed. The vial was capped and stirred at room temperature overnight. The solids were recovered through filtration and dried in a vacuum oven at room temperature.


Characterization of the resulting material demonstrated a crystalline Form A of compound 4 in the form of a hydrate.


Table 10, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 4.









TABLE 10





XRPD Peak Positions for Form A of Compound 4


Position (°2θ)















7.5


9.3


11.2


11.9


14.2


15.0


15.3


15.7


21.4


21.9


22.6


22.8


23.4


24.6


24.8


25.2


26.4


26.8


30.5


34.1






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 32 depicts an XRPD pattern of Form A of compound 4.



FIG. 33 depicts a DSC thermogram of Form A of compound 4.



FIG. 34 depicts a TGA trace of Form A of compound 4.



FIG. 35 depicts a DVS plot of Form A of compound 4.


Elemental analysis—Calculated: C, 51.52; H, 4.72; Cl, 6.91; N, 16.38; Found: C, 51.89; H, 4.51; Cl, 7.11; N, 16.52.


Karl Fischer titration: 2.56%


Form B of Compound 4


Form B of compound 4 was prepared as follows.


Procedure: Compound 1 was dissolved in tetrahydrofuran. An equal molar equivalent of 1M HCl in diethyl ether was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetonitrile with a stirring bar at ambient temperature overnight, then filtered and dried in a vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline Form B of compound 4.


Table 11, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 4.









TABLE 11





XRPD Peak Positions for Form B of Compound 4


Position (°2θ)















8.0


8.4


9.1


11.0


11.8


12.7


16.2


17.2


17.8


18.9


20.4


20.9


23.7


23.9


24.7


25.4


25.6


25.9


29.2


30.7






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 36 depicts an XRPD pattern of Form B of compound 4.



FIG. 37 depicts a DSC thermogram and TGA trace of Form B of compound 4.


Form C of Compound 4


Form C of compound 4 was prepared as follows.


Procedure: Form B of compound 4 was slurried in acetone for 5 days at room temperature.


Characterization of the resulting material demonstrated a crystalline Form C of compound 4.


Table 12, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 4.









TABLE 12





XRPD Peak Positions for Form C of Compound 4


Position (°2θ)















7.7


8.2


9.0


9.5


10.2


12.1


12.4


12.6


13.5


15.3


16.4


18.4


21.0


21.3


23.1


23.4


23.7


24.2


25.5


25.7






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 38 depicts an XRPD pattern of Form C of compound 4.



FIG. 39 depicts a DSC thermogram of Form C of compound 4.


Form D of Compound 4


Form D of compound 4 was prepared as follows.


Procedure: Compound 1 (3 g, 6.5 mmol) was charged to a 150 mL round bottom flask. Methanol (15×) was charged. A solution of HCl (3.27 mL, 6.54 mmol; 2M in diethyl ether) was charged to the flask. The solids completely dissolved. The solution was stirred overnight, then transferred to a crystallizing dish for slow evaporation of the solvent. Solids were then placed to dry in a vacuum oven with a nitrogen bleed at 40° C.


Characterization of the resulting material demonstrated a crystalline Form D of compound 4.


Table 13, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form D of compound 4.









TABLE 13





XRPD Peak Positions for Form D of Compound 4


Position (°2θ)















7.1


9.1


11.2


12.7


14.0


14.4


14.6


17.0


17.2


21.7


22.0


22.4


22.8


23.7


24.8


25.3


25.5


28.0


34.2


35.7






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 40 depicts an XRPD pattern of Form D of compound 4.



FIG. 41 depicts a DSC thermogram of Form D of compound 4.



FIG. 42 depicts a DSC thermogram and TGA trace of Form D of compound 4.



FIG. 43 depicts a DVS plot of Form D of compound 4.


Karl Fischer titration: 1.21%


Form E of Compound 4


Form E of compound 4 was prepared as follows.


Procedure: Form B of compound 4 was slurried in acetonitrile/water (1:1) for five days at room temperature.


Characterization of the resulting material demonstrated a crystalline Form E of compound 4.


Table 14, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form E of compound 4.









TABLE 14





XRPD Peak Positions for Form E of Compound 4


Position (°2θ)















7.7


9.0


10.7


14.4


15.5


17.1


18.2


19.8


20.4


21.5


23.1


24.4


24.6


25.0


27.0


27.3


28.1


30.9


31.2


37.6






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 44 depicts an XRPD pattern of Form E of compound 4.



FIG. 45 depicts a DSC thermogram of Form E of compound 4.


Form F of Compound 4


Form F of compound 4 was prepared as follows.


Procedure: Form B of compound 4 was slurried in isopropanol for five days at room temperature.


Characterization of the resulting material demonstrated a crystalline Form F of compound 4.


Table 15, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form F of compound 4.









TABLE 15





XRPD Peak Positions for Form F of Compound 4


Position (°2θ)















6.1


7.7


11.3


12.4


13.8


15.4


16.9


17.7


18.6


19.5


22.6


22.9


23.1


23.6


24.9


25.7


26.1


30.2


34.2


35.0






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 46 depicts an XRPD pattern of Form F of compound 4.



FIG. 47 depicts a DSC thermogram of Form F of compound 4.


Form G of Compound 4


Form G of compound 4 was prepared as follows.


Procedure: Form B of compound 4 was slurried in acetonitrile for five days at room temperature.


Characterization of the resulting material demonstrated a crystalline Form G of compound 4.


Table 16, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form G of compound 4.









TABLE 16





XRPD Peak Positions for Form G of Compound 4


Position (°2θ)















5.2


8.5


8.7


10.5


14.8


15.6


19.0


19.5


20.7


21.0


21.9


22.2


23.3


25.7


27.0


27.4


29.6


30.7


31.3






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 48 depicts an XRPD pattern of Form G of compound 4.



FIG. 49 depicts a DSC thermogram of Form G of compound 4.


Form H of Compound 4


Form H of compound 4 was prepared as follows.


Procedure: 1 g of the HCl salt of compound 1 was dissolved in methanol (4 mL) and filtered through a syringe filter. The solvent was evaporated slowly under a stream of nitrogen until the reaction mixture was slightly cloudy. The salt solution was stirred at room temperature overnight, filtered, and dried at room temperature under a stream of nitrogen for 15 minutes.


Characterization of the resulting material demonstrated a crystalline Form H of compound 4.


Table 17, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form H of compound 4.









TABLE 17





XRPD Peak Positions for Form H of Compound 4


Position (°2θ)















7.2


8.5


9.1


10.2


13.1


14.5


15.8


18.1


18.8


20.5


21.0


21.7


22.5


23.3


24.2


24.6


25.3


25.8


28.8


29.9






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 50 depicts an XRPD pattern of Form H of compound 4.



FIG. 51 depicts a DSC thermogram of Form H of compound 4.


Form I of Compound 4


Form I of Compound 4 was prepared as follows.


Procedure: Form H of compound 4 was slurried in water for two hours at room temperature.


Characterization of the resulting material demonstrated a crystalline Form I of compound 4.


Table 18, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form I of compound 4.









TABLE 18





XRPD Peak Positions for Form I of Compound 4


Position (°2θ)















4.7


7.1


9.3


10.8


11.9


14.1


15.0


16.7


18.8


20.3


21.1


22.2


23.1


23.5


24.2


25.4


26.0


29.1






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 52 depicts an XRPD pattern of Form I of compound 4.


Example 5
Preparation of Forms A-E of Compound 5



embedded image



Form A of Compound 5


Form A of compound 5 was prepared as follows.


Procedure A: 4.1 g of compound 1 was dissolved in tetrahydrofuran (15×). Hydrobromic acid (1 molar equivalent of 48% HBr diluted to 2M in acetonitrile) was charged and agitated for 2 hours at 20° C. The solvent was removed under reduced pressure and the resulting solids were slurried in acetone for 16 hours at 20° C., filtered and dried to yield 4.89 g product.


Procedure B: Compound 1 was dissolved in tetrahydrofuran. Equal molar equivalent of 8.84 M HBr in water was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetonitrile (or acetone) with a stirring bar at ambient temperature overnight, then filtered and dried in vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 5.


Table 19, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 5.









TABLE 19





XRPD Peak Positions for Form A of Compound 5


Position (°2θ)















7.5


9.5


11.3


13.0


14.3


14.7


15.0


15.7


17.3


20.3


20.9


21.6


22.6


23.2


23.7


24.8


26.0


28.8


30.5


33.8






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 53 depicts an XRPD pattern of Form A of compound 5.



FIG. 54 depicts a DSC thermogram of Form A of compound 5.



FIG. 55 depicts a TGA trace of Form A of compound 5.



FIG. 56 depicts a DVS plot of Form A of compound 5.


Elemental analysis—Calculated: C, 48.99; H, 4.11; Br, 14.81; N, 15.58; Found: C, 48.37; H, 4.19; Br, 15.34; N, 15.28.


Karl Fischer titration: 0.35%


Form B of Compound 5


Form B of compound 5 was prepared as follows.


Procedure: Compound 1 was dissolved in tetrahydrofuran. Equal molar equivalent of 8.84 M HBr in water was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methyl ethyl ketone or toluene with magnetic stirring at ambient temperature overnight, then filtered and dried in a vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline Form B of compound 5.


Table 20, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 5.









TABLE 20





XRPD Peak Positions for Form B of Compound 5


Position (°2θ)















8.3


11.8


12.3


14.2


16.2


17.9


18.7


18.9


20.3


20.5


20.8


21.1


23.8


24.5


25.5


25.7


26.3


29.2


30.7


31.9






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 57 depicts an XRPD pattern of Form B of compound 5.



FIG. 58 depicts a DSC thermogram and TGA trace of Form B of compound 5.


Form C of Compound 5


Form C of compound 5 was prepared as follows.


Procedure: Compound 1 (0.5 g, 1.091 mmol) was dissolved in THF (5 ml, 61.0 mmol) at room temperature. HBr (0.148 ml, 1.309 mmol) was added to solution to form a slightly hazy solution, which was stirred at room temperature for 15 min. Next was added acetonitrile (5 mL). Nitrogen was streamed over the hazy solution until solvent was fully evaporated. Pale yellow solid formed. The product was charged with acetonitrile (5 mL), slurried overnight, filtered, and dried.


Characterization of the resulting material demonstrated a crystalline Form C of compound 5.


Table 21, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 5.









TABLE 21





XRPD Peak Positions for Form C of Compound 5


Position (°2θ)















7.46x


8.4


10.5


15.4


15.8


17.4


20.1


20.8


22.1


22.3


22.6


23.2


24.2


24.5


25.9


26.6


29.0


31.8


33.8


37.7






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 59 depicts an XRPD pattern of Form C of compound 5.



FIG. 60 depicts a DSC thermogram and TGA trace of Form C of compound 5.


Form D of Compound 5


Form D of compound 5 was prepared as follows.


Procedure: Compound 1 (3.0 g, 6.54 mmol) was dissolved in 60 mL tetrahydrofuran. HBr (48% diluted to 1M with water) was charged in a 1:1 ratio. The solution was stirred for one hour after which the solvent was evaporated under reduced pressure. Acetonitrile (10×) was charged and the slurry was vigorously stirred overnight. Solids were filtered and dried in a vacuum oven at 40° C. with a nitrogen bleed.


Characterization of the resulting material demonstrated a crystalline Form D of compound 5.


Table 22, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form D of compound 5.









TABLE 22





XRPD Peak Positions for Form D of Compound 5


Position (°2θ)















7.2


9.4


10.8


11.8


15.1


16.9


18.9


20.4


21.6


22.3


23.7


24.3


24.7


25.5


26.1


29.0


30.3


30.6


32.1


35.2






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 61 depicts an XRPD pattern of Form D of compound 5.



FIG. 62 depicts a DSC thermogram and TGA trace of Form D of compound 5.


Form E of Compound 5


Form E of compound 5 was prepared as follows.


Procedure: Compound 1 (3 g, 6.54 mmol) was dissolved in ˜60 ml tetrahydrofuran. HBr (48% diluted to ˜1M with water) was charged in a 1:1 ratio. The solution was stirred for one hour, after which the solvent was evaporated under reduced pressure. Acetonitrile (10×) was charged and the slurry was vigorously stirred overnight. The solvent was removed under reduced pressure. Tetrahydrofuran (20×) was charged. The slurry was vigorously stirred, then the tetrahydrofuran was removed under reduced pressure to facilitate the removal of water. Acetonitrile (10×) was charged and the slurry was again vigorously stirred overnight. Solids were filtered and dried in a vacuum oven at 40° C., with nitrogen bleed.


Characterization of the resulting material demonstrated a crystalline Form E of compound 5.


Table 23, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form E of compound 5.









TABLE 23





XRPD Peak Positions for Form E of Compound 5


Position (°2θ)















7.8


8.7


10.0


11.6


15.0


16.4


17.5


18.7


21.5


22.8


23.8


24.3


25.2


25.8


28.8


29.1


30.3


31.7


33.7


36.8






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 63 depicts an XRPD pattern of Form E of compound 5.



FIG. 64 depicts a DSC thermogram of Form E of compound 5.



FIG. 65 depicts a DVS plot of Form E of compound 5.


Example 6
Preparation of Forms A-C of Compound 6



embedded image



Form A of Compound 6


Form A of compound 6 was prepared as follows.


Procedure A: Compound 1 (4.1 g, 8.94 mmol) was dissolved in 15× tetrahydrofuran. One molar equivalent of 2 M sulfuric acid in acetonitrile was charged. Solids precipitated as the acid was charged. The slurry was stirred for 2 hours at 20° C., then the solvent was removed under reduced pressure. The resulting solids were slurried in ethyl acetate at 20° C. for about 16 hours, filtered, and dried.


Procedure B: Compound 1 was dissolved in tetrahydrofuran. Equal molar equivalent of 1.18 M H2SO4 in acetonitrile was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in ethyl acetate with a stirring bar at ambient temperature overnight, then filtered and dried in vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 6.


Table 24, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 6.









TABLE 24





XRPD Peak Positions for Form A of Compound 6


Position (° 2θ)















6.2


7.1


9.9


14.2


14.7


19.4


19.5


20.1


20.2


20.7


21.4


23.4


23.9


24.3


24.8


25.3


26.0


26.9


28.7


29.8






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 66 depicts an XRPD pattern of Form A of compound 6.



FIG. 67 depicts a DSC thermogram of Form A of compound 6.



FIG. 68 depicts a TGA trace of Form A of compound 6.



FIG. 69 depicts a DVS plot of Form A of compound 6.


Elemental analysis—Calculated: C, 47.48; H, 4.17; N, 15.10; S, 5.76; Found: C, 46.89; H, 4.31; N, 14.73; S, 6.05.


Karl Fischer titration: 0.69%


Form B of Compound 6


Form B of compound 6 was prepared as follows.


Procedure A: Compound 1 (3 g, 6.54 mmol) was dissolved in ˜60 ml tetrahydrofuran. H2SO4 (95% diluted to ˜1M with water) was charged. The solution was stirred for more than one hour, after which the solvent was removed under reduced pressure. Ethyl acetate (10×) was charged. The slurry was vigorously stirred overnight, after which the solids were filtered and dried in a vacuum oven at 40° C. with nitrogen bleed.


Procedure B: Compound 1 was dissolved in tetrahydrofuran. Equal molar equivalent of 1.18 M H2SO4 in acetonitrile was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetone with a stirring bar at ambient temperature overnight, then filtered and dried in vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline Form B of compound 6.


Table 25, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 6.









TABLE 25





XRPD Peak Positions for Form B of Compound 6


Position (° 2θ)















7.1


7.6


10.1


11.4


11.6


12.4


13.7


15.2


17.3


17.8


18.7


18.7


20.3


21.7


22.9


24.1


25.4


26.0


29.7


34.7






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 70 depicts an XRPD pattern of Form B of compound 6.



FIG. 71 depicts a DSC thermogram and TGA trace of Form B of compound 6.


Form C of Compound 6


Form C of compound 6 was prepared as follows.


Procedure: Compound 1 (3 g, 6.54 mmol) was dissolved in tetrahydrofuran. Equimolar equivalent of 1.18 M H2SO4 in acetonitrile was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methanol with a stirring bar at ambient temperature overnight, after which the solids were filtered and dried in a vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline Form C of compound 6.


Table 26, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 6.









TABLE 26





XRPD Peak Positions for Form C of Compound 6


Position (° 2θ)















7.1


7.6


8.3


9.3


12.6


13.5


14.2


17.4


18.1


19.6


20.2


20.5


20.9


21.2


23.1


24.4


25.1


26.2


27.9


29.9






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 72 depicts an XRPD pattern of Form C of compound 6.



FIG. 73 depicts a DSC thermogram and TGA trace of Form C of compound 6.


Example 7
Preparation of Form A of Compound 7



embedded image



Form A of Compound 7


Form A of compound 7 was prepared as follows.


Procedure: 0.22 g of 98% sulphuric acid was diluted in 10 mL acetonitrile. 0.5 g of compound 1 was charged to the sulphuric acid solution and agitated at 22° C. for 2 hours. The solids were filtered, washed three times, each with 2 mL acetonitrile, and dried under reduced pressure at 40° C. to yield 0.61 g of the bis-sulfate salt.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 7.


Table 27, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 7.









TABLE 27





XRPD Peak Positions for Form A of Compound 7


Position (° 2θ)















7.3


8.7


9.6


11.4


11.5


14.1


17.9


18.2


19.0


19.2


20.5


20.7


21.0


22.2


23.5


24.3


24.3


27.5


28.6


31.1






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 74 depicts an XRPD pattern of Form A of compound 7.



FIG. 75 depicts a DSC thermogram and TGA trace of Form A of compound 7.



FIG. 76 depicts a DVS plot of Form A of compound 7.


Elemental analysis—Calculated: C, 40.37; H, 3.85; N, 12.84; S, 9.80; Found: C, 40.27; H, 3.92; N, 12.69; S, 9.63.


Example 8
Preparation of Forms A-D of Compound 8



embedded image



Form A of Compound 8


Form A of compound 8 was prepared as follows.


Procedure: Compound 1 was dissolved in tetrahydrofuran. An equal molar equivalent of 1.3 M p-toluenesulfonic acid monohydrate in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetonitrile with a stir bar at ambient temperature overnight, then filtered and dried in a vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline Form A of compound 8.


Table 28, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 8.









TABLE 28





XRPD Peak Positions for Form A of Compound 8


Position (° 2θ)















8.6


9.0


12.4


12.8


13.4


16.1


16.4


16.6


17.0


17.9


19.1


20.0


20.5


22.9


23.5


23.7


23.8


24.8


27.8


30.7






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 77 depicts an XRPD pattern of Form A of compound 8.



FIG. 78 depicts a DSC thermogram and TGA trace of Form A of compound 8.


Form B of Compound 8


Form B of compound 8 was prepared as follows.


Procedure: Compound 1 was dissolved in tetrahydrofuran. An equal molar equivalent of 1.3 M p-toluenesulfonic acid monohydrate in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetone with a stir bar at ambient temperature overnight, then filtered and dried in a vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline Form B of compound 8.


Table 29, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 8.









TABLE 29





XRPD Peak Positions for Form B of Compound 8


Position (° 2θ)















8.6


8.9


12.2


16.2


16.5


16.9


17.2


17.9


19.0


19.1


20.1


23.1


23.4


23.6


24.6


24.7


25.1


26.3


27.6


28.2






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 79 depicts an XRPD pattern of Form B of compound 8.



FIG. 80 depicts a DSC thermogram and TGA trace of Form B of compound 8.


Form C of Compound 8


Form C of compound 8 was prepared as follows.


Procedure: Compound 1 was dissolved in tetrahydrofuran. An equal molar equivalent of 1.3 M p-toluenesulfonic acid monohydrate in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methylethyl ketone with a stir bar at ambient temperature overnight, then filtered and dried in a vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form C of compound 8.


Table 30, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 8.









TABLE 30







XRPD Peak Positions for Form C of Compound 8










Peak No.
Position (° 2θ)














8.8
23.1



9.5
23.8



12.5
24.2



13.3
24.6



14.2
25.2



16.9
26.3



17.4
30.3



17.8
31.1



18.9
31.8



20.1
38.0








1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 81 depicts an XRPD pattern of Form C of compound 8.



FIG. 82 depicts a DSC thermogram and TGA trace of Form C of compound 8.



FIG. 83 depicts a DVS plot of Form C of compound 8.


Elemental analysis—Calculated: C, 55.23; H, 4.64; N, 13.33; S, 5.08; Found: C, 54.78; H, 4.58; N, 13.18; S, 5.35.


Form D of Compound 8


Form D of compound 8 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). An equal molar equivalent of 0.31 M p-toluenesulfonic acid monohydrate in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methanol at ambient temperature overnight. No solids were obtained. The solution was dried under nitrogen purge, then was added methyl tert-butyl ether, and the solution was slurried overnight. No solids were obtained. The solution was then dried, hexane was added, and the solution was slurried overnight, then dried in a vacuum oven.


Characterization of the resulting material demonstrated an amorphous Form D of compound 8.



FIG. 84 depicts a DSC thermogram and TGA trace of Form D of compound 8.


Example 9
Preparation of Forms A-B of Compound 9



embedded image



Form A of Compound 9


Form A of compound 9 was prepared as follows.


Procedure: Compound 1 was dissolved in tetrahydrofuran. An equal molar equivalent of 1.25 M benzenesulfonic acid in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methyl ethyl ketone (or acetone) with a stir bar at ambient temperature overnight, then filtered and dried in vacuum oven at 30° C. overnight.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 9.


Table 31, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 9.









TABLE 31







XRPD Peak Positions for Form A of Compound 9










Peak No.
Position (° 2θ)














8.9
23.6



12.5
24.0



13.2
25.2



15.6
25.5



17.6
26.1



18.0
27.0



18.5
27.9



19.1
29.6



20.6
30.3



21.7
39.5








1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 85 depicts an XRPD pattern of Form A of compound 9.



FIG. 86 depicts a DSC thermogram and TGA trace of Form A of compound 9.



FIG. 87 depicts a DVS plot of Form A of compound 9.


Elemental analysis—Calculated: C, 54.54; H, 4.41; N, 13.63; S, 5.20; Found: C, 54.20; H, 4.38; N, 13.60; S, 5.59.


Form B of Compound 9


Form B of compound 9 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). Equal molar equivalent of 0.14 M benzenesulfonic acid in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methanol at ambient temperature overnight, no solids were obtained. So it was dried under nitrogen purge, then added with methyl tert-butyl ether and slurried overnight, then dried in a vacuum oven.


Characterization of the resulting material demonstrated an amorphous Form B of compound 9.



FIG. 88 depicts a DSC thermogram and TGA trace of Form B of compound 9.


Example 10
Preparation of Forms A-B of Compound 10



embedded image



Form A of Compound 10


Form A of compound 10 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). An equal molar equivalent of 0.31 M methanesulfonic acid in acetonitrile was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in methanol at ambient temperature overnight, no solids were obtained. The material was dried under nitrogen purge, followed by the addition of methyl tert-butyl ether, overnight slurry, filtration and drying in a vacuum oven.


Characterization of the resulting material demonstrated an amorphous Form A of compound 10.



FIG. 89 depicts a DSC thermogram and TGA trace of Form A of compound 10.


Form B of Compound 10


Form B of compound 10 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). An equal molar equivalent of 0.31 M methanesulfonic acid in acetonitrile was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. Acetonitrile/water (1/1 v/v) was charged and slurried overnight, no solids were obtained. It was dried and slurried in hexane overnight and then dried in vacuum oven.


Characterization of the resulting material demonstrated a partially crystalline Form B of compound 10.


Table 32, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 10.









TABLE 32





XRPD Peak Positions for Form B of Compound 10


Position (° 2θ)















6.1


7.9


8.3


16.3


22.6






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 90 depicts an XRPD pattern of Form B of compound 10.



FIG. 91 depicts a DSC thermogram and TGA trace of Form B of compound 10.


Example 11
Preparation of Form A of Compound 11



embedded image



Form A of Compound 11


Form A of compound 11 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). Equal molar equivalent of 0.25 M maleic acid in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in ethyl acetate at ambient temperature overnight, then filtered and dried in vacuum oven at ambient temperature.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 11.


Table 33, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 11.









TABLE 33





XRPD Peak Positions for Form A of Compound 11


Position (° 2θ)















5.2


8.9


9.2


10.4


12.0


13.3


13.4


15.3


16.1


18.7


20.9


23.0


23.2


23.9


24.6


25.6


26.1


26.3


26.7


27.0






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 92 depicts an XRPD pattern of Form A of compound 11.



FIG. 93 depicts a DSC thermogram and TGA trace of Form A of compound 11.


Elemental analysis—Calculated: C, 54.54; H, 4.41; N, 13.63; S, 5.20; Found: C, 54.20; H, 4.38; N, 13.60; S, 5.59.


Example 12
Preparation of Forms A-C of Compound 12



embedded image



Form A of Compound 12


Form A of compound 12 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). Equal molar equivalent of 0.15 M oxalic acid in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetonitrile at ambient temperature overnight, then filtered and dried in vacuum oven at ambient temperature.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form A of compound 12.


Table 34, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form A of compound 12.









TABLE 34





XRPD Peak Positions for Form A of Compound 12


Position (° 2θ)















5.4


5.8


6.8


9.7


10.3


12.3


13.4


14.4


16.4


17.4


17.7


20.3


22.0


23.4


23.7


24.9


25.2


26.9


30.2


35.4






1 In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 94 depicts an XRPD pattern of Form A of compound 12.



FIG. 95 depicts a DSC thermogram and TGA trace of Form A of compound 12.


Form B of Compound 12


Form B of compound 12 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). Equal molar equivalent of 0.15 M oxalic acid in MeOH was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in acetone at ambient temperature overnight, then filtered and dried in vacuum oven at ambient temperature.


Characterization of the resulting material demonstrated a crystalline Form B of compound 12.


Table 35, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form B of compound 12.









TABLE 35





XRPD Peak Positions for Form B of Compound 12


Position (° 2θ)















5.0


5.8


8.3


9.1


9.9


12.5


13.4


14.8


15.2


16.7


17.4


20.3


21.7


24.8


25.3


26.3


26.8


28.1


29.2


30.6






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 96 depicts an XRPD pattern of Form B of compound 12.



FIG. 97 depicts a DSC thermogram and TGA trace of Form B of compound 12.


Form C of Compound 12


Form C of compound 12 was prepared as follows.


Procedure: Compound 1 was dissolved in MeOH/CH2Cl2 (1/1 v/v, pre-mixed). Equal molar equivalent of 0.15 M oxalic acid in methanol was charged. The sample was shaken at ambient temperature at 200 RPM for 1 hour. The solvent was removed under nitrogen purge. The resulting solids were slurried in ethyl acetate at ambient temperature overnight, then filtered and dried in vacuum oven at ambient temperature.


Characterization of the resulting material demonstrated a crystalline, anhydrous Form C of compound 12.


Table 36, supra, is reproduced below and sets forth the X-ray diffraction peaks observed for Form C of compound 12.









TABLE 36





XRPD Peak Positions for Form C of Compound 12


Position (° 2θ)















5.6


5.8


8.4


9.3


10.1


12.4


13.4


14.9


16.2


16.6


17.5


18.5


21.8


22.2


23.4


25.2


25.9


26.3


26.9


36.5






1In this and all subsequent tables, the position 2θ is within ±0.2.








FIG. 98 depicts an XRPD pattern of Form C of compound 12.



FIG. 99 depicts a DSC thermogram and TGA trace of Form C of compound 12.


Example 13
Solubility in Water at Ambient Temperature

Each of compounds 1 through 9 were weighed into separate 8 mL clear glass vials. After addition of 3.2 mL of water, the vials were capped and shaken on an orbital shaker at 300 RPM at ambient temperature for 24 hours. The samples were removed from the shaker and the pH of each was measured using a calibrated pH meter. Next, a portion of the each sample was withdrawn and filtered using a PVDF-membrane syringe filter. The filtrate was analyzed using HPLC/UV with appropriate dilution. The solubility was reported as free base-equivalent value because the free base was used as the standard for HPLC analysis. The rest of the samples were shaken for another 24 hours for 48-hour solubility measurement using the same procedure. After the 48-hour solubility tests were run, the solid residues were recovered through filtration using 0.2 μm Nylon-membrane centrifuge tube filters at 14000 RPM for 5 min and analyzed using XRPD.


For the bis-phosphate complex and bis-sulfate salt, measurements were only performed at the 24-hour time point. The results are shown in Table 37, below.









TABLE 37







Solubility in Water at Ambient Temperature











Conc. (μg/mL)
Conc. (μg/mL)



Sample
at 24 hours
at 48 hours
pH at 48 hrs













Compound 1
0.07
0.05
6.33


(free base)


Compound 2
19.40
91.69
2.48


(phosphate salt)


Compound 3
113
Not
Not


(bis-phosphate

Determined
Determined


complex)


Compound 4
162.33
134.08
2.29


(HCl salt)


Compound 5
190.63
207.42
2.27


(HBr salt)


Compound 6
560.02
409.83
1.98


(sulfate salt)


Compound 7
1170
Not
Not


(bis-sulfate salt)

Determined
Determined


Compound 8
218.34
184.83
2.00


(tosylate salt)


Compound 9
173.63
143.49
2.10


(besylate salt)









Example 14
A Single Oral Dose Pharmacokinetic (PK) Study in Dogs with Various Salt and Free Base Forms of Compound 1

The pharmacokinetics of compound 1 (free base, administered to dosing group 1), compound 2 (phosphate salt, administered to dosing group 4), compound 4 (hydrochloride salt, administered to dosing group 2), compound 5 (bromide salt, administered to dosing group 5), and compound 6 (sulfate salt, administered to dosing group 3) were evaluated in dogs (2/sex/group) in a parallel design study. In addition, the effect of food on the systemic exposure to compounds 1, 2, 4, 5 and 6 was also evaluated. Compounds 1, 2, 4, 5 and 6 were orally administered at 15 mg/kg to dogs in a fasted state (overnight fast) on day 1 and administered under fed conditions (a meal was provided one hour prior to dosing) on day 9. Doses were administered as a suspension in 5% Captisol/0.2% Tween 80/0.05M Citrate buffer (compound 1) and 5% Captisol/0.2% Tween (compounds 2, 4, 5 and 6). Animals were habituated (days 2 to 8) to consume their entire daily ration (˜400 g) in 1 hour. Pharmacokinetic samples were collected on days 1 and 9 at 0, 0.5, 1, 2, 4, 6, 8, 12, and 24 hours after dosing. The summary of pharmacokinetic parameters are shown in Table 38 below.









TABLE 38







Mean (SD) Pharmacokinetic Parameters in Dogs










Day 1 (Fasted)
Day 9 (Fed)












Cmax
AUClast
Cmax
AUClast


Salt Type
ng/mL
(ng · h/mL)
ng/mL
(ng · h/mL)





Compound 1,
  98.1
286
293
1890


Form C
  (51.9)
(162)
(112)
 (980)


Compound 4,
323
856
818
3350


Form A
(135)
(285)
(47)
 (839)


Compound 2,
345
1080 
790
3880


Form A
 (78)
(511)
(218)
(1250)


Compound 6,
364
1120 
740
5590


Form A
(204)
(597)
(433)
(2480)


Compound 5,
393
1680 
826
5030


Form A
(171)
(1210) 
(211)
(1020)





Number of animals used were 2/sex/group






Compounds 2, 4, 5 and 6 demonstrated enhanced oral bioavailability in dogs when compared to Compound 1 under both fasted and fed conditions with their mean systemic exposure (AUClast) to Compound 1 increasing by approximately either 3- to 6-fold (fasted; see FIG. 100(a)) or 2- to 3-fold (fed; see FIG. 100(b)).


In dogs, a positive food effect was observed with both free base (Compound 1) and salt forms (Compounds 2, 4, 5 and 6). Systemic exposure increased by approximately 3- to 7-fold under fed conditions as compared to fasted conditions.


While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example.

Claims
  • 1. Compound 2:
  • 2. The compound according to claim 1, wherein said compound is crystalline.
  • 3. The compound according to claim 1, wherein said compound is a crystalline solid substantially free of amorphous compound 2.
  • 4. The compound according to claim 2, having one or more peaks in its XRPD selected from those at about 6.8, about 10.1, and about 20.8 degrees 2-theta.
  • 5. The compound according to claim 4, having at least two peaks in its XRPD selected from those at about 6.8, about 10.1, and about 20.8 degrees 2-theta.
  • 6. The compound according to claim 5, wherein said compound is of Form A.
  • 7. The compound according to claim 2, having an XRPD substantially similar to that depicted in FIG. 13.
  • 8. The compound according to claim 2, having one or more peaks in its XRPD selected from those at about 3.6, about 7.3, and about 15.0 degrees 2-theta.
  • 9. The compound according to claim 8, having at least two peaks in its XRPD selected from those at about 3.6, about 7.3, and about 15.0 degrees 2-theta.
  • 10. The compound according to claim 9, wherein said compound is of Form B.
  • 11. The compound according to claim 2, having an XRPD substantially similar to that depicted in FIG. 17.
  • 12. The compound according to claim 2, having one or more peaks in its XRPD selected from those at about 8.4, about 9.3, and about 16.5 degrees 2-theta.
  • 13. The compound according to claim 12, having at least two peaks in its XRPD selected from those at about 8.4, about 9.3, and about 16.5 degrees 2-theta.
  • 14. The compound according to claim 13, wherein said compound is of Form C.
  • 15. The compound according to claim 2, having an XRPD substantially similar to that depicted in FIG. 21.
  • 16. The compound according to claim 2, having one or more peaks in its XRPD selected from those at about 9.1, about 10.4, and about 25.1 degrees 2-theta.
  • 17. The compound according to claim 16, having at least two peaks in its XRPD selected from those at about 9.1, about 10.4, and about 25.1 degrees 2-theta.
  • 18. The compound according to claim 17, wherein said compound is of Form D.
  • 19. The compound according to claim 2, having an XRPD substantially similar to that depicted in FIG. 25.
  • 20. A pharmaceutical composition comprising the compound according to claim 1 and a pharmaceutically acceptable carrier or excipient.
  • 21. The pharmaceutical composition according to claim 20, wherein said pharmaceutical composition is substantially free of impurities.
CROSS-REFERENCE TO RELATED APPLICATIONS

The present application is a U.S. national stage entry of International PCT application number PCT/US15/44783, filed Aug. 12, 2015, which claims priority to U.S. provisional application No. 62/037,066, filed Aug. 13, 2014, the entirety of each of which is hereby incorporated herein by reference.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2015/044783 8/12/2015 WO 00
Publishing Document Publishing Date Country Kind
WO2016/025561 2/18/2016 WO A
US Referenced Citations (278)
Number Name Date Kind
3609152 Hess et al. Sep 1971 A
4337341 Zimmerman Jun 1982 A
4879303 Davison et al. Nov 1989 A
5786476 Fuso Jul 1998 A
5958935 Davis et al. Sep 1999 A
6093716 Davis et al. Jul 2000 A
6114333 Davis et al. Sep 2000 A
6127376 Davey et al. Oct 2000 A
6160010 Uckun et al. Dec 2000 A
6262088 Phillips Jul 2001 B1
6469168 Simonek et al. Oct 2002 B1
6579983 Batchelor et al. Jun 2003 B1
6593326 Bradbury et al. Jul 2003 B1
6838464 Pease et al. Jan 2005 B2
6878717 De Corte et al. Apr 2005 B2
6908906 Takita et al. Jun 2005 B2
6939874 Harmange et al. Sep 2005 B2
7037917 De Corte et al. May 2006 B2
7060827 Singh et al. Jun 2006 B2
7122542 Singh et al. Oct 2006 B2
7125879 Guillemont et al. Oct 2006 B2
7176212 Breault et al. Feb 2007 B2
7202033 Prescott et al. Apr 2007 B2
7241769 Stadtmueller et al. Jul 2007 B2
7276510 Kukla et al. Oct 2007 B2
7282504 Armistead et al. Oct 2007 B2
7288547 Lucking et al. Oct 2007 B2
7329671 Singh et al. Feb 2008 B2
7329672 Singh et al. Feb 2008 B2
7332484 Singh et al. Feb 2008 B2
7432377 Chew et al. Oct 2008 B2
7435814 Singh et al. Oct 2008 B2
7449458 Bhamidipati et al. Nov 2008 B2
7452879 Singh et al. Nov 2008 B2
7485724 Singh et al. Feb 2009 B2
7491732 Li et al. Feb 2009 B2
7498435 Singh et al. Mar 2009 B2
7500137 Park Mar 2009 B2
7504396 Nunes et al. Mar 2009 B2
7514444 Honigberg et al. Apr 2009 B2
7514445 Freyne et al. Apr 2009 B2
7514446 Davis-Ward et al. Apr 2009 B2
7517886 Singh et al. Apr 2009 B2
7528143 Noronha et al. May 2009 B2
7531548 Guillemont et al. May 2009 B2
7540908 Sao et al. Jun 2009 B2
7540909 Sao et al. Jun 2009 B2
7550460 Singh et al. Jun 2009 B2
7553357 Sao et al. Jun 2009 B2
7557207 Cooper et al. Jul 2009 B2
7557210 Singh et al. Jul 2009 B2
7582648 Singh et al. Sep 2009 B2
7589200 Singh et al. Sep 2009 B2
7642351 Singh et al. Jan 2010 B2
7655797 Singh et al. Feb 2010 B2
7659280 Clough et al. Feb 2010 B2
7713987 Bhamidipati et al. May 2010 B2
7718662 Chen et al. May 2010 B1
7741330 Chen et al. Jun 2010 B1
7803939 Singh et al. Sep 2010 B2
7812029 Singh et al. Oct 2010 B1
7820819 Singh et al. Oct 2010 B2
7825116 Singh et al. Nov 2010 B2
7834024 Li et al. Nov 2010 B2
7858633 Li et al. Dec 2010 B2
7863286 Argade et al. Jan 2011 B2
7879984 Martin et al. Feb 2011 B2
7884111 Argade et al. Feb 2011 B2
7893074 Garcia-Echeverria et al. Feb 2011 B2
7906644 Singh et al. Mar 2011 B2
7915273 Argade et al. Mar 2011 B2
7947698 Atuegbu et al. May 2011 B2
7989465 Singh et al. Aug 2011 B2
8003789 De Corte et al. Aug 2011 B2
8088781 Honigberg et al. Jan 2012 B2
8133900 Hood et al. Mar 2012 B2
8148525 Singh et al. Apr 2012 B2
8153640 Guillemont et al. Apr 2012 B2
8158621 Singh et al. Apr 2012 B2
8163902 Bhamidipati et al. Apr 2012 B2
8188276 Singh et al. May 2012 B2
8193197 Li et al. Jun 2012 B2
8206711 White et al. Jun 2012 B2
8263590 Garcia-Echeverria et al. Sep 2012 B2
8299087 Li et al. Oct 2012 B2
8304422 Atuegbu et al. Nov 2012 B2
8334296 Singh et al. Dec 2012 B2
8338439 Singh et al. Dec 2012 B2
8399433 Appari et al. Mar 2013 B2
8399450 Michellys et al. Mar 2013 B2
8399472 Li et al. Mar 2013 B2
8410266 Singh et al. Apr 2013 B2
8415365 Li et al. Apr 2013 B2
8450335 Singh et al. May 2013 B2
8501751 Honigberg et al. Aug 2013 B2
8530466 Masuda et al. Sep 2013 B2
8530655 De Corte et al. Sep 2013 B2
8557806 Singh et al. Oct 2013 B2
8563568 Witowski et al. Oct 2013 B2
8609679 Singh et al. Dec 2013 B2
8697694 Arasappan et al. Apr 2014 B2
8710222 Singh et al. Apr 2014 B2
8735404 Honigberg et al. May 2014 B2
8748438 Honigberg et al. Jun 2014 B2
8748597 Singh et al. Jun 2014 B2
8796255 Lee et al. Aug 2014 B2
8822685 Singh et al. Sep 2014 B2
8835430 Singh et al. Sep 2014 B2
8853397 Singh et al. Oct 2014 B2
8883435 Honigberg et al. Nov 2014 B2
8883803 Honigberg et al. Nov 2014 B2
8975249 Lee et al. Mar 2015 B2
9012462 Wang et al. Apr 2015 B2
9145387 Haq Sep 2015 B2
9212181 Singh et al. Dec 2015 B2
9296737 Singh et al. Mar 2016 B2
9409921 Singh et al. Aug 2016 B2
9504686 Haq et al. Nov 2016 B2
9561228 Haq et al. Feb 2017 B2
9796700 Haq et al. Oct 2017 B2
20020147339 Batchelor et al. Oct 2002 A1
20040002395 Poynor Jan 2004 A1
20040019067 Armistead et al. Jan 2004 A1
20040023957 Wang et al. Feb 2004 A1
20040077661 Arbiser Apr 2004 A1
20040180914 Batchelor et al. Sep 2004 A1
20050004125 Freyne et al. Jan 2005 A1
20050014753 Ding et al. Jan 2005 A1
20050085637 Cheung et al. Apr 2005 A1
20050209221 Nunes et al. Sep 2005 A1
20050272083 Seshagiri Dec 2005 A1
20060030018 Zuccola et al. Feb 2006 A1
20060079543 Sum et al. Apr 2006 A1
20060084644 Pal et al. Apr 2006 A1
20060084645 Pal et al. Apr 2006 A1
20060100227 Baenteli et al. May 2006 A1
20060148800 Stadtmueller et al. Jul 2006 A1
20060160803 Adams et al. Jul 2006 A1
20060247241 Garcia-Echeverria et al. Nov 2006 A1
20060247262 Baenteli et al. Nov 2006 A1
20060270694 Wong Nov 2006 A1
20060293311 Li et al. Dec 2006 A1
20070010668 Davis-Ward et al. Jan 2007 A1
20070032493 Foley et al. Feb 2007 A1
20070066658 Chappell Mar 2007 A1
20070141143 Smithey et al. Jun 2007 A1
20070203161 Argade et al. Aug 2007 A1
20070203162 Li et al. Aug 2007 A1
20070208022 Guillemont et al. Sep 2007 A1
20070208034 Stadlwieser Sep 2007 A1
20070259904 Noronha et al. Nov 2007 A1
20080009484 Argade et al. Jan 2008 A1
20080009494 Li et al. Jan 2008 A1
20080021020 Argade et al. Jan 2008 A1
20080027045 Argade et al. Jan 2008 A1
20080039622 Singh et al. Feb 2008 A1
20080058358 Luecking et al. Mar 2008 A1
20080076921 Honigberg et al. Mar 2008 A1
20080132504 Garcia-Echeverria et al. Jun 2008 A1
20080139582 Honigberg et al. Jun 2008 A1
20080167330 Luecking et al. Jul 2008 A1
20080176866 Jautelat et al. Jul 2008 A1
20080182852 Johnson et al. Jul 2008 A1
20080194603 Li et al. Aug 2008 A1
20080194605 Heinrich et al. Aug 2008 A1
20080207613 Styles et al. Aug 2008 A1
20080207913 Breitenkamp et al. Aug 2008 A1
20080214501 Pan et al. Sep 2008 A1
20080221089 Argade et al. Sep 2008 A1
20080260754 Li et al. Oct 2008 A1
20080261977 Eatherton et al. Oct 2008 A1
20080279867 Atuegbu et al. Nov 2008 A1
20080300268 Singh et al. Dec 2008 A1
20080312438 Singh et al. Dec 2008 A1
20090030197 Chew et al. Jan 2009 A1
20090088371 Grossbard Apr 2009 A1
20090131436 Imbach et al. May 2009 A1
20090137588 Singh et al. May 2009 A1
20090156622 Singh et al. Jun 2009 A1
20090171086 Singh et al. Jul 2009 A1
20090181987 Honigberg et al. Jul 2009 A1
20090215803 Rice et al. Aug 2009 A1
20090286778 Combs et al. Nov 2009 A1
20090298830 Mann et al. Dec 2009 A1
20090318407 Bauer et al. Dec 2009 A1
20100004270 Honigberg et al. Jan 2010 A1
20100016296 Singh et al. Jan 2010 A1
20100022561 Honigberg et al. Jan 2010 A1
20100029610 Singh et al. Feb 2010 A1
20100041677 Honigberg et al. Feb 2010 A1
20100081679 Greul et al. Apr 2010 A1
20100088912 Higgs et al. Apr 2010 A1
20100144706 Zahn et al. Jun 2010 A1
20100173285 Varmus et al. Jul 2010 A1
20100197918 Singh et al. Aug 2010 A1
20100249092 Singh et al. Sep 2010 A1
20100254905 Honigberg et al. Oct 2010 A1
20110027856 Li et al. Feb 2011 A1
20110028405 Harrison et al. Feb 2011 A1
20110039868 Honigberg et al. Feb 2011 A1
20110071158 Sapountzis et al. Mar 2011 A1
20110098280 Garcia-Echeverria et al. Apr 2011 A1
20110098288 Major et al. Apr 2011 A1
20110105472 Greul et al. May 2011 A1
20110144330 Singh et al. Jun 2011 A1
20110190261 Dong et al. Aug 2011 A1
20110207736 Gray et al. Aug 2011 A1
20110212077 Noronha et al. Sep 2011 A1
20110224235 Honigberg et al. Sep 2011 A1
20110245156 Sielecki-Dzurdz Oct 2011 A1
20110245284 Greul et al. Oct 2011 A1
20110281322 Honigberg et al. Nov 2011 A1
20110281850 Flynn et al. Nov 2011 A1
20120021434 Foley et al. Jan 2012 A1
20120040968 Shimada et al. Feb 2012 A1
20120065201 Honigberg et al. Mar 2012 A1
20120071497 Buggy et al. Mar 2012 A1
20120077832 Witowski et al. Mar 2012 A1
20120083006 Ramsden et al. Apr 2012 A1
20120087915 Buggy et al. Apr 2012 A1
20120088912 Honigberg et al. Apr 2012 A1
20120094999 Gray et al. Apr 2012 A1
20120101113 Honigberg et al. Apr 2012 A1
20120101114 Honigberg et al. Apr 2012 A1
20120142667 Ramsden et al. Jun 2012 A1
20120149687 Lee et al. Jun 2012 A1
20120149722 Lee et al. Jun 2012 A1
20120157426 Lee et al. Jun 2012 A1
20120165328 Honigberg et al. Jun 2012 A1
20120165332 Major et al. Jun 2012 A1
20120172385 Harrison et al. Jul 2012 A1
20120184013 Honigberg et al. Jul 2012 A1
20120184567 Honigberg et al. Jul 2012 A1
20120190697 Guillemont et al. Jul 2012 A1
20120202264 Honigberg et al. Aug 2012 A1
20120213795 Li et al. Aug 2012 A1
20120270237 Ramsden et al. Oct 2012 A9
20120296089 Honigberg et al. Nov 2012 A1
20120316135 Dalgarno et al. Dec 2012 A1
20120329130 Honigberg et al. Dec 2012 A1
20130035334 Honigberg et al. Feb 2013 A1
20130065879 Singh et al. Mar 2013 A1
20130065899 Singh et al. Mar 2013 A1
20130072469 Singh et al. Mar 2013 A1
20130109709 Witowski et al. May 2013 A1
20130137708 Garske et al. May 2013 A1
20130142807 Li et al. Jun 2013 A1
20130150349 Singh et al. Jun 2013 A1
20130165462 Singh et al. Jun 2013 A1
20130231306 Crew et al. Sep 2013 A1
20130267530 Lai Oct 2013 A1
20130267531 Lai et al. Oct 2013 A1
20140057929 Witowski et al. Feb 2014 A1
20140134265 Buggy et al. May 2014 A1
20140140991 Daniel et al. May 2014 A1
20140142123 Honigberg et al. May 2014 A1
20140142128 Daniel et al. May 2014 A1
20140142129 Daniel et al. May 2014 A1
20140163027 Verner et al. Jun 2014 A1
20140163046 Honigberg et al. Jun 2014 A1
20140179720 Tester et al. Jun 2014 A1
20140187564 Honigberg et al. Jul 2014 A1
20140187565 Honigberg et al. Jul 2014 A1
20140213574 Singh et al. Jul 2014 A1
20140228322 Haq et al. Aug 2014 A1
20140303154 Singh et al. Oct 2014 A1
20140303191 Buggy et al. Oct 2014 A1
20140330007 Singh et al. Nov 2014 A1
20140371241 Buggy et al. Dec 2014 A1
20150005297 Singh et al. Jan 2015 A1
20150025055 Lee et al. Jan 2015 A1
20150038518 Balasubramanian Feb 2015 A1
20160002176 Haq et al. Jan 2016 A1
20160082008 Haq et al. Mar 2016 A1
20160303121 Singh et al. Oct 2016 A1
20170100397 Singh et al. Apr 2017 A1
20170137406 Haq et al. May 2017 A1
20170210729 Haq et al. Jul 2017 A1
Foreign Referenced Citations (169)
Number Date Country
2009251863 Dec 2009 AU
2375182 Dec 2000 CA
2463822 May 2003 CA
2553729 Aug 2005 CA
2608367 Dec 2006 CA
2673125 Apr 2008 CA
2710118 Jul 2009 CA
2717529 Sep 2009 CA
2757671 Oct 2010 CA
2760061 Nov 2010 CA
2763720 Dec 2010 CA
102558149 Jul 2012 CN
103159742 Jun 2013 CN
103664878 Mar 2014 CN
1 054 004 Nov 2000 EP
1187816 Mar 2002 EP
1448556 Aug 2004 EP
1597251 Nov 2005 EP
1904457 Apr 2008 EP
1990342 Nov 2008 EP
2089369 Aug 2009 EP
2234986 Oct 2010 EP
2276747 Jan 2011 EP
2414337 Feb 2012 EP
2428508 Mar 2012 EP
2443095 Apr 2012 EP
H07-41461 Feb 1995 JP
WO-9628427 Sep 1996 WO
WO-9719065 May 1997 WO
WO-9931073 Jun 1999 WO
WO-00027825 May 2000 WO
WO-00046203 Aug 2000 WO
WO-00078731 Dec 2000 WO
WO-01047897 Jul 2001 WO
WO-01060816 Aug 2001 WO
WO-01064654 Sep 2001 WO
WO-01064655 Sep 2001 WO
WO-01085699 Nov 2001 WO
WO-02064586 Aug 2002 WO
WO-02083653 Oct 2002 WO
WO-03016306 Feb 2003 WO
WO-03030909 Apr 2003 WO
WO-03037891 May 2003 WO
WO-03063794 Aug 2003 WO
WO-03066601 Aug 2003 WO
WO-2004014382 Feb 2004 WO
WO-2004031232 Apr 2004 WO
WO-2004056786 Jul 2004 WO
WO-2004069812 Aug 2004 WO
WO-2004074244 Sep 2004 WO
WO-2004080980 Sep 2004 WO
WO-2004096224 Nov 2004 WO
WO-2005013996 Feb 2005 WO
WO-2005016893 Feb 2005 WO
WO-2005016894 Feb 2005 WO
WO-2005026130 Mar 2005 WO
WO-2005026158 Mar 2005 WO
WO-2005063722 Jul 2005 WO
WO-2005070890 Aug 2005 WO
WO-2005118544 Dec 2005 WO
WO-2006021544 Mar 2006 WO
WO-2006038001 Apr 2006 WO
WO-2006045066 Apr 2006 WO
WO-2006053109 May 2006 WO
WO-2006055561 May 2006 WO
WO-2006068770 Jun 2006 WO
WO-2006074057 Jul 2006 WO
WO-2006078846 Jul 2006 WO
WO-2006101977 Sep 2006 WO
WO-2006108487 Oct 2006 WO
WO-2006124874 Nov 2006 WO
WO-2006128129 Nov 2006 WO
WO-2006129100 Dec 2006 WO
WO-2006133426 Dec 2006 WO
WO-2007027238 Mar 2007 WO
WO-2007048064 Apr 2007 WO
WO-2007053452 May 2007 WO
WO-2007056151 May 2007 WO
WO-2007085833 Aug 2007 WO
WO-2007089768 Aug 2007 WO
WO-2007098507 Aug 2007 WO
WO-2007113254 Oct 2007 WO
WO-2007113256 Oct 2007 WO
WO-2007120339 Oct 2007 WO
WO-2007120980 Oct 2007 WO
WO-2007125351 Nov 2007 WO
WO-2008005538 Jan 2008 WO
WO-2008009458 Jan 2008 WO
WO-2008025556 Mar 2008 WO
WO-2008049123 Apr 2008 WO
WO-2008064274 May 2008 WO
WO-2008073687 Jun 2008 WO
WO-2008074515 Jun 2008 WO
WO-2008079719 Jul 2008 WO
WO-2008079907 Jul 2008 WO
WO-2008080964 Jul 2008 WO
WO-2008080965 Jul 2008 WO
WO-2008088303 Jul 2008 WO
WO-2008092199 Aug 2008 WO
WO-2008093687 Aug 2008 WO
WO-2008107096 Sep 2008 WO
WO-2008115738 Sep 2008 WO
WO-2008115742 Sep 2008 WO
WO-2008118822 Oct 2008 WO
WO-2008118823 Oct 2008 WO
WO-2008147831 Dec 2008 WO
WO-2009012421 Jan 2009 WO
WO-2009017838 Feb 2009 WO
WO-2009029682 Mar 2009 WO
WO-2009032668 Mar 2009 WO
WO-2009032694 Mar 2009 WO
WO-2009032703 Mar 2009 WO
WO-2009080638 Jul 2009 WO
WO-2009105675 Aug 2009 WO
WO-2009112490 Sep 2009 WO
WO-2009115267 Sep 2009 WO
WO-2009127642 Oct 2009 WO
WO-2009132202 Oct 2009 WO
WO-2009136995 Nov 2009 WO
WO-2009143389 Nov 2009 WO
WO-2009158571 Dec 2009 WO
WO-2010025833 Mar 2010 WO
WO-2010077740 Jul 2010 WO
WO-2010081679 Jul 2010 WO
WO-2010112210 Oct 2010 WO
WO-2010128659 Nov 2010 WO
WO-2010129053 Nov 2010 WO
WO-2010141406 Dec 2010 WO
WO-2010146132 Dec 2010 WO
WO-2011079231 Jun 2011 WO
WO-2011090760 Jul 2011 WO
WO-2011140338 Nov 2011 WO
WO-2011153514 Dec 2011 WO
WO-2011153553 Dec 2011 WO
WO-2012021444 Feb 2012 WO
WO-2012061299 May 2012 WO
WO-2012061303 May 2012 WO
WO-2012061415 May 2012 WO
WO-2012064706 May 2012 WO
WO-2012078492 Jun 2012 WO
WO-2012100459 Aug 2012 WO
WO-2012135801 Oct 2012 WO
WO-2012151561 Nov 2012 WO
WO-2012158843 Nov 2012 WO
WO-2012170976 Dec 2012 WO
WO-2013063401 May 2013 WO
WO-2013138495 Sep 2013 WO
WO-2013138502 Sep 2013 WO
WO-2013173518 Nov 2013 WO
WO-2014025128 Feb 2014 WO
WO-2014039452 Mar 2014 WO
WO-2014074580 May 2014 WO
WO-2014081709 May 2014 WO
WO-2014081712 May 2014 WO
WO-2014081714 May 2014 WO
WO-2014100748 Jun 2014 WO
WO-2014124230 Aug 2014 WO
WO-PCTUS1544783 Aug 2015 WO
WO-PCTUS1544793 Aug 2015 WO
WO-PCTUS1544890 Aug 2015 WO
WO-PCTUS1544917 Aug 2015 WO
WO-PCTUS1544918 Aug 2015 WO
WO-PCTUS1544919 Aug 2015 WO
WO-PCTUS1544928 Aug 2015 WO
WO-PCTUS1544929 Aug 2015 WO
WO-PCTUS1544930 Aug 2015 WO
WO-PCTUS1544931 Aug 2015 WO
WO-PCTUS1544932 Aug 2015 WO
WO-PCTUS1544936 Aug 2015 WO
Non-Patent Literature Citations (143)
Entry
Deschenes-Simard et al. Cancer Res. 2014, 74(2); 412-9.
Little et al. Oncogene (2013) 32, 1207-1215.
Cohen et al., Current Opinion in Chemical Biology, 3,459-465, 1999.
Freshney et al.,Culture of Animal Cells, A Manual of Basic Technique, Alan R. Liss, Inc., 1983, New York, p. 4.
Dermer et al., Bio/Technology, 1994, 12:320.
Golub et al., Science, 286, 531-537, 1999.
U.S. Appl. No. 13/518,833, filed Jun. 22, 2012, Gray et al.
U.S. Appl. No. 13/882,958, filed Jul. 8, 2013, Pandey et al.
U.S. Appl. No. 15/422,617, filed Feb. 2, 2017, Haq et al.
Adult Non-Hodgkin Lymphoma Treatment (PDQ®), Nat. Can. Inst., retreived Jul. 28, 2014 from web: http://www.cancer.gov/cancertopics/pdg/treatment/adult-non-hodqkins/HealthProfessional/page1.
Advani, R.H. et al., Bruton Tyrosine Kinase Inhibitor Ibrutinib (PCI-32765) Has Significant Activity in Patients With Relapsed/Refractory B-Cell Malignancies, J. Clin. Oncol., pp. 1-9 (2012).
Aliagas-Martin, I. et al., A class of 2,4-bisanilinopyrimidine Aurora A inhibitors with unusually high selectivity against Aurora B, J. Med. Chem. 52:3300-3307 (2009).
Andrulis, I. et al., Neu/ErbB-2 amplification identifies a poor-prognosis group of women with node-negative breast cancer, J Clin Oncol 16:1340-9 (1998).
Aronov, A.M. et al., Flipped out: structure-guided design of selective pyrazolylpyrrole ERK inhibitors, J. Med. Chem., 50(6)1280-7 (2007).
Aronov, A.M. et al., Structure-guided design of potent and selective pyrimidylpyrrole inhibitors of extracellular signal-regulated kinase (ERK) using conformational control, J. Med. Chem., 52(20):6362-8 (2009).
Balmana et al., BRCA in breast cancer: ESMO Clinical Recommendations, Annals of Oncology, 20(Supplement 4): iv19-iv20 (2009).
Bamborough, P. et al., N-4-Pyrimidinyl-1H-indazol-4-amine inhibitors of Lck: Indazoles as phenol isosteres with improved pharmacokinetics, Bioorg. & Med. Chem. Lett. 17:4363-4368 (2007).
Brown, J.R. et al., Phase Ib trial of AVL-292, a covalent inhibitor of Bruton's tyrosine kinase (Btk), in chronic lymphocytic leukemia (CLL) and B-non-Hodgkin lymphoma (B-NHL), J. Clin. Oncol., 30, (2012).
Brunton, L.L. et al., eds., Chemotherapy of Neoplastic Diseases, in Goodman & Gilman's: The Pharmacological Basis of Therapeutics, 11th edition, pp. 853-908 (2008).
Buggy, J.J. et al., Bruton tyrosine kinase (BTK) and its role in B-cell malignancy, Int. Rev. Immunol., 31(2): 119-32 (2012).
Calvo, E. et al., Administration of CI-1033, an Irreversible Pan-erbB Tyrosine Kinase Inhibitor, Is Feasible on a 7-Day Off Schedule: A Phase I Pharmacokinetic and Food Effect Study, Clinical Cancer Research, 10: 7112-7120 (2004).
Carter, T. et al, Inhibition of drug-resistant mutants of ABL, KIT, and EGF receptor kinases, Proc. Natl. Acad. Sci. USA 102(31):11011-11016 (2005).
Chabner, B.A. et al., Chemotherapy of Neoplastic Diseases, Neoplastic Agents, Goodman & Gilman's: The Pharmacological Basis of Therapeutics, 11th edition, Brunton, L.L. et al., eds., pp. 1315-1403 (2006).
Clovis Oncology, Press Release, “Clovis Oncology's CO-1686 Demonstrates Compelling Clinical Activity and Progression-free Survival (PFS) in Updated Phase 1/2 Study Results in Patients with EGFR-Mutant Non-small Cell Lung Cancer (NSCLC)”, May 31, 2014.
Cohen, M. et al., Structural bioinformatics-based design of selective, irreversible inhibitors, Science 308:1318-1321 (2005).
Curto, M. et al., Contact-dependent inhibition of EGFR signaling by Nf2/Merlin, J Cell Biol 177:893-903 (2007).
Dickson, M.A., and Schwartz, G.K., Development of cell-cycle inhibitors for cancer therapy, Current Oncology, 16(2): 36-43 (2009).
Ding, K. et al., Design, Synthesis and Biological Evaluation of Novel Conformationally Constrained Inhibitors Targeting Epidermal Growth Factor Receptor T790M mutant, J. Med. Chem., Just Accepted Manuscript, 1-36 (2012).
Evans, E. et al.,. Clinical Development of AVL-292; A Potent, Selective Covalent Btk Inhibitor for the Treatment of B Cell Malignancies, Blood (ASH Annual Meeting Abstracts), 118: 3485 (2011).
Eve, H.E. et al.,. Single-agent lenalidomide in relapsed/refractory mantle cell lymphoma: results from a UK phase II study suggest activity and possible gender differences, Br. J. Haematol., 159(2): 154-63 (2012).
Fabian, M. et al., A small molecule-kinase interaction map for clinical kinase inhibitors, Nature Biotechnology, 23(3): 329 (2005).
Fallon, K. et al., Constitutive activation of the neuregulin-1/erbB signaling pathway promotes the proliferation of a human peripheral neuroepithelioma cell line, J Neuro Oncol 66:273-84 (2004).
Frank, D., STAT signaling in the pathogenesis and treatment of cancer, Mol. Med. 5 :432-456 (1999).
Friday, B.B., and Adjei, A.A., Advances in targeting the Ras/Raf/MEK/Erk mitogen-activated protein kinase cascade with MEK inhibitors for cancer therapy, Clinical Cancer Research, 14(2): 342-346 (2008).
Friedberg, J.W. et al., Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia, Blood, 115(13): 2578-85 (2010).
Fry, D. et al., Specific, irreversible inactivation of the epidermal growth factor receptor and erbB2, by a new class of tyrosine kinase inhibitor, Proc. Natl. Acad. Sci. USA 95:12022-12027 (1998).
Galustian, C. et al., Thalidomide-derived immunomodulatory drugs as therapeutic agents, Expert. Opin. Biol. Ther., 4(12): 1963-70 (2004).
Ghoneim, K., Synthesis and evaluation of some 2-, 4-, di- substituted -6-methylpyrimidine derivatives for antimicrobial activity, J. Indian Chem. Soc. 63(10):914-917 (1986).
Ghosh, D., 2-4-bis (arylamino) -5- methylpyrimidines as antimicrobial agents, J. Med. Chem. 10(5):974 (1967).
Ghosh, D., 2-4-bis (arylamino) -6- methylpyrimidines as antimicrobial agents, J. Indian Chem. Soc. 58(5):512-573 (1981).
Gonzales, A. et al, Antitumor activity and pharmacokinetic properties of PF-00299804, a second-generation, irreversible pan-erbB receptor tyrosine kinase inhibitor, Mol. Cancer Ther. 7(7):1880-1889 (2008).
Gunnellini, M. and Falchi, L., Therapeutic Activity of Lenalidomide in Mantle Cell Lymphoma and Indolent Non-Hodgkin's Lymphomas, Adv. Hematol., Article ID 523842, 7 pages (2012).
Hacken, E.T. and Burger, J.A., Microenvironment dependency in Chronic Lymphocytic Leukemia: The basis for new targeted therapies, Pharmacology & Therapeutics, http://dx.doi.org/10.1016/j.pharmthera.2014.07.003, (2014).
Harris N.L. et al., Lymphoma classification: from Real to WHO and beyond. In: DeVita VT, Hellman S, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology Updates. Philadelphia, Pa: Lippincott-Raven, 13(3): 1-14 (1999).
Hernandez-Ilizaliturri, F.J. et al., Higher response to lenalidomide in relapsed/refractory diffuse large B-cell lymphoma in nongerminal center B-cell-like than in germinal center B-cell-like phenotype, Cancer, 117(22): 5058-66 (2011).
Hur, W. et al., Clinical stage EGFR inhibitors irreversibly alkylate Bmx kinase, Bioorg. Med. Chem. Lett. 18:5916-5919 (2008).
International Search Report for PCT/US09/48784, 8 pages (dated Nov. 16, 2009).
International Search Report for PCT/US10/31714, 4 pages (dated Aug. 13, 2010).
International Search Report for PCT/US10/62432, 4 pages (dated May 26, 2011).
International Search Report for PCT/US11/58610, 4 pages (dated Mar. 27, 2012).
International Search Report for PCT/US11/58616, 3 pages (dated Mar. 27, 2012).
International Search Report for PCT/US11/59726, 3 pages (dated Mar. 20, 2012).
International Search Report for PCT/US13/70772, 3 pages (dated Mar. 25, 2014).
International Search Report for PCT/US13/70776, 4 pages (dated Mar. 25, 2014).
International Search Report for PCT/US2014/015256, 6 pages (dated Aug. 5, 2014).
International Search Report for PCT/US2015/044783, 2 pages (dated Oct. 30, 2015).
International Search Report of PCT/US09/48784, 8 pages (dated Nov. 16, 2009).
International Search Report of PCT/US11/46926, 2 pages (dated Dec. 22, 2011).
International Search Report of PCT/US13/30982, 2 pages (dated May 30, 2013).
International Search Report of PCT/US13/30996, 2 pages (dated May 30, 2013).
International Search Report of PCT/US13/70766, 4 pages (dated Mar. 25, 2014).
Irish, J. et al., Altered B-cell receptor signaling kinetics distinguish human follicular lymphoma B cells from tumor-infiltrating nonmalignant B cells, Blood, 108(9): 3135-42 (2006).
Jaffe E.S. and Pittaluga S., Aggressive B-cell lymphomas: a review of new and old entities in the WHO classification, Hematology Am. Soc. Hematol. Educ. Program, 506-14 (2011).
Jemal, A. et al., Cancer statistics, CA Cancer J. Clin., 53(1): 5-26 (2003).
Johnson, J. et al., Relationships between drug activity in NCI preclinical in vitro and in vivo models and early clinical trials, British Journal of Cancer, 84(10): 1424-1431 (2001).
Kataja V. and Castiglione M., Primary breast cancer: ESMO clinical recommendations for diagnosis, treatment and follow-up, Annals of Oncology, 20(Supplement 4): iv10-iv14 (2009).
Kiesewetter, B. et al., A phase II study of lenalidomide in patients with extranodal marginal zone B-cell lymphoma of the mucosa associated lymphoid tissue (MALT-lymphoma), Haematologica, 98(3):353-6 (2013).
Kirken, R., Targeting Jak3 for immune suppression and allograft acceptance, Transplant. Proc. 33 :3268-3270 (2001).
Kumar, A., et al, Structure and Clinical Relevance of the Epidermal Growth Factor Receptor in Human Cancer, Journal of Clinical Oncology 26(10):1742-1751 (2008).
Kuster, B., ed., Chapters 1 and 2, Kinase Inhibitors: Methods and Protocols, Methods in Molecular Biology, vol. 795, Humana Press, 46 pages (2012).
Kwak, E. et al., Irreversible inhibitors of the EGF receptor may circumvent acquired resistance to gefitinib, Proc. Natl. Acad. Sci. USA 102:7665-7670 (2005).
Lajeunesse, D. et al., A systematic screen for dominant second-site modifiers of Merlin/NF2 phenotypes reveals an interaction with blistered/DSRF and scribbler, Genetics 158:667-79 (2001).
Leonard, J. et al., A randomized trial of lenalidomide alone versus lenalidomide plus rituximab in patients with recurrent follicular lymphoma, J. Clin. Oncol., 30 (2012).
Li, D. et al., BIBW2992, an irreversible EGFR/HER2 inhibitor highly effective in preclinical lung cancer models, Oncogene 27:4702-4711 (2008).
Liddle, J. et al., Discovery of GSK143, a highly potent, selective and orally efficacious spleen tyrosine kinase inhibitor, Bioorg. Med. Chem. Lett., 21(20):6188-94 (2011).
Lin, N. and Winer, E., New targets for therapy in breast cancer: Small molecule tyrosine kinase inhibitors, Breast Cancer Res 6:204-210 (2004).
Liu, Q. et al., Developing Irreversible Inhibitors of the Protein Kinase Cysteinome, Chemistry & Biology, 20:146-159 (2013).
Malaviya, R. et al., Targeting Janus Kinase 3 in Mast Cells Prevents Immediate Hypersensitivity Reactions and Anaphylaxis, J. Biol. Chem. 274 :27028-27038 (1999).
McClatchey, A. and Giovannini, M., Membrane organization and tumorigenesis—the NF2 tumor suppressor, Merlin, Genes Dev 19:2265-77 (2005).
McCubrey, J.A. et al., Targeting survival cascades induced by activation of Ras/Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways for effective leukemia therapy, Leukemia, 22(4): 708-722 (2008).
McDaniel J.M. et al., Molecular action of lenalidomide in lymphocytes and hematologic malignancies, Adv. Hematol., Article ID 513702, 9 pages (2012).
Merriam-Webster's Online Directory, “Prevent” download on Apr. 7, 2008 from “http//www.merriam-webster.com/dictionary/prevent”, p. 1 of 1.
Minkovsky, N. and Berezov, A., BIBW-2992, a dual receptor tyrosine kinase inhibitor for the treatment of solid tumors, Curr Opin Invest Drugs 9:1336-1346 (2008).
Montagut, C. and Settleman, J., Targeting the RAF-MEK-ERK pathway in cancer therapy, Cancer Letters, 283(2): 125-134 (2009).
Nastoupil, L.J. et al., Diffuse large B-cell lymphoma: current treatment approaches, Oncology, 26(5): 488-95 (2012).
Nelson et al., Screening for breast cancer: an update for the U.S. Preventive Services Task Force, Ann. Intern Med, 151(10): 727-737 (2009).
Ogiso, et al., Crystal Structure of the Complex of Human Epidermal Growth Factor and Receptor Extracellular Domains, Cell, vol. 110, 775-787 (2002).
Pearce, H.L. et al., Failure modes in anticancer drug discovery and development, Cancer Drug Design and Discovery Edited by Stephen Neidle, 18: 424-435 (2008).
Pelton, P. et al., Ruffling membrane, stress fiber, cell spreading and proliferation abnormalities in human Schwann cells, Oncogene 17:2195-2209 (1998).
PubChem CID 44594695. Feb. 1, 2010. [Retrieved from the Internet May 15, 2011: http://pubchem.ncbi.nlm.nih.gov/summary.cgi?cid=44594695&loc=ec_rcs].
Readinger, J. et al., Selective Targeting of ITK Blocks Multiple Steps of HIV Replication, Proc. Natl. Acad. Sci. USA 105: 6684-6689 (2008).
Rinaldi, A. et al., Genomic and expression profiling identifies the B-cell associated tyrosine kinase Syk as a possible therapeutic target in mantle cell lymphoma, Br. J. Haematol., 132(3): 303-16 (2006).
Roberts, P.J., and Der, C.J., Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer, Oncogene, 26(22): 3291-3310 (2007).
Ruiz-Ballesteros, E. et al., Splenic marginal zone lymphoma: proposal of new diagnostic and prognostic markers identified after tissue and cDNA microarray analysis. Blood, 106(5): 1831-8 (2005).
Sakamoto, T. et al., Blockade of the ERK pathway enhances the therapeutic efficacy of the histone deacetylase inhibitor MS-275 in human tumor xenograft models, Biochem. Biophys. Res. Commun., 433(4):456-62 (2013).
Schlessingerman, Mass of an Adult Male, The Physics Factbook (2003), retreived Jul. 22, 2014 from web: http://hypertexbook.com/facts/2003/AlexSchleesingerman.shtml.
Seidel, H. et al., Pharmaceutical intervention in the JAK/STAT signaling pathway, Oncogene 19: 2645-2656 (2000).
Seipelt, I et al., Dual Inhibition of P13K and Erk1/2 shows synergy and efficacy in human tumor cells, either by using drug combinations or novel dual P13K/Erk inhibitors, Aeterna Zentaris GmbH, AACR Poster, Abstract #871 (2012).
Sequist, L., Second—Generation Epidermal Growth Factor Receptor Tyrosine Kinase Inhibitors in Non-Small Cell Lung Cancer, The Oncologist 12(3):325-330 (2007).
Simone, J.V., Oncology: Introduction, Cecil Textbook of Medicine, 20th Edition, 1: 1004-1010 (1995).
Singh, J. et al, Structure-based design of a potent, selective, and irreversible inhibitor of the catalytic domain of the erbB receptor subfamily of protein tyrosine kinases, J. Med. Chem. 40:1130-1135 (1997).
Soria, J-C. et al., “Abstract # 1354: First-In-Human Evaluation of CO-1686, an Irreversible, Highly Selective Tyrosine Kinase Inhibitor of Mutations of EGFR (Activating and T790M),” 15th World Conference on Lung Cancer, Oct. 27, 2013.
Steelman, L.S. et al., Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to leukemia, Leukemia, 22(4): 686-707 (2008).
Steelman, L.S. et al., Roles of the Ras/Raf/MEK/ERK pathway in leukemia therapy, Leukemia, 25(7): 1080-1097 (2011).
Steinhardt, J.J. and Gartenhaus, R.B., Promising Personalized Therapeutic Options for Diffuse Large B-cell Lymphoma Subtypes with Oncogene Addictions, Clin. Cancer Res., 18(17): 4538-48 (2012).
Stonecypher, M. et al., Activation of the neuregulin-1/ErbB signaling pathway promotes the proliferation of neoplastic Schwann cells in human malignant peripheral nerve sheath tumors, Oncogene 24:5589-5605 (2005).
Sudbeck, E. et al., Structure-based Design of Specific Inhibitors of Janus Kinase 3 as Apoptosis-inducing Antileukemic Agents, Clin. Cancer Res. 5: 1569-1582 (1999).
Tohnya, T.M. et al., A phase I study of oral CC-5013 (lenalidomide, Revlimid), a thalidomide derivative, in patients with refractory metastatic cancer, Clin Prostate Cancer, 2(4): 241-3 (2004).
Trieu, V. et al., A specific inhibitor of janus kinase-3 increases survival in a transgenic mouse model of amyotrophic lateral sclerosis, Biochem. Biophys. Res. Commun. 267 :22-25 (2000).
Walter, A. O. et al., “Discovery of a mutant-selective covalent inhibitor of EGFR that overcomes T790M-mediated resistance in NSCLC,” Cancer Discov. Dec. 2013; 3(12): 1404-1415.
Walter, A.O. et al., Discovery of a Mutant-Selective Covalent Inhibitor of EGFR that Overcomes T790M Mediated Resistance in NSCLC, Cancer Discovery, 3(12): 1405-1415 (2013).
Westlin, W. et al., Translational medicine enables rapid early clinical development of AVL-292, a highly selective, orally available inhibitor of Bruton's tyrosine kinase, in a phase 1b clinical trial, Cancer Res., 72 (2012).
Wiernik, P.H. et al., Lenalidomide monotherapy in relapsed or refractory aggressive non-Hodgkin's lymphoma, J. Clin. Oncol., 26(30): 4952-7 (2008).
Winer, E.S. et al., A novel Bruton's tyrosine kinase inhibitor for the treatment of lymphoid malignancies, Expert Opin. Investig. Drugs, 21(3): 355-61 (2012).
Witzig, T.E. et al., Lenalidomide oral monotherapy produces durable responses in relapsed or refractory indolent non-Hodgkin's Lymphoma, J. Clin. Oncol., 27(32): 5404-5409 (2009).
Wong, K-K., Recent developments in anti-cancer agents targeting the Ras/Raf/ MEK/ERK pathway, Recent Patents on Anti-Cancer Drug Discovery, 4(1): 28-35 (2009).
Wong, K. et al, A phase I study with neratinib (HKI-272), an irreversible pan Erb B receptor tyrosine kinase inhibitor, in patients with solid tumors, Clin. Cancer Res. 15(7):2552-2558 (2009).
Written Opinion for PCT/US09/48784, 9 pages (dated Nov. 16, 2009).
Written Opinion for PCT/US10/31714, 7 pages (dated Aug. 13, 2010).
Written Opinion for PCT/US10/62432, 14 pages (dated May 26, 2011).
Written Opinion for PCT/US11/46926, 9 pages (dated Dec. 22, 2011).
Written Opinion for PCT/US11/58610, 8 pages (dated Mar. 27, 2012).
Written Opinion for PCT/US11/58616, 9 pages (dated Mar. 27, 2012).
Written Opinion for PCT/US11/59726, 7 pages (dated Mar. 20, 2012).
Written Opinion for PCT/US13/70772, 10 pages (dated Mar. 25, 2014).
Written Opinion for PCT/US13/70776, 11 pages (dated Mar. 25, 2014).
Written Opinion for PCT/US2013/070766, 11 pages (dated Mar. 25, 2014).
Written Opinion for PCT/US2014/015256, 6 pages (dated Aug. 5, 2014).
Written Opinion for PCT/US2015/044783, 7 pages (dated Oct. 30, 2015).
Written Opinion of PCT/US13/30982, 12 pages (dated May 30, 2013).
Written Opinion of PCT/US13/30996, 12 pages (dated May 30, 2013).
Yang, Y. et al., Exploiting Synthetic Lethality for the Therapy of ABC Diffuse Large B Cell Lymphoma, Cancer Cell, 21: 723-737 (2012).
Zhang, J. et al., Targeting Cancer with Small Molecule Kinase Inhibitors, Nature Rev. Cancer 9:28-39 (2009).
Zhang, Y. et al., Antitumor Activity of Epidermal Growth Factor Receptor-Related Protein Is Mediated by Inactivation of ErbB Receptors and Nuclear Factor-kB in Pancreatic Cancer, Cancer Res 66:1025-1032 (2006).
Zhou, W. et al. Novel mutant-selective EGFR kinase inhibitors against EGFR T790M, Nature, 462(7276): 1070-1074 (2009).
Zhou, W. et al., Novel mutant-selective EGFR kinase inhibitors against EGFR T790M, Nature, 462(7276): 1070-1074 (2009).
Zhu, D. et al., Immunomodulatory drugs Revlimid (lenalidomide) and CC-4047 induce apoptosis of both hematological and solid tumor cells through NK cell activation. Cancer Immunol. Immunother., 57(12): 1849-59 (2008).
Australian Examination Report for 2011323626, dated Sep. 1, 2014, 3 pages.
Extended European Search Report for EP11816874.9, 5 pages (dated Dec. 12, 2013).
Extended European Search Report for EP11838624.2, 5 pages (dated Jun. 6, 2014).
Extended European Search Report for EP11838628.3, 7 pages (dated Jun. 20, 2014).
Extended European Search Report for EP11839800.7, 8 pages (dated Jun. 24, 2014).
Supplementary European Search Report for EP10844293.0, 8 pages (dated Jun. 27, 2013).
Related Publications (1)
Number Date Country
20170226083 A1 Aug 2017 US
Provisional Applications (1)
Number Date Country
62037066 Aug 2014 US