CN117794935A - Salts and solid forms of kinase inhibitors - Google Patents

Salts and solid forms of kinase inhibitors Download PDF

Info

Publication number
CN117794935A
CN117794935A CN202280033117.2A CN202280033117A CN117794935A CN 117794935 A CN117794935 A CN 117794935A CN 202280033117 A CN202280033117 A CN 202280033117A CN 117794935 A CN117794935 A CN 117794935A
Authority
CN
China
Prior art keywords
salt
compound
crystalline
ray powder
powder diffraction
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202280033117.2A
Other languages
Chinese (zh)
Inventor
凯特林·金克马
布莱恩·海因里奇
李辉
克里斯多夫·李
克莱尔·奥布里·梅登多普
尼米塔·戴夫
图安·董·希
劳伦·麦凯克恩
埃里卡·巴特勒
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cable Chart Pharmaceutical Co
Original Assignee
Cable Chart Pharmaceutical Co
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cable Chart Pharmaceutical Co filed Critical Cable Chart Pharmaceutical Co
Priority claimed from PCT/US2022/019776 external-priority patent/WO2022192558A1/en
Publication of CN117794935A publication Critical patent/CN117794935A/en
Pending legal-status Critical Current

Links

Abstract

Various salt forms and free base solid forms of compound (I) are disclosed, represented by the following formula.

Description

Salts and solid forms of kinase inhibitors
Cross Reference to Related Applications
The present application claims priority from U.S. provisional application No. 63/159,107 filed on day 3 and day 10 of 2021 and U.S. provisional application No. 63/208,641 filed on day 6 and day 9 of 2021. The entire contents of the above-mentioned application are incorporated herein by reference.
Background
KIT enzyme (also known as CD 117) is a receptor tyrosine kinase expressed on a variety of cell types. KIT receptor proteins belong to the class III Receptor Tyrosine Kinase (RTK) family, which also includes the structurally related proteins pdgfrα (platelet derived growth factor receptor a), pdgfrβ, FLT3 (FMS-like tyrosine kinase 3) and CSF1R (colony stimulating factor 1 receptor). KIT molecules contain long extracellular domains, transmembrane segments, and intracellular portions. The ligand for KIT is Stem Cell Factor (SCF). Typically, stem Cell Factor (SCF) binds to and activates KIT by inducing dimerization, autophosphorylation, and downstream signaling initiation. However, in several tumor types, the activation of somatic mutations in KIT drives constitutive activity independent of the ligand.
KIT mutations usually occur in the DNA encoding the membrane proximal domain (exon 11). KIT mutations also occur at lower frequencies in exons 7, 8, 9, 13, 14, 17 and 18. Mutations make KIT function independent of SCF activation, resulting in high cell division rates and possible genomic instability. Mutant KIT is associated with the pathogenesis of several disorders and conditions such as mastocytosis, gastrointestinal stromal tumor (GIST), acute Myelogenous Leukemia (AML), melanoma, and seminoma.
The structurally related Platelet Derived Growth Factor Receptor (PDGFR) is a cell surface tyrosine kinase receptor of a member of the Platelet Derived Growth Factor (PDGF) family. PDGF subunits α and β regulate cell proliferation, cell differentiation, cell growth and cell development. Changes (e.g., mutations) in PDGF subunits α and PDGF subunits β are associated with a number of diseases, including some cancers. For example, the exon 18 pdgfrad 842V mutation has been found in a unique subset of GIST (usually from the stomach). The D842V mutation is also associated with tyrosine kinase inhibitor resistance. In addition, other exon 18 mutations (such as pdgfra D842I and pdgfra D842Y) are associated with ligand independent constitutive activation of pdgfra. In GIST, functionally acquired mutations that confer pdgfrα signaling independent of constitutive activation of the ligand (such as, for example, pdgfrαd842I, D842V and D842Y) have been identified as drivers of disease.
U.S. patent No. 10,829,493, the entire teachings of which are incorporated herein by reference, discloses potent, highly selective inhibitors of KIT, including exon 17 mutants and/or PDGFR alpha exon 18 mutant proteins. The structure of one of the inhibitors disclosed in U.S. patent No. 10,829,493 (referred to herein as "compound (I)") is shown below:
compound (I) is a potent and selective small molecule inhibitor of KIT exon 17 mutant enzyme KIT D816V. Its efficacy was demonstrated in an in vitro biochemical (dissociation constant, kd=0.24 nM) and cellular (half maximal inhibitory concentration, [ IC50] =4.3 nM) environment. Compound (I) is highly selective for KIT D816V compared to other kinases, transmembrane or soluble receptors, ion channels, transporters and other enzymes. Compound (I) has limited brain penetration potential.
Successful development of pharmaceutically active agents such as compound (I) generally requires the identification of solid forms having the following properties: can be isolated and purified immediately after synthesis, is suitable for large-scale production, can be stored for long periods of time and has little absorption of water, decomposition or conversion to other solid forms, is suitable for formulation, and can be readily absorbed (e.g., soluble in water and gastric juice) after administration to a subject.
Disclosure of Invention
It has now been found that the free base of compound (I) has a plurality of crystalline forms which can be interconverted with one another due to the presence of varying amounts of water in the crystal lattice. Thus, it was found that the preparation of the stable crystalline form of the free base of compound (I) is difficult to reproduce on a production scale.
It has also been found that 1:1 phosphate, 2:1 besylate, 1:1 benzoate and 1:1 sulfate can crystallize under well-defined conditions to provide a non-hygroscopic crystalline form. These four salts exhibit good thermal behavior and high melting point onset temperatures and are suitable for large scale synthesis. Minimal mass loss was observed during thermogravimetric analysis. The salts disclosed herein also exhibit similar solubility in simulated fluids and water at 37 ℃.
In addition, the 1:1 phosphate maintains a purity of greater than 98%. Among the crystalline forms of the 1:1 phosphate, anhydrous crystalline form a of the phosphate was found to be the most stable form. Form a is highly crystalline with a melting onset temperature of 199.7 ℃. Form a shows low residual solvent (0.45 wt%) and good solubility in simulated fluids and water. Form a also exhibits low solubility in selected organic solvents and solvent mixtures, but is soluble in high boiling solvents such as dimethylacetamide (DMAc), N-methylpyrrolidone (NMP) and Dimethylsulfoxide (DMSO), and in aqueous/organic mixtures, particularly with Tetrahydrofuran (THF). Form a was obtained from different crystallization experiments and in slurries of alcohol, acetate, acetone, methyl Ethyl Ketone (MEK), trifluoroethanol (TFE), dioxane, chlorobenzene, chloroform, anisole and in many aqueous/organic mixtures. Crystallization using 2-MeTHF is advantageous because it provides crystalline compound (I) phosphate form a, the residual solvent of which is robustly controlled.
The symbol "1:1" is the molar ratio between compound (I) and an acid (e.g. sulfuric acid, phosphoric acid or benzoic acid); and the symbol "2:1" is the molar ratio between compound (I) and the acid (e.g. benzenesulfonic acid).
In one aspect, the present disclosure provides a phosphate salt of compound (I), wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
In another aspect, the present invention provides a benzenesulfonate salt of compound (I), wherein the molar ratio between compound (I) and benzenesulfonic acid is 2:1.
In yet another aspect, the present invention provides a sulfate salt of compound (I), wherein the molar ratio between compound (I) and sulfuric acid is 1:1.
In yet another aspect, the present invention provides a benzoate salt of compound (I), wherein the molar ratio between compound (I) and benzoic acid is 1:1.
In another aspect, the present disclosure provides a pharmaceutical composition comprising a salt or free base (including amorphous and crystalline forms) of compound (I) disclosed herein and a pharmaceutically acceptable carrier or diluent.
The present disclosure provides a method of treating disorders and conditions associated with oncogenic KIT and PDGFRA alterations comprising administering to a patient in need thereof a salt or free base (including amorphous and crystalline forms) of compound (I) disclosed herein or a corresponding pharmaceutical composition thereof.
The present disclosure provides a method of treating a disease or condition in a patient in need thereof, comprising administering to the patient in need thereof a salt or free base (including amorphous and crystalline forms) of compound (I) disclosed herein or a corresponding pharmaceutical composition thereof, wherein the disease or condition is selected from the group consisting of systemic mastocytosis, gastrointestinal stromal tumor, acute myelogenous leukemia, melanoma, seminoma, intracranial germ cell tumor, mediastinal B cell lymphoma, ewing's sarcoma, diffuse large B cell lymphoma, asexual cell tumor, myelodysplastic syndrome, nasal NK/T cell lymphoma, chronic myelomonocytic leukemia, and brain cancer. In one embodiment, the disease or condition is selected from the group consisting of systemic mastocytosis, gastrointestinal stromal tumor, acute myelogenous leukemia, melanoma, seminoma, mediastinal B-cell lymphoma, ewing's sarcoma, diffuse large B-cell lymphoma, anaplastic cytoma, myelodysplastic syndrome, nasal NK/T-cell lymphoma, and chronic myelomonocytic leukemia. In one embodiment, the disease or condition is systemic mastocytosis. In one embodiment, the systemic mastocytosis is selected from the group consisting of indolent systemic mastocytosis and smoky systemic mastocytosis.
The present disclosure also provides the use of a salt or free base of compound (I) of the present disclosure or a pharmaceutical composition comprising said salt or free base for the treatment of any of the diseases listed in the preceding paragraph. In one embodiment, provided are salts or free bases of the present disclosure or pharmaceutical compositions comprising the salts or free bases for use in any of the methods of the present disclosure described herein. In another embodiment, there is provided the use of a salt or free base of the present disclosure or a pharmaceutical composition comprising said salt or free base for the manufacture of a medicament for any of the methods of the present disclosure as described.
Drawings
FIG. 1A shows an X-ray powder diffraction (XRPD) pattern of compound (I) phosphate form A, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
Figure 1B shows thermograms of thermogravimetric analysis (TGA) and differential scanning calorimetric analysis (DSC) (5 mW) of compound (I) phosphate form a, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
FIG. 1C shows a differential scanning calorimetric analysis (DSC) (10 mW) thermogram of compound (I) phosphate form A, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
Fig. 2A and 2B (y-axis magnified) show X-ray powder diffraction (XRPD) patterns of compound (I) phosphate form G, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
Figure 2C shows thermograms of thermogravimetric analysis (TGA) and differential scanning calorimeter analysis (DSC) of compound (I) phosphate form G, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
Fig. 2D shows a differential scanning calorimetric analysis (DSC) thermogram of compound (I) phosphate form G, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
FIG. 3 shows an X-ray powder diffraction (XRPD) pattern of compound (I) phosphate form O, wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
FIG. 4A shows an X-ray powder diffraction (XRPD) pattern of the benzenesulfonate salt form 1-A of compound (I), wherein the molar ratio between compound (I) and benzenesulfonic acid is 1:2.
FIG. 4B shows thermograms of thermogravimetric analysis (TGA) and Differential Scanning Calorimetry (DSC) of the benzenesulfonate form 1-A of compound (I) with a molar ratio between compound (I) and benzenesulfonic acid of 1:2.
FIG. 4C shows a Differential Scanning Calorimetric (DSC) thermogram of the benzenesulfonate form 1-A of compound (I), wherein the molar ratio between compound (I) and benzenesulfonic acid is 1:2.
FIG. 5 shows an X-ray powder diffraction (XRPD) pattern of the benzenesulfonate salt form 1-B of compound (I), wherein the molar ratio between compound (I) and benzenesulfonic acid is 1:2.
FIG. 6A shows an X-ray powder diffraction (XRPD) pattern of compound (I) benzoate form 2-A, with a molar ratio between compound (I) and benzoic acid of 1:1.
FIG. 6B shows thermograms of thermogravimetric analysis (TGA) and Differential Scanning Calorimetry (DSC) of the benzoic acid form 2-A of compound (I), wherein the molar ratio between compound (I) and benzoic acid is 1:1.
FIG. 6C shows a Differential Scanning Calorimetric (DSC) thermogram of the benzoic acid form 2-A of compound (I), wherein the molar ratio between compound (I) and benzoic acid is 1:1.
FIG. 7 shows an X-ray powder diffraction (XRPD) pattern of compound (I) benzoate form 2-B, with a molar ratio between compound (I) and benzoic acid of 1:1.
FIG. 8A shows an X-ray powder diffraction (XRPD) pattern of compound (I) sulfate form 9-A, with a molar ratio of 1:1 between compound (I) and sulfuric acid.
FIG. 8B shows a Differential Scanning Calorimetric (DSC) thermogram of the sulfate form 9-A of compound (I), wherein the molar ratio between compound (I) and sulfuric acid is 1:1.
FIG. 9A shows an X-ray powder diffraction (XRPD) pattern of compound (I) free base form FB-A-0.
Figure 9B shows thermogravimetric analysis (TGA) and differential scanning calorimeter analysis (DSC) thermograms of compound (I) crystalline free base.
Fig. 9C shows a differential scanning calorimetric analysis (DSC) thermogram of compound (I) crystalline free base.
FIG. 10 shows an X-ray powder diffraction (XRPD) pattern of compound (I) free base form FB-A-1.
FIG. 11 shows an X-ray powder diffraction (XRPD) pattern of compound (I) free base form FB-A-2.
Fig. 12 shows an X-ray powder diffraction (XRPD) pattern of amorphous phosphate of compound (I).
Figure 13 shows the DSC thermogram of the lyophilized amorphous phosphate of compound (I).
Fig. 14 shows X-ray powder diffraction (XRPD) patterns of amorphous compound (I) free base prepared by lyophilization of crystalline solids in various solvent systems. (1) amorphous form prepared from ACN: water (7:3 volume); (2) amorphous form prepared from t-BuOH (L.C); (3) amorphous form prepared from t-BuOH in water (9:1 volumes).
FIG. 15 shows an X-ray powder diffraction (XRPD) pattern of the free base of compound (I) prepared in example 10. Instrument: bruker D8 advanced X-ray diffractometer, X-ray source:working electric quantity: 40kV,40mA, initial angle: 4 °; stop angle: 40 °, increment: 0.05 degree/step, scan speed: 0.5 seconds/step.
Detailed Description
The present disclosure relates to: i) Pharmaceutically acceptable salts of compound (I); ii) new solid forms of the free base of compound (I) and new solid forms of the pharmaceutically acceptable salts of compound (I), including unsolvated forms, solvated forms, amorphous forms, and crystalline forms (hereinafter collectively referred to as "salts and solid forms of the present disclosure"); and iii) methods of use and preparation of the salts and solid forms of the present disclosure.
Phosphates of the Compound (I)
In one aspect, the present disclosure provides a phosphate salt of compound (I), wherein the molar ratio between compound (I) and phosphoric acid is 1:1.
In some embodiments, the phosphonate is crystalline. In some embodiments, the phosphate is in a single crystalline form.
In some embodiments, the phosphate salt is unsolvated. In other embodiments, the phosphate salt is solvated.
In some embodiments, the present disclosure provides crystalline form a of the phosphate salt of compound (I). XRPD patterns and peaks are shown in fig. 1A. Thermogravimetric analysis (TGA) and differential scanning calorimeter analysis (DSC) thermograms are shown in figure 1B.
Table 1-list of peaks for compound (I) phosphate form a.
Only peaks with a relative intensity of 3 or more are reported.
Table 2-list of concentrated peaks for compound (I) phosphate form a #1.
Table 3-list of concentrated peaks for compound (I) phosphate form a #2.
Table 4-list of concentrated peaks for compound (I) phosphate form a #3.
In some embodiments, the present disclosure provides crystalline form G of the phosphate salt of compound (I). XRPD patterns and peaks are shown in fig. 2A and 2B. Thermogravimetric analysis (TGA) and differential scanning calorimeter analysis (DSC) thermograms are shown in figures 2C and 2D.
Table 5-list of peaks for compound (I) phosphate form G.
Only peaks with a relative intensity of 0.5 or more are reported.
Table 6-list of concentrated peaks for compound (I) phosphate form G #1.
Only peaks with a relative intensity of 0.5 or more are reported.
Table 7-list of concentrated peaks for compound (I) phosphate form G #2.
Table 8-concentrated peak list #3 for Compound (I) phosphate form G.
In some embodiments, the present disclosure provides crystalline form O of the phosphate salt of compound (I). XRPD patterns and peaks are shown in fig. 3.
Table 9-list of peaks for compound (I) phosphate form O.
Only peaks with a relative intensity of 5 or more are reported.
Table 10-concentrated peaks list #1 for compound (I) phosphate form O.
Table 11-concentrated peak list #2 for compound (I) phosphate form O.
Table 12-list of concentrated peaks for compound (I) phosphate form O #3.
Benzenesulfonate of Compound (I)
In one aspect, the present invention provides a benzenesulfonate salt of compound (I), wherein the molar ratio between compound (I) and benzenesulfonic acid is 1:2.
In some embodiments, the benzenesulfonate salt is crystalline. In some embodiments, the benzenesulfonate salt is in a single crystalline form.
In some embodiments, the benzenesulfonate salt is unsolvated. In other embodiments, the benzenesulfonate salt is solvated.
In some embodiments, the present disclosure provides crystalline form 1-a of the benzenesulfonate salt of compound (I). XRPD patterns and peaks are shown in fig. 4A. Thermogravimetric analysis (TGA) and differential scanning calorimeter analysis (DSC) thermograms are shown in figures 4B and 4C.
Table 13-Peak List of the benzenesulfonate form 1-A of Compound (I).
/>
Only peaks with a relative intensity of 5 or more are reported.
Table 14-concentrated peaks list #1 for the benzenesulfonate form 1-A of compound (I).
Table 15-concentrated peaks list #2 for the benzenesulfonate form 1-A of compound (I).
/>
Table 16-concentrated peaks list #3 for the benzenesulfonate form 1-A of compound (I).
In some embodiments, the present disclosure provides crystalline forms 1-B of the benzenesulfonate salt of compound (I). XRPD patterns and peaks are shown in fig. 5.
Table 17-Peak List of the benzenesulfonate form 1-B of Compound (I).
Only peaks with a relative intensity of 5 or more are reported.
Table 18-concentrated peaks list #1 for the benzenesulfonate form 1-B of compound (I).
Table 19-concentrated peaks list #2 for the benzenesulfonate form 1-B of compound (I).
Table 20-concentrated peaks list #3 for the benzenesulfonate form 1-B of compound (I).
Benzoate of Compound (I)
In one aspect, the present disclosure provides a benzoate salt of compound (I), wherein the molar ratio between compound (I) and benzoic acid is 1:1.
In some embodiments, the benzoate salt is crystalline. In some embodiments, the benzoate salt is in a single crystalline form.
In some embodiments, the benzoate salt is unsolvated. In other embodiments, the benzoate salt is solvated.
In some embodiments, the present disclosure provides crystalline form 2-a of the benzoate salt of compound (I). XRPD patterns and peaks are shown in fig. 6A. Thermogravimetric analysis (TGA) and differential scanning calorimeter analysis (DSC) thermograms are shown in fig. 6B and 6C.
Table 21-Peak List of Compound (I) benzoate form 2-A.
Only peaks with a relative intensity of 5 or more are reported.
Table 22-concentrated peak list #1 for benzoate form 2-A of compound (I).
/>
Table 23-concentrated peak list #2 for benzoate form 2-A of compound (I).
Table 24-concentrated peak list #3 for benzoate form 2-A of compound (I).
In some embodiments, the present disclosure provides crystalline form 2-B of the benzoate salt of compound (I). XRPD patterns and peaks are shown in fig. 7.
Table 25-Peak List of the benzoate form 2-B of Compound (I).
/>
Only peaks with a relative intensity of 5 or more are reported.
Table 26-concentrated peak list #1 for benzoate form 2-B of compound (I).
Table 27-concentrated peak list #2 for benzoate form 2-B of compound (I).
Table 28-concentrated peak list #3 for benzoate form 2-B of compound (I).
Sulfate of Compound (I)
In one aspect, the present disclosure provides a sulfate salt of compound (I), wherein the molar ratio between compound (I) and sulfuric acid is 1:1.
In some embodiments, the sulfate salt is crystalline. In some embodiments, the sulfate salt is in a single crystalline form.
In some embodiments, the sulfate salt is unsolvated. In other embodiments, the sulfate salt is solvated.
In some embodiments, the present disclosure provides crystalline form 9-a of the sulfate salt of compound (I). XRPD patterns and peaks are shown in fig. 8A. A differential scanning calorimetric analysis (DSC) thermogram is shown in figure 8B.
Table 29-Peak List of Compound (I) sulfate form 9-A.
Only peaks with a relative intensity of 5 or more are reported.
Table 30-concentrated peaks list #1 for Compound (I) sulfate form 9-A.
Table 31-concentrated peaks list #2 for Compound (I) sulfate form 9-A.
Table 32-concentrated peaks list #3 for Compound (I) sulfate form 9-A.
Free base of Compound (I)
In one aspect, the present disclosure provides the free base of compound (I).
In some embodiments, the free base of compound (I) is in an amorphous form.
In some embodiments, the free base of compound (I) is crystalline. In some embodiments, the free base of compound (I) is in a single crystalline form.
In some embodiments, the free base of compound (I) is non-solvated. In other embodiments, the free base of compound (I) is solvated.
In some embodiments, the present disclosure provides ase:Sub>A crystalline family of free base form FB-A of compound (I) that includes form FB-A-0 (monohydrate), form FB-A-1 (metastable hydrate), and form FB-A-2 (dehydrate).
As described herein, a family of crystals is a group of crystalline forms including the same crystalline form, which may be described as "interconversions" due to the presence of varying amounts of water in the crystal lattice. Different amounts of water resulted in slight peak shifts in the XRPD.
The family of crystals of form FB-A-0 was found to be channel hydrates which can hold varying amounts of water in the crystal lattice depending on the ambient humidity. XRPD patterns and peaks are shown in fig. 9A.
Table 33-Peak List of free base form FB-A-0 of Compound (I).
Only peaks with a relative intensity of 5 or more are reported.
Table 34-concentrated peak list #1 of free base form FB-A-0 of Compound (I).
Table 35-concentrated peak list #2 of free base form FB-A-0 of Compound (I).
Table 36-concentrated peak list #3 of free base form FB-A-0 of Compound (I).
In some embodiments, the present disclosure provides ase:Sub>A crystalline form of FB-A-1 of the free base of compound (I). XRPD patterns and peaks are shown in fig. 10.
Table 37-Peak List of free base form FB-A-1 of Compound (I).
Only peaks with a relative intensity of 3 or more are reported.
TABLE 38 concentrated peak list #1 for compound (I) free base form FB-A-1.
Table 39-concentrated peak list #2 of free base form FB-A-1 of Compound (I).
TABLE 40 concentrated peak list #3 for compound (I) free base form FB-A-1.
In some embodiments, the present disclosure provides crystalline form FB-A-2 of the free base of compound (I). XRPD patterns and peaks are shown in fig. 11.
Table 41-Peak List of free base form FB-A-2 of Compound (I).
Only peaks with a relative intensity of 5 or more are reported.
Table 42-concentrated peak list #1 of free base form FB-A-2 of Compound (I).
/>
Table 43-concentrated peak list #2 of free base form FB-A-2 of Compound (I).
Table 44-concentrated peak list #3 of free base form FB-A-2 of Compound (I).
In some embodiments, the present disclosure provides crystalline forms of the free base of compound (I) represented by the formula:
wherein the crystalline form comprises ase:Sub>A family of form A selected from the group consisting of crystalline forms of form FB-A-0, form FB-A-1 and form FB-A-2, characterized by at least one of the following:
(a) An X-ray powder diffraction pattern (XRPD) substantially the same as shown in figures 9A, 10 or 11;
(b) An X-ray powder diffraction pattern (XRPD) comprising at least three peaks selected from the group consisting of 10.6 °, 12.2 °, 14.2 °, 14.8 °, 17.1 °, 18.3 °, 23.1 ° and 24.1;
(c) Thermogravimetric analysis (TGA) substantially similar to fig. 9B.
(d) A DSC thermogram substantially similar to the one set forth in figure 9C.
(e) DSC thermograms with endotherm onset temperatures of 61.6+ -2deg.C and 171.4+ -2deg.C; or (b)
(f) A combination thereof.
Pharmaceutical composition
Some embodiments of the present disclosure relate to a pharmaceutical composition comprising a salt or solid form of the present disclosure and a pharmaceutically acceptable excipient. Some embodiments of the present disclosure relate to a pharmaceutical composition comprising: a pharmaceutically acceptable excipient; and a phosphate of the compound (I), wherein the molar ratio between the compound (I) and phosphoric acid is 1:1. In some embodiments, the phosphonate is crystalline. In some embodiments, the phosphate is in a single crystalline form. In some embodiments, the phosphate salt is unsolvated. In other embodiments, the phosphate salt is solvated. In some embodiments, the phosphate salt of compound (I) is crystalline form a. In some embodiments, the phosphate salt of compound (I) is crystalline form G. In some embodiments, the phosphate salt of compound (I) is crystalline form O.
Some embodiments of the present disclosure relate to a pharmaceutical composition comprising: a pharmaceutically acceptable excipient; and a benzenesulfonate salt of the compound (I), wherein the molar ratio between the compound (I) and benzenesulfonic acid is 1:2. In some embodiments, the benzenesulfonate salt is crystalline. In some embodiments, the benzenesulfonate salt is in a single crystalline form. In some embodiments, the benzenesulfonate salt is unsolvated. In other embodiments, the benzenesulfonate salt is solvated. In some embodiments, the benzenesulfonate salt of compound (I) is crystalline form 1-a. In some embodiments, the benzenesulfonate salt of compound (I) is crystalline form 1-B.
Some embodiments of the present disclosure relate to a pharmaceutical composition comprising: a pharmaceutically acceptable excipient; and a benzoate salt of the compound (I), wherein the molar ratio between the compound (I) and benzoic acid is 1:1. In some embodiments, the benzoate salt is crystalline. In some embodiments, the benzoate salt is in a single crystalline form. In some embodiments, the benzoate salt is unsolvated. In other embodiments, the benzoate salt is solvated. In some embodiments, the benzoate salt of compound (I) is crystalline form 2-A. In some embodiments, the benzoate salt of compound (I) is crystalline form 2-B.
Some embodiments of the present disclosure relate to a pharmaceutical composition comprising: a pharmaceutically acceptable excipient; and a sulfate of the compound (I), wherein the molar ratio between the compound (I) and sulfuric acid is 1:1. In some embodiments, the sulfate salt is crystalline. In some embodiments, the sulfate salt is in a single crystalline form. In some embodiments, the sulfate salt is unsolvated. In other embodiments, the sulfate salt is solvated. In some embodiments, the sulfate salt of compound (I) is crystalline form 9-a.
Some embodiments of the present disclosure relate to a pharmaceutical composition comprising a pharmaceutically acceptable excipient; and compound (I) free base. In some embodiments, the free base is in an amorphous form. In some embodiments, the free base is crystalline. In some embodiments, the free base is in a single crystalline form. In some embodiments, the free base is unsolvated. In other embodiments, the free base is solvated. In some embodiments, the free base of compound (I) is the crystalline form FB-A-0. In some embodiments, the free base of compound (I) is the crystalline form FB-A-1. In some embodiments, the free base of compound (I) is the crystalline form FB-A-2.
The salts or solid forms of the present disclosure may be formulated for administration in any convenient manner for use in human or veterinary medicine. In some embodiments, the compound or salt included in the pharmaceutical composition may be active itself, or may be a prodrug, for example, capable of being converted to the active compound in a physiological environment.
The phrase "pharmaceutically acceptable" is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
Examples of pharmaceutically acceptable carriers/excipients include: (1) sugars such as, for example, lactose, glucose, and sucrose; (2) Starches such as, for example, corn starch and potato starch; (3) Cellulose and its derivatives such as, for example, sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) gum tragacanth powder; (5) malt; (6) gelatin; (7) talc; (8) excipients such as, for example, cocoa butter and suppository waxes; (9) Oils such as, for example, peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) ethylene glycol, such as, for example, propylene glycol; (11) Polyols such as, for example, glycerol, sorbitol, mannitol and polyethylene glycol; (12) esters such as, for example, ethyl oleate and ethyl laurate; (13) agar; (14) Buffering agents such as, for example, magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) ringer's solution; (19) ethanol; (20) phosphate buffer solution; (21) Cyclodextrins, such as, for example And (22) other non-toxic compatible substances employed in pharmaceutical formulations.
Examples of pharmaceutically acceptable antioxidants include: (1) Water-soluble antioxidants such as, for example, ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite, and the like; (2) Oil-soluble antioxidants such as, for example, ascorbyl palmitate, butylated Hydroxyanisole (BHA), butylated Hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelators such as, for example, citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.
Solid dosage forms (e.g., capsules, tablets, pills, dragees, powders, granules, and the like) may contain one or two pharmaceutically acceptable carriers (such as, for example, sodium citrate or dicalcium phosphate) and/or any of the following: (1) Fillers or extenders such as, for example, starch, lactose, sucrose, glucose, mannitol and/or silicic acid; (2) Binders such as, for example, carboxymethyl cellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and/or acacia; (3) humectants such as, for example, glycerol; (4) Disintegrants such as, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarders such as, for example, paraffin wax; (6) absorption accelerators such as, for example, quaternary ammonium compounds; (7) Wetting agents such as, for example, cetyl alcohol and glycerol monostearate; (8) absorbents such as, for example, kaolin and bentonite; (9) Lubricants such as, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, and mixtures thereof; and (10) a colorant.
Liquid dosage forms may include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage form may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as, for example, ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1, 3-butylene glycol, oils (such as, for example, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, tragacanth, and mixtures thereof.
In addition to the active compounds, the ointments, pastes, creams and gels may contain excipients such as, for example, animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc, zinc oxide or mixtures thereof.
Powders and sprays can contain, in addition to the active compound, excipients such as, for example, lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder or mixtures of these substances. The spray may additionally contain conventional propellants such as, for example, chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons such as, for example, butane or propane.
The salts or solid forms of the present disclosure may be given as such or as a pharmaceutical composition containing, for example, from 0.1 to 99.5% (such as from 0.5 to 90%) of the active ingredient in combination with a pharmaceutically acceptable carrier.
The formulations may be administered topically, orally, transdermally, rectally, vaginally, parenterally, intranasally, intrapulmonary, intraocular, intravenous, intramuscular, intraarterial, intrathecally, intracapsular, intradermal, intraperitoneal, subcutaneous, subcuticular, or by inhalation.
Therapeutic method
Some embodiments of the present disclosure relate to methods of treating patients in need of KIT or pdgfrα inhibitors by administering a therapeutically effective amount of a salt or solid form of the present disclosure.
Salts and solid forms of the present disclosure are selective KIT inhibitors. In some embodiments, the salts and solid forms of the present disclosure are selective D816V KIT inhibitors. In some embodiments, the salts and solid forms of the present disclosure are selective pdgfrα inhibitors. In some embodiments, the salts and solid forms of the present disclosure are selective PDGFR alpha exon 18 inhibitors. In some embodiments, the salts and solid forms of the present disclosure are selective PDGFR αd842V inhibitors. As used herein, "selective KIT inhibitor" or "selective PDGFR alpha inhibitor" refers to a salt or solid form of the disclosure that selectively inhibits a KIT protein kinase or PDGFR alpha protein kinase relative to another protein kinase, and exhibits at least 2-fold selectivity for the KIT protein kinase or PDGFR alpha protein kinase over the other kinase. For example, a selective KIT inhibitor or selective PDGFRA inhibitor exhibits at least 9-fold, 10-fold selectivity for a KIT protein kinase or PDGFRA protein kinase over another kinase; at least 15 times; at least 20 times; at least 30 times; at least 40 times; at least 50 times; at least 60 times; at least 70 times; at least 80 times; at least 90 times; at least 100-fold, at least 125-fold, at least 150-fold, at least 175-fold, or at least 200-fold. In some embodiments, the selective KIT inhibitor or selective PDGFR alpha inhibitor exhibits a selectivity that is at least 150-fold over another kinase, e.g., VEGFR2 (vascular epidermal growth factor receptor 2), SRC (non-receptor protein tyrosine kinase), and FLT3 (Fms-like tyrosine kinase 3). In some embodiments, the selective KIT or selective pdgfrα inhibitor exhibits selectivity for pdgrfβ, CSF1R (colony stimulating factor receptor 1), and FLT 3.
In some embodiments, the selective KIT or selective pdgfrα inhibitor exhibits selectivity for LCK (lymphocyte-specific protein kinase), ABL (nucleoprotein tyrosine kinase), immortalized mitotic gene a (NIMA) associated kinase 5 (NEK 5) and ROCK1 (Rho-associated coil-coil-continuing protein kinase) -1. In some embodiments, the selectivity for KIT protein kinase or PDGFR alpha protein kinase relative to another kinase is measured in a cellular assay (e.g., a cellular assay). In some embodiments, the selectivity for KIT protein kinase or PDGFR alpha protein kinase over another kinase is measured in a biochemical assay (e.g., a biochemical assay).
The salts and solid forms of the present disclosure are selective for ion channels. In some embodiments, the selective KIT or selective PDGFR alpha inhibitor has limited potential to inhibit human voltage-gated sodium channels (hNav 1.2).
The salts and solid forms of the present disclosure are more selective for mutant KIT than for wild-type KIT. In some embodiments, the salts and solid forms of the present disclosure are selective for exon 17 mutant KIT over wild-type KIT.
The salts and solid forms of the present disclosure are useful for treating diseases or conditions in humans or non-humans that are associated with mutant KIT or mutant PDGFRA activity. In some embodiments, the salts and solid forms of the present disclosure are used as medicaments. In some embodiments, the salts and solid forms of the present disclosure are used in therapy. In some embodiments, the salts and solid forms of the present disclosure are used in the manufacture of medicaments. In some embodiments, the present disclosure provides methods for treating KIT-driven malignancies, including mastocytosis (SM), GIST (gastrointestinal stromal tumor), AML (acute myelogenous leukemia), melanoma, seminoma, intracranial germ cell tumor, and/or mediastinal B cell lymphoma. In addition, KIT mutations are associated with ewing's sarcoma, DLBCL (diffuse large B-cell lymphoma), asexual cell tumor, MDS (myelodysplastic syndrome), NKTCL (nasal NK/T cell lymphoma), CMML (chronic myelomonocytic leukemia) and brain cancer. In some embodiments, the present disclosure provides methods for treating ewing's sarcoma, DLBCL, asexual cytoma, MDS, NKTCL, CMML, and/or brain cancer. KIT mutations can also be found in thyroid, colorectal, endometrial, bladder, NSCLC and breast cancers (AACR Project GENIE). In some embodiments, the salts and solid forms of the present disclosure are useful for treating Mast Cell Activation Syndrome (MCAS). The salts and solid forms of the present disclosure are useful for treating systemic mastocytosis. The salts and solid forms of the present disclosure are useful for treating advanced systemic mastocytosis. Salts and solid forms of the present disclosure are useful for treating inert SM and smoky SM. Salts and solid forms of the present disclosure are useful for treating GIST.
Salts and solid forms of the present disclosure are useful for treating diseases or conditions associated with KIT mutations in exon 9, exon 11, exon 14, exon 17, and/or exon 18 of KIT gene sequences. Salts and solid forms of the present disclosure are useful for treating diseases or conditions associated with PDGFRA mutations in exon 12, exon 14, and/or exon 18 of the PDGFRA gene sequence. In some embodiments, provided herein are methods for treating a disease or condition associated with at least one KIT mutation in exon 9, exon 11, exon 14, exon 17, and/or exon 18 of a KIT gene sequence. In some embodiments, methods are provided for treating a disease or condition associated with at least one PDGFRA mutation in exon 12, exon 14, and/or exon 18 of a PDGFRA gene sequence.
The salts and solid forms of the present disclosure may be active against one or more KIT protein kinases having mutations in exon 17 of the KIT gene sequence (e.g., KIT protein mutation D816V, D816Y, D816F, D816K, D816A, D816G, D816E, D816I, D816F, D820A, D820E, D820G, D820Y, N822K, N822H, V560G, Y823D and a 829P) and much less active against wild-type KIT protein kinases. In some embodiments, provided herein are methods for treating a disease or condition associated with at least one KIT mutation, such as a KIT mutation selected from the group consisting of D816V, D816Y, D816F, D816K, D816H, D816A, D816G, D816E, D816I, D816F, D820A, D820E, D820G, D820Y, N822H, V560G, Y823D and a 829P. In some embodiments, provided herein are methods for treating a disease or condition associated with at least one KIT mutation, such as, for example, those selected from C809, C809G, D816H, D820A, D820G, N822H, N822K and Y823D.
Salts and solid forms of the present disclosure may be active against one or more KIT protein kinases having a mutation in exon 11 of the KIT gene sequence (e.g., KIT protein mutation del557-559insF, V559G/D). In some embodiments, provided herein are methods for treating a disease or condition associated with at least one KIT mutation, such as, for example, those selected from the group consisting of L576P, V559D, V560D, V560G, W557G, del 554-558EVQWK, del557-559insF, del EVQWK554-558, del EVQWKVVEEINGNNYVYI554-571, del KPMYEVQWK550-558, del KPMYEVQW550-557FL, del KV558-559N, del MYEVQW552-557, del PMYE551-554, del VV559-560, del wkv 557-561, del WK557-558, del wkv 557-560C, del wkv 557-F, delYEVQWK-558, and insert K558.
Salts and solid forms of the present disclosure may be active against one or more KIT protein kinases having mutations in exon 11/13 of the KIT gene sequence (e.g., KIT protein mutations V559D/V654A, V G/D816V and V560G/822K). In some embodiments, provided herein are methods for treating a disease or condition associated with one or more KIT mutations in exons 11/13.
Salts and solid forms of the present disclosure may be active against one or more KIT protein kinases having a mutation in exon 9 of the KIT gene sequence. In some embodiments, provided herein are methods for treating a disease or condition associated with at least one KIT mutation in exon 9.
In some embodiments, the salts and solid forms of the present disclosure are not active against KIT protein kinase having mutations V654A, N655T, T670I and/or N680.
Salts and solid forms of the present disclosure may be active against one or more pdgfrα protein kinases having mutations. In some embodiments, provided herein are methods for treating a disease or condition associated with at least one PDGFRA mutation in exon 12 of a PDGFRA gene sequence, such as, for example, PDGFRA protein mutation V561D, del RV560-561, del RVIES560-564, ins ER561-562, spdche 566-571R, SPDGHE566-571K, or Ins YDSRW582-586. In some embodiments, provided herein are methods for treating a disease or condition associated with at least one PDGFRA mutation in exon 14 of a PDGFRA gene, such as, for example, PDGFRA protein mutation N659K. In some embodiments, provided herein are methods for treating diseases or conditions associated with at least one PDGFRA mutation in exon 18 of a PDGFRA gene, such as, for example, PDGFRA protein mutation D842V, D842Y, D842-843 IM, D846Y, Y849C, del D842, del I843, del RD841-842, del DIM842-845, del DIMH842-845, del IMHD843-846, del MHDS844-847, RD841-842KI, DIMH842-845A, DIMH842-845V, DIMHD842-846E, DIMHD 842-S, DIMHD842-846G, IMHDS843-847T, IMHDS8843-847M, or HDSN 848P.
Salts and solid forms of the present disclosure may be active against one or more PDGFRA protein kinases having mutations in exon 18 of the PDGFRA gene sequence (e.g., the protein mutations PDGFRA D842V, PDGFR a D842I or PDGFRA D842Y). In some embodiments, provided herein are methods for treating a disease or condition associated with at least one PDGFRA mutation in exon 18, such as, for example, the protein mutation pdgfrad 842V.
Salts and solid forms of the present disclosure are useful for treating eosinophilic disorders. In some embodiments, the eosinophil disorder is mediated by mutant KIT or pdgfrα. In some embodiments, the eosinophil disorder is mediated by wild-type KIT or PDGFR alpha. In some embodiments, provided herein are methods for treating an eosinophil disorder comprising administering to a subject a therapeutically effective amount of a salt of the present disclosure and a solid form or a pharmaceutically acceptable salt thereof and/or a solvate of any of the foregoing. In one embodiment, the eosinophilic disorder is selected from the group consisting of eosinophilia syndrome (hypereosinophilic syndrome), eosinophilia (eosinophia), eosinophilic gastroenteritis (eosinophilic enterogastritis), eosinophilic leukemia (eosinophilic leukemia), eosinophilic granuloma (eosinophilic granuloma) and Mumura's disease.
In some embodiments, the eosinophilic disorder is selected from the group consisting of eosinophilia syndrome, eosinophilia, eosinophilic gastroenteritis, eosinophilic leukemia, eosinophilic granuloma and Mucun's disease. Other eosinophilic conditions include eosinophilic esophagitis, eosinophilic gastroenteritis, eosinophilic fasciitis and Churg-Strauss syndrome.
In one embodiment, the eosinophilic disorder is eosinophilia syndrome. In particular embodiments, the eosinophilia syndrome is idiopathic eosinophilia syndrome. In one embodiment, the eosinophilic disorder is eosinophilic leukemia. In a specific embodiment, the eosinophilic leukemia is chronic eosinophilic leukemia. In another embodiment, eosinophilic disorders are refractory to treatment with imatinib (imatinib), sunitinib (sunitinib), and/or regorafenib (regorafenib). In particular embodiments, eosinophilic conditions are refractory to treatment with imatinib.
The salts and solid forms of the present disclosure are useful for reducing the number of eosinophils in a subject in need thereof. In some embodiments, provided herein are methods for reducing the number of eosinophils in a subject in need thereof, comprising administering to the subject a therapeutically effective amount of a salt of the present disclosure and a solid form or a pharmaceutically acceptable salt thereof and/or a solvate of any of the foregoing.
In one embodiment, the disclosed methods reduce the number of eosinophils in the blood, bone marrow, gastrointestinal tract (e.g., esophagus, stomach, small intestine, and colon), or lung. In another embodiment, the methods disclosed herein reduce the number of blood eosinophils. In another embodiment, the methods disclosed herein reduce the number of lung eosinophils. In yet another embodiment, the methods disclosed herein reduce the number of eosinophil precursor cells.
In another embodiment, the disclosed methods reduce (after administration) the number of eosinophils by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%; at least about 90%, at least about 95%, or at least about 99%. In particular embodiments, the methods disclosed herein reduce the number of eosinophils below the limit of detection.
In another embodiment, the disclosed methods reduce the number of eosinophil precursors (after administration) by at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, or at least about 99%. In particular embodiments, the methods disclosed herein reduce the number of eosinophil precursors below the limit of detection.
Salts and solid forms of the present disclosure are useful for treating mast cell disorders. Salts and solid forms of the present disclosure are useful for treating mastocytosis. Mastocytosis is subdivided into two groups: (1) Cutaneous Mastocytosis (CM) descriptions are limited to the forms of skin; (2) Systemic Mastocytosis (SM) describes the form of mast cells infiltrating the extradermal organ with or without skin involvement. SM is further subdivided into five forms: inert (ISM); smoke emission (SSM); aggressiveness (ASM); SM is associated with related hematological non-mast cell lineage diseases (SM-AHNMD); and Mast Cell Leukemia (MCL).
The diagnosis of SM is based in part on histological and cytological studies of bone marrow, which show mast cell infiltration that is often morphologically atypical, often abnormally expressing non-mast cell markers (CD 25 and/or CD 2). Diagnosis of SM was confirmed when bone marrow mast cell infiltration occurred in the background of one of the following cases: (1) mast cell morphology abnormalities (spindle cells); (2) Serum tryptase levels were elevated above 20ng/mL; or (3) the presence of an activating KIT protein mutation, such as, for example, an exon 17 mutation, such as a D816 mutation, such as D816V.
The activating mutation at the D816 position was found in most cases of mastocytosis (90-98%), the most common mutations being D816V, D816H and D816Y. The D816V mutation is present in the activation loop of the protein kinase domain and results in constitutive activation of KIT kinase.
No drugs have been approved for use in non-advanced forms of systemic mastocytosis ISM or SSM. Current methods of controlling these chronic diseases include non-specific symptomatic-directed therapies that have varying degrees of efficacy and have no impact on MC burden. Cytoreductive therapies such as cladribine (cladribine) and interferon alpha are occasionally used for refractory symptoms. Based on the current treatment situation, the medical needs of ISM and SSM patients with moderate to severe symptoms remain unmet and available symptom-directed therapies do not adequately control the symptoms.
Salts and solid forms of the present disclosure are useful for treating ISM or SSM. In some embodiments, a patient with ISM or SSM has at least one, at least two, at least three symptoms that are not adequately controlled by symptomatic treatment. Symptoms can be assessed using patient report results (PRO) tools such as the inert systemic mastocytosis symptom assessment table (ISM-SAF) (ispro Europe 2019,Copenhagen Denmark,2019, 11 months 2-6 days). The salts and solid forms of the present disclosure may be used to ameliorate symptoms associated with ISM or SSM, for example, to reduce or eliminate itching, flushing, headache, and/or GI events, such as vomiting, diarrhea, and abdominal pain. Improvement of symptoms can be assessed using ISM-SAF.
The salts and solid forms of the present disclosure are useful for treating other mast cell disorders, such as Mast Cell Activation Syndrome (MCAS) and Hereditary Alpha Tryptase (HAT) (Picard clin.ther.2013, month 5 (5) 548;Akin J.Allergy Clin.Immuno.140 (2) 349 62 the salts and solid forms of the present disclosure are useful for treating mast cell disorders associated with KIT and PDGFR alpha mutations.
The salts and solid forms of the present disclosure are useful in the treatment of Mast Cell Activation Syndrome (MCAS), an immune condition in which mast cells inappropriately and excessively release chemical mediators, resulting in a range of chronic symptoms, sometimes including allergic reactions or near-allergic reaction attacks. Unlike mastocytosis, where the patient's number of mast cells is abnormally increased, MCAS patients have normal numbers of mast cells, which are not functioning properly and are defined as "hyperreactive". Types of MCAS include primary MCAS (monoclonal mast cell activation syndrome (MMAS)), secondary MCAS (MCAS caused by another disease), and idiopathic MCAS (MCAS excluding primary or secondary MCAS).
Disclosed herein are improved methods for treating Inert Systemic Mastocytosis (ISM) and monoclonal mast cell activation syndrome in a patient using compound (I) or a pharmaceutically acceptable salt thereof (e.g., salts and solid forms of the present disclosure). The present disclosure provides dosing regimens for compound (I) for the treatment of ISM and mcas. More specifically, the present disclosure provides methods for treating ISM and mcas in patients by administering compound (I) in a once daily dose in an amount of 15mg to 200 mg. It is an object of the present disclosure to provide a novel method of treating ISM and mcas in safe and effective once daily doses.
Non-limiting embodiments of the present disclosure include:
embodiment 1. A method of treating Inert Systemic Mastocytosis (ISM) or monoclonal mast cell activation syndrome (mcas), comprising orally administering to a patient in need thereof an amount of 15mg to 200mg of compound (I) once daily
Or a pharmaceutically acceptable salt thereof in an amount equivalent to 15mg to 200mg of compound (I).
Embodiment 2. The method of embodiment 1, wherein the patient has ISM.
Embodiment 3. The method of embodiment 1 or 2, wherein the patient has moderate to severe ISM.
Embodiment 4. The method of embodiment 3, wherein the patient has a Total Symptom Score (TSS) of 28 or greater at baseline of the inert systemic mastocytosis-symptom assessment table (ISM-SAF). In an alternative embodiment, the patient has a TSS <28 of ISM-SAF at baseline.
Embodiment 5. The method of any of embodiments 1-4, wherein the patient has ≡1 symptom at baseline in the skin or GI region of the inert systemic mastocytosis-symptom assessment table (ISM-SAF).
Embodiment 6. The method of embodiment 1, wherein the patient has mcas.
Embodiment 7. The method of any one of embodiments 1-6, wherein the amount is 25 to 100mg.
Embodiment 8. The method of embodiment 7, wherein the amount is 100mg.
Embodiment 9. The method of embodiment 7, wherein the amount is 50mg.
Embodiment 10. The method of embodiment 7, wherein the amount is 25mg.
Embodiment 11. The method of any one of embodiments 1-10, wherein compound (I) is administered as the free base.
Embodiment 12. The method of any one of embodiments 1-10, wherein compound (I) is administered as a pharmaceutically acceptable salt.
Embodiment 13. The method of embodiment 12, wherein the pharmaceutically acceptable salt of compound (I) is the salt. In some aspects, the salt is a phosphate salt. In some aspects, the salt is a benzenesulfonate salt. In some aspects, the salt is a benzoate salt. In some aspects, the salt is a sulfate salt.
Embodiment 14. The method of any one of embodiments 1-13, wherein the treatment results in a decrease in mast cell burden.
Embodiment 15. The method of any one of embodiments 1-14, wherein the treatment results in one or more symptoms of systemic mastocytosis.
Embodiment 16. The method of any one of embodiments 1-15, wherein the treatment results in a reduction in allergy onset.
Embodiment 17. The method of any one of embodiments 1-16, wherein the treatment results in an improvement in quality of life (QoL) as measured by one or more questionnaires.
Embodiment 18. The method of any one of embodiments 1-17, wherein treatment results in a decrease in the patient's TSS score as compared to the patient's TSS baseline score, as assessed by ISM-SAF.
In some embodiments, compound (I) in the form of a free base (including various solid forms of the free base disclosed herein) is administered to a patient. In some embodiments, compound (I) is administered to a patient in the form of a pharmaceutically acceptable salt, including various solid forms of the pharmaceutically acceptable salts disclosed herein. In some embodiments, the pharmaceutically acceptable salt is a phosphate salt.
In some embodiments, treating a patient with an amount of compound (I) disclosed herein or a pharmaceutically acceptable salt thereof comprises reducing mast cell burden. In some embodiments, the objective measure of mast cell burden comprises serum tryptase levels, bone marrow mast cell numbers, skin mast cell infiltration, and KIT D816V mutant allele burden in blood. In some embodiments, the objective measure of mast cell burden includes serum tryptase, bone marrow mast cell number, and KIT D816V mutant allele burden in the blood. In some embodiments, treating a patient with an amount of 50mg of compound (I) or an equivalent amount of a pharmaceutically acceptable salt thereof reduces serum tryptase levels in the patient. In some embodiments, treating a patient with an amount of 100mg of compound (I) or an equivalent amount of a pharmaceutically acceptable salt thereof reduces serum tryptase levels in the patient.
In some embodiments, treating a patient with an amount of compound (I) disclosed herein or a pharmaceutically acceptable salt thereof comprises reducing one or more symptoms of systemic mastocytosis. Systemic mastocytosis symptoms include, but are not limited to, itching, flushing, GI cramps, diarrhea, allergic reactions (especially to bee venom), bone pain, osteoporosis and urticaria pigmentosa. In some embodiments, the symptomatic improvement is assessed using ISM-SAF (self-assessment table) Patient Report Outcome (PRO) tools as defined herein. In some embodiments, the patient completes the ISM-SAF once a day prior to receiving the treatment, and the patient also completes the ISM-SAF once a day during the treatment. For example, the patient completes ISM-SAF within a period of time (e.g., four weeks) from the time of informed consent, during which the best support therapy (BSC) medication is optimized and stabilized. Once the data for the period of time (e.g., four weeks) is collected, ISM-SAF is completed once daily for an additional period of time (e.g., two weeks (14 days)) and patient eligibility is determined based on ISM-SAF symptom thresholds. Patients meeting the ISM-SAF qualification threshold then complete ISM-SAF once a day, while completing the screening procedure to assess study qualification.
Once all screening procedures were completed, baseline symptoms were collected over a period of time (e.g., 14 days) immediately prior to study entry. These data were used as baseline TSS. ISM-SAF was completed by patients once daily until the study was completed. In some embodiments, the primary endpoint of the study is the average change in ISM-SAF TSS from baseline. In some embodiments, treatment with compound (I) or a pharmaceutically acceptable salt thereof reduces the number of episodes of the allergic reaction. In some embodiments, an "allergic episode" is an allergic episode treated with epinephrine.
In some embodiments, treatment with an amount of compound (I) disclosed herein, or a pharmaceutically acceptable salt thereof, improves quality of life (QoL), as measured by one or more questionnaires. Non-limiting examples of QoL questionnaires include MC-QoL, PGIS, SF-12, PGIC, and EQ-5D-EL. MC-QoL is a disease-specific QoL tool developed specifically for ISM and CM patients (Siebenhaar, F. Et al, allergy 71 (6): 869-77 (2016)). MC-QoL contains 27 items, evaluating four areas: symptoms, emotions, social life/function, and skin. The project was evaluated in a 5 component table with a review period of two weeks. PGIS is a single item scale that evaluates a patient's perception of disease symptoms at a point in time. PGIS has been widely used to evaluate the overall perception of whether a patient is beneficial to treatment. SF-12 was developed for medical outcome studies, a study directed to chronically ill patients for many years. The tool is designed to ease the burden on the interviewee while achieving minimum accuracy criteria for purposes of group comparison involving multiple health dimensions. The questionnaire uses 8 health domains from the patient's perspective to measure health and wellbeing. The review period is four weeks. PGIC is a single item scale that evaluates a patient's perception of a change in symptoms of a disease at a point in time. EQ-5D-5L is a standardized tool for measuring general health. It consists of two parts, health description and evaluation. Health is measured by five dimensions (5D): ability to move; self-care; daily activities; pain/discomfort; and anxiety/depression. The interviewee self-assessed the severity of each dimension using a 5-component scale. The retrospective period is "today" (Whynes, d.k., health Qual Life Outcomes 6:94 (2008)).
In some embodiments, treating a patient with an amount of compound (I) disclosed herein, or a pharmaceutically acceptable salt thereof, improves bone density. Bone density was measured by scanning the lumbar vertebrae and hips by dual energy x-ray absorptiometry. In some embodiments, the treatment does not affect bone density.
As used herein, "SD" means stable disease.
As used herein, "CR" means complete reaction.
As used herein, "PFS" means progression free survival.
As used herein, the "inert systemic mastocytosis symptom assessment table" ("a"Medicines Corporation) or "ISM-SAF" (ispro European 2019, copenhagen Denmark,2019, 11 months 2-6) for example, for daily patient reporting results (PRO) assessment on electronic diaries. ISM-SAF is a 12-item PRO, specifically developed to evaluate symptoms in ISM and SSM patients. Although ISM-SAF was developed primarily for evaluating therapeutic efficacy hypotheses, it can also be used to screen participants for entry (or exit) into clinical studies based on minimal levels of sign and symptom severity. Eleven items shown in the table below were rated by a 10 component table (0 to 10, no to the most severe), and 1 item (diarrhea) was also evaluated for frequency.
/>
For the fields of skin/Skin Symptom Score (SSS), GI/Gastrointestinal Symptom Score (GSS), nonspecific symptom score, total Symptom Score (TSS), ISM-SAF generates a score for each item. TSS is the sum of all symptoms. In one aspect, the TSS is items 1-10 and 12. In one aspect, the GSS is items 2-3 and 12. In one aspect, the SSS is items 4-6. In one aspect, the patient completes the ISM-SAF daily within 4 weeks from the time of informed consent, during which the BSC medication is optimized and stabilized. Once the 4 week data was collected, ISM-SAF was completed daily for an additional 2 weeks (14 days) to determine patient eligibility based on ISM-SAF symptom thresholds. Patients meeting the ISM-SAF qualification threshold complete ISM-SAF daily, while completing the screening procedure to assess study qualification. Once all screening procedures were completed, baseline symptoms were collected within 14 days immediately prior to study entry. These data will be used as baseline TSS.
In one aspect, ISM patients have moderate to severe symptoms characterized by minimal TSS. In one aspect, ISM patients have moderate to severe symptoms characterized by a minimum TSS of 28 or more, e.g., usingISM-SAF evaluation. In one aspect, the minimum TSS of the skin and GI regions of ISM-SAF at baseline is provided for ISM or SSM patients with moderate to severe symptoms >28 and symptoms are more than or equal to 1. In one aspect, the TSS of an ISM patient<28. In one aspect, the baseline is a 14 day period prior to cycle 1, day 1 (C1D 1). In one aspect, the patient does not experience an acute symptom onset beyond their typical baseline symptoms. In one aspect, the patient fails to achieve symptomatic control of 1 or more baseline symptoms, as determined by the investigator, at least 2 of the following symptomatic treatments are administered at the optimal (approved) dose for a minimum of 4 weeks (28 days) before starting ISM-SAF to determine qualification: an H1 blocker, an H2 blocker, a proton pump inhibitor, a leukotriene inhibitor, cromolyn sodium, a corticosteroid, or omalizumab. In one aspect, the patient has<Baseline serum tryptase at 20 ng/mL. In one aspect, the patient has a baseline serum tryptase of ≡20 ng/mL.
In one aspect, treating a patient with compound (I) or a pharmaceutically acceptable salt thereof results in a decrease in TSS score as compared to the patient's TSS baseline score, as assessed by ISM-SAF. In one aspect, the patient's TSS is reduced by 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23.
In one aspect, ISM patients have a KIT D816V mutation. KIT D816V mutation can be detected by a high sensitivity assay, such as a drop digital polymerase chain reaction (ddPCR) assay with a limit of detection (LOD) of 0.022% Mutant Allele Frequency (MAF).
As used herein, "BSC" means optimal support therapy. More specifically, examples of best support therapeutic agents include:
in some embodiments, the proton pump inhibitor is co-administered to a patient treated with compound (I) or a pharmaceutically acceptable salt disclosed herein. In some embodiments, the proton pump inhibitor is co-administered to a patient treated with compound (I) or a pharmaceutically acceptable salt thereof under fasting conditions. In some embodiments, the patient is treated with compound (I) or a pharmaceutically acceptable salt herein, and the proton pump inhibitor is co-administered after a medium fat meal. In some embodiments, the patient is treated with compound (I) or a pharmaceutically acceptable salt herein, and the proton pump inhibitor is co-administered after a high fat meal.
As used herein, an "adverse event" or "AE" is any adverse medical event associated with the use of a drug in a human, whether or not it is considered to be associated with the drug. An AE (also referred to as an adverse experience) may be any adverse and unexpected sign (e.g., abnormal laboratory findings), symptom, or disease that is temporally related to the use of a drug without any judgment of causality. AE may be caused by any use of a drug (e.g., non-labeled use, use in combination with another drug) as well as any route of administration, formulation, or dosage (including excess).
As used herein, the terms "about" and "approximately" when used in connection with a dose, amount, or weight percent of a composition or component of a dosage form, include a specified dose, amount, or weight percent value or range of doses, amounts, or weight percent recognized by one of ordinary skill in the art that provides a pharmacological effect equivalent to that obtained from the specified dose, amount, or weight percent.
The present disclosure provides a method of treating inert systemic mastocytosis comprising administering to a patient in need thereof an amount of compound (I) of 15mg to 200mg or a pharmaceutically acceptable salt thereof in an amount equivalent to 55mg to 200mg once daily. In some embodiments, compound (I) is administered to a patient in need thereof in an amount of 10mg to 200mg once daily. In some embodiments, compound (I) (or a pharmaceutically acceptable salt thereof in an amount equivalent to 15mg, 25mg, 50mg, 100mg, 125mg, 150mg, 175mg, or 200mg of compound (I)) is administered to a patient in need thereof once daily. In some embodiments, the amount is 50mg to 200mg once a day. In some embodiments, the amount is 100mg to 200mg once a day. In some embodiments, the amount is 50mg to 100mg once a day.
The salts and solid forms of the present disclosure are useful for treating Hereditary Alpha Tryptase (HAT) (elevated tryptase caused by overexpression of TPSAB 1).
Other mast cell diseases include mast cell mediated asthma, allergic reactions (including idiopathic, ig-E and non-Ig-E mediated), urticaria (including idiopathic and chronic), atopic dermatitis, swelling (angioedema), irritable bowel syndrome, mastocytic gastroenteritis, mast cell colitis, pruritus, chronic pruritus, pruritus secondary to chronic renal failure, and cardiac, vascular, intestinal, brain, kidney, liver, pancreas, muscle, bone, and skin conditions associated with mast cells. In some embodiments, the mast cell disease is independent of mutant KIT or mutant pdgfrα.
KIT and PDGFRA mutations have been extensively studied in GIST. Salts and solid forms of the present disclosure are useful for treating GIST associated with KIT mutations. Salts and solid forms of the present disclosure are useful for treating unresectable or metastatic GIST. Nearly 80% of the metastatic GIST have primary activating mutations in the extracellular region (exon 9) or the membrane-proximal (JM) domain (exon 11) of the KIT gene sequence. Many mutant KIT tumors respond to treatment with targeted therapies, such as imatinib, which is a selective tyrosine kinase inhibitor that specifically inhibits BCR-ABL, KIT and PDGFRA proteins. However, most GIST patients eventually relapse due to the secondary mutation of KIT significantly reducing the binding affinity of imatinib. These resistance mutations always occur in the adenosine 5-triphosphate (ATP) binding pocket (exons 13 and 14) or in the activation loop (exons 17 and 18) of the kinase gene. None of the agents currently approved for GIST are selective targeting agents. Imatinib is currently approved for the treatment of GIST; multiple kinase inhibitors were used after imatinib. In many cases, these multi-kinase inhibitors, such as, for example, sunitinib, regorafenib, and Mi Duosi tenib (midostaurin), only weakly inhibit imatinib resistant mutants, and/or multi-kinase inhibitors are limited by more complex safety and small therapeutic windows. In some embodiments, the salts and solid forms of the present disclosure are useful for treating GIST of a patient who has been treated with imatinib. Salts and solid forms of the present disclosure may be used to treat GIST as a first line (1L), second line (2L), third line (3L), or fourth line (4L) therapy.
Salts and solid forms of the present disclosure are useful for treating GIST when no specific mutations are present or present in KIT. In some embodiments, the salts and solid forms of the present disclosure are capable of treating GIST when no specific mutation is present in KTI. In certain embodiments, the salts and solid forms of the present disclosure are incapable of treating GIST when a specific mutation is present in KTI. In some embodiments, the salts and solid forms of the present disclosure do not provide clinical benefit to patients carrying KIT ATP-binding pocket mutations (KIT protein mutations V654A, N655T and/or T670I).
Salts and solid forms of the present disclosure are useful for treating GIST associated with PDGFRA mutations. In 5% to 6% of unresectable or metastatic GIST patients, an activation loop mutation at protein amino acid 842 in exon 18 of the PDGFRA gene sequence occurs as the primary mutation.
Salts and solid forms of the present disclosure are also useful for treating AML. AML patients also carry KIT mutations, most of which are located at the D816 position of KIT proteins.
Disclosed herein is an improved process for preparing crystalline phosphate form a of compound (I):
comprising forming a phosphate of compound (I) with phosphoric acid in an organic solvent mixture comprising 2-MeTHF/acetone/water. In one embodiment, the ratio of the organic solvent mixture is 1.0 volume of 2-MeTHF to 1.0 volume of acetone to 1.3-2.0 volumes of water. In one embodiment, the amount of phosphoric acid is 1.1 equivalent to 1.0 equivalent of compound (I). In one embodiment, the method further comprises crystallizing the phosphate form a compound (I) by adding acetone. In one embodiment, acetone is added over a period of 4-8 hours.
The following examples are intended to be illustrative and are not intended to limit the scope of the present disclosure in any way.
Experiment
Abbreviations:
/>
analysis conditions
X-ray powder diffraction (XRPD)
Bruker
XRPD of the free base and phosphate disclosed herein was performed in reflection mode (i.e., bragg-Brentano geometry) using Bruker D8 Advance equipped with LYNXEYE detector, except for example 10 (the conditions indicated therein). Samples were prepared on silicon zero recovery wafers. The parameters of the XRPD method used are listed below:
Rigaku
the X-ray powder diffraction of the benzene sulfonates, benzoates, and sulfates disclosed herein was performed in reflection mode (i.e., bragg-Brentano geometry) using the Rigaku MiniFlex 600. Samples were prepared on silicon zero recovery wafers. The parameters of the XRPD method used are listed below:
simultaneous thermogravimetric analysis and differential scanning calorimetry (TGA and DSC)
Using Mettler Toledo TGA/DSC 3+ TGA and DSC were performed simultaneously on the same sample. The flow rates of the protective gas and the purge gas are respectively 20-30mL/min and 50-100mL/min. The required amount of sample (5-10 mg) was weighed directly into a sealed aluminum pan with pinholes and analyzed according to the following parameters:
differential Scanning Calorimetry (DSC)
Using Mettler Toledo DSC 3+ DSC is performed. Samples (1-5 mg) were weighed directly in 40 μl sealed aluminum pans with pinholes and analyzed according to the following parameters:
1 H-NMR Spectroscopy 1 H-NMR)
Proton NMR was performed on a Bruker Avance 300MHz spectrometer. The solid was dissolved in 0.75mL of deuterated solvent in a 4mL vial, transferred to an NMR tube (Wilmad 5mm thin wall 8"200MHz, 506-PP-8) and analyzed according to the following parameters:
high Performance Liquid Chromatography (HPLC)
HPLC was performed using Agilent 1220 info LC. The flow rate of the instrument is in the range of 0.2-5.0mL/min, the working pressure is in the range of 0-600 bar, the temperature is 5-60 ℃ higher than the ambient temperature, and the wavelength is in the range of 190-600nm.
HPLC was performed using Agilent 1220infinity 2lc equipped with a Diode Array Detector (DAD). The flow rate of the instrument is in the range of 0.2-5.0mL/min, the working pressure is in the range of 0-600 bar, the temperature is 5-60 ℃ higher than the ambient temperature, and the wavelength is in the range of 190-600nm.
HPLC was performed using Hitachi LaChrom HPLC. The flow rate of the instrument is in the range of 0.2-10.0mL/min, the working pressure is in the range of 0-412 bar, the temperature is 5-60 ℃ higher than the ambient temperature, and the wavelength is in the range of 190-600nm.
The HPLC method used in this study is shown below:
Karl fischer titration
KF titration of moisture determination was performed using a Mettler Toledo C20S coulometer KF titrator equipped with a current generator unit with a diaphragm and a double platinum needle electrode. The detection range of the instrument is 1ppm to 5% water. Will Aquastar TM CombiCoulomat fritless reagents are used for the anode and cathode compartments. About 0.03-0.10g of the sample was dissolved in the anode chamber and titrated until the solution potential dropped below 100mV. Verification was performed using a hydraulic 1 wt% water standard prior to sample analysis.
Microscopic method
Optical microscopy was performed using a Zeiss AxioScope A1 digital imaging microscope equipped with 2.5X, 10X, 20X and 40X objective lenses and polarizers. Images were taken with an built-in Axiocam 105 digital camera and processed using ZEN 2 (blue version) software supplied by Zeiss. Optical microscopy was performed using a Wetzlar Wilovert 30 inverted microscope equipped with a MiniVID USB 2.0,5.1MP digital camera.
Example 1: preparation of salts of Compound (I)
Stock solutions were prepared in TFE at a concentration of 85 mg/mL. Stock solutions of fourteen different counterions (e.g., benzenesulfonic acid, benzoic acid, phosphoric acid, sulfuric acid, citric acid, glucuronic acid, glutamic acid, hydrochloric acid, malic acid, methanesulfonic acid, succinic acid, tartaric acid, toluenesulfonic acid, and acetic acid) were prepared in EtOH. 352 μl (30 mg) of the free base solution was added to each vial. 1.1 and/or 2.2 equivalents of the appropriate counter ion stock solution are added to each vial. The vial was stirred at 45 ℃ for two hours, then the cap was opened and stirred in the atmosphere overnight and evaporated at 40 ℃. The vials were then placed under active vacuum at 50 ℃ for 3 hours to thoroughly dry. About 20 volumes (600 μl) of screening solvent were then added to each vial and the sample was heated to 45 ℃ with stirring (450 rpm). Vials exhibiting significant sedimentation or gumming are often vortexed to ensure thorough mixing. After two hours, the sample was stirred at room temperature. When the slurry was observed at room temperature, the solids were filtered for characterization.
Similar experiments were also performed in EtOAc and IPA in water (9:1 volumes).
From this salt screen, crystalline salts of benzenesulfonic acid, benzoic acid, hydrochloric acid, methanesulfonic acid, sulfuric acid, succinic acid and phosphoric acid were observed.
The crystalline salts were characterized and evaluated for feasibility (availability) based on melting point, crystallinity, stability to dry and humidity exposure, water solubility, polymorphism, and acceptability of the counterion.
The benzenesulfonates, benzoates and phosphates were all selected for further investigation on scale as they were stable to drying and/or re-drying after humidification and all melted at temperatures above 150 ℃. Sulfate was also selected for further study.
In contrast, solids isolated from maleic and acetic acid screens were not selected because they were not confirmed to be salts. Although a stable crystallization pattern was given throughout the screening process, no counter ion stoichiometry of acetate and malate was observed by NMR. Malate also has a significantly different appearance after humidification and exhibits a low melting point and poor water solubility.
Although HCl is the desired counterion, it was not selected due to its poor physicochemical properties; both the observed crystalline mode and amorphous solids deliquesce at high humidity. Methanesulfonate and toluenesulfonate were not selected because of poor acceptability of the counterions, their low crystallinity and deliquescence of the solids (amorphous or crystalline) upon humidification. Sulfuric acid shows 2 modes of stabilization to humidification, however, 9-a shows poor thermal behavior, while 9-B becomes a hard viscous solid after humidification.
Example 2: preparation of amorphous phosphate of Compound (I)
The amorphous material was produced by lyophilization from water and t-BuOH: water (6:4 volumes). Approximately 540mg of compound (I) form a phosphate (example 5) was weighed into a 20mL scintillation vial, and then the solvent was added. For water, 20mL was added in three steps while stirring at RT, but no dissolution was obtained. The vials were transferred to a hot plate at 75 ℃ and complete dissolution was achieved in a few minutes. However, the solution precipitated again after stirring for about 10 minutes. For t-BuOH: water, the initial mixing ratio was (9:1 volumes), but readjusted to (6:4 volumes) to achieve dissolution at 75 ℃. The solution was transferred to a 300mL beaker and a total of about 500 volumes of t-BuOH: water (6:4 volumes) were added.
The solution was split into 20mL vials and each vial was immersed in liquid nitrogen for about 2 minutes to freeze. The solid was transferred to a freeze dryer and kept overnight. XRPD analysis of the solid formed by lyophilization confirmed the amorphous pattern. See fig. 12.
Example 3: preparation and characterization of phosphates of Compound (I) form G
Amorphous slurry experiments:
about 62mg of amorphous material (phosphate of compound (I)) was weighed into a 4mL vial and 1mL of IPA was added to water (9:1 volumes) while stirring at RT. A clear solution with glue formed at the bottom of the vial and the glue was not destroyed by sonication or vortexing. The sample was stirred at RT for 2 days, after which it became a thick, flowable white slurry. An aliquot was removed and XRPD analysis confirmed form g+form a. XRPD analysis confirmed high crystalline form G after 9 days on a stir plate at RT. After drying overnight under static vacuum at 50 ℃, the crystallinity of form G decreased, and the high crystalline G sample left on the XRPD plate at ambient conditions decreased in crystallinity after 4 days.
Characterization:
the simultaneous TGA/DSC thermogram of crystalline form G (wet) shows a mass loss of 30 wt% between 40 ℃ and 140 ℃, in which three endothermic events occur, fig. 2C. A fourth endothermic peak corresponding to a melting starting temperature of 149.6 ℃ was observed and was consistent with the endothermic peak of 144.7 ℃ starting temperature of mode G obtained in the previous work, fig. 2D.
DSC and NMR analysis showed that form G was a hydrate (mass loss at <100℃was observed by DSC and only 0.3 wt% IPA was observed by NMR). The KF analysis determined that the sample contained 3.75 wt% water, confirming that the sample contained 1.3 molar equivalents of water.
Form G was left to stand at 40 ℃ and 75% relative humidity for 7 days to test its stability. About 10mg of compound (I) form G phosphate was weighed into a 4mL vial and covered with KimWipe. The vial was placed in a 20mL vial containing saturated aqueous NaCl. The system was placed on a 40 ℃ hotplate and an atmosphere of 75% relative humidity was created for 7 days. The solids were then spread for XRPD analysis. After one week, some peak shifts in XRPD were observed, which correspond well to form O, although they were not well resolved.
Example 4: preparation and characterization of phosphate of Compound (I) form O
Experiments using polymers to promote precipitation were performed using two solvent systems and two different polymers, table 45. About 30mg of compound (I) phosphate form a was weighed into a 2mL vial and the solvent was added while stirring at RT. For acetone to water (7:3 volumes), 1.4mL was added to achieve complete dissolution, and for THF to water (7:3 volumes), 400. Mu.L was added. Polyethylene glycol (PEG) and polyvinylpyrrolidone (PVP) polymers were gradually added, one wiper tip at a time, while precipitation was monitored. As the polymer was added, the solution became more viscous and became a thick, clear viscous solution, and no precipitation was observed in all four samples. The sample was left on the stir plate and after eight days a thick white slurry formed in the sample prepared in PEG-containing THF to water (7:3 volumes). XRPD analysis of the wet cake collected from this sample confirmed form O.
Table 45-summary of polymer crystallization experiments.
The blank cells indicate that no data is available.
Example 5: preparation and characterization of Compound (I) phosphate form A
5.1 Small Scale preparation
399.4mg of compound (I) (free base) was weighed into a 20mL scintillation vial and a stirring bar was added. 10 volumes (4 mL) of TFE were added to dissolve the solids with stirring at 45 ℃. 1.1 equivalent (3.5 mL) of the phosphoric acid stock solution was added drop-wise to the free base solution, immediately a white precipitate was seen. The solution was stirred for an additional 1 hour at 45 ℃, then the cover was removed, the temperature was adjusted to 40 ℃, and the solvent was allowed to evaporate for the whole weekend. Upon stirring at 45 ℃, the white precipitate disappeared and only peach gum was present. The glue will break due to sonication and vortexing, but the "glue ball" will quickly reform upon agitation. The vials were placed under active vacuum at 50 ℃ for 3 hours. Once removed, the vial contained an off-white solid. 20 volumes of EtOH (8.0 mL) were added to the vial and an off-white slurry appeared almost immediately. The slurry was allowed to stir at 45 ℃ for 1 hour, then at RT overnight.
The off-white slurry was sampled for XRPD analysis and form a was confirmed. The slurry was filtered, washed with 2x 1.5 volumes (599 μl) of EtOH, and dried under vacuum at 50 ℃.
385.2mg (counterion corrected yield 81%) of a white solid was collected and a Loss On Drying (LOD) of 63% was observed.
TGA analysis showed a gradual loss of mass of 0.3 wt% at up to 120 ℃ and then 1.9 wt% at up to 210 ℃ which corresponds to melting (fig. 1B). DSC shows two endothermic events; a small endothermic peak at an onset temperature of 161.3 ℃ and a larger endothermic peak at an onset temperature of 191.8 ℃ (fig. 1C).
Failed to pass 1 H NMR confirmed API: CI (counterion) ratio; however, the residual solvent was found to be 0.45 wt% EtOH. Microscopy revealed small needles.
The purity of the phosphate was found to be 98.20% a/a by HPLC.
5.2 Large Scale preparation
5.2.1
A solution of compound (I) (free base) (21.8 kg) in THF (405 kg) was filtered through a carbon filter at 45-55deg.C. The resulting solution was heated to 60-70 ℃ and distilled to remove 321kg THF. To this concentrated solution was added 16.5% by weight aqueous phosphoric acid (27 kg,1.1 eq) and mixed at 40-50 ℃. Water rinse (15 kg) was added and then further vacuum distilled at 40-50℃to remove about 40kg. Acetone (24 kg) was added to the resulting mixture at 40-50 ℃, followed by compound (I) phosphate seed crystals (26 g). Additional acetone (260 kg) was added and the mixture was then cooled to 15-25 ℃. The phosphate of compound (I) was isolated by filtration and washed twice with a mixture of acetone/water/THF (47 kg/6.4kg/7.3 kg) to give 26.1kg, >99% yield and 99.9% purity.
5.2.2
2-MeTHF (52 mL), acetone (52 mL), water (70 mL) and then compound (I) free base (25 g,1 eq.) and concentrated H were added to reactor R1 with stirring at ambient temperature 3 PO 4 (6.7 g,76.3 wt% assay, 1.1 eq). The mixture in R1 was heated to 50 ℃ and then fine filtered into a separate reactor R2, which reactor R2 was preheated at about 47 ℃. Acetone (8.7 mL) was added to R2 at about 47 ℃ followed by compound (I) phosphate seed (0.25 g,1 wt%). The mixture was stirred at about 47℃for 0.5h. The remaining acetone (450 mL) was added over 6h (240 mL over the first 4h, then) with slow stirring at the same temperatureThe remaining 210mL was added over 2 h). The resulting slurry was cooled to 20-25 ℃ for 18h and filtered. The wet cake was washed with a mixture of 2-MeTHF: acetone: water (9:80:11, 38mL x 2) and then dried under vacuum at 50deg.C to give 27.5g of compound (I) phosphate as a solid in 93% yield and 99.94% HPLC purity. XRPD analysis showed that the phosphate salt of compound (I) obtained was crystalline form a.
The original process for preparing the phosphate form a of compound (I) is carried out in a solvent mixture THF/acetone/water. However, as shown in Table A, the residual THF in phosphate form A of compound (I) prepared using this solvent mixture was near or above the maximum acceptable limit (.ltoreq.720 ppm) according to the guidelines for THF residual solvent of International pharmaceutical Commission (International Council for Harmonization, ICH). THF is a class II solvent, which is considered to be limited in pharmaceutical products due to its inherent toxicity according to the ICH guidelines.
TABLE A residual THF in a batch of compound (I) phosphate form A cGMP prepared with THF
Thus, an improved process for preparing compound (I) phosphate form a using 2-MeTHF instead of THF during crystallization was developed. 2-MeTHF is a class III solvent (potentially less toxic solvent) with a maximum acceptable limit of 5000ppm according to ICH guidelines. The R & D data indicate that the newly developed 2-MeTHF process yields compound (I) phosphate form a of good quality and has robust control over residual 2-MeTHF (table B).
TABLE B residual 2-MeTHF in Compound (I) phosphate form A cGMP batch prepared with 2-MeTHF
5.2 API to counterion ratio of phosphate form A of Compound (I)
To determine the ratio of API to counterion, a known concentration of the free base of compound (I) and the phosphate form a of compound (I) were formulated in a volumetric flask to a concentration of about 0.5 mg/mL. These samples were then injected for HPLC analysis. The total area counts under the main API peak were compared for intensity assessment. The strength was determined to be 88%, corresponding to 0.8 molar equivalents of phosphoric acid. Inductively coupled plasma atomic emission spectroscopy (ICP OES) was also performed on the form a sample. The phosphorus content was determined to be 4.65% by weight, which is very good agreement with the theoretical phosphorus content of 4.94% by weight of the monophosphate.
5.3 stability test
A. Short-term slurry
After the gravimetric solubility evaluation in 14 solvents at both temperatures, a short-term slurry preparation was performed. The vial containing the slurry was centrifuged and the settled solids were recovered and filtered for XRPD analysis. A summary of the results is presented in table 46. Samples prepared in NMP, DMAc and DMSO at RT remained in solution and no solids were collected. All other 23 samples formed a slurry and form a was obtained from 21 samples. Free base form A (FB-A) +phosphate form A was obtained in water at 50℃indicating disproportionation of the phosphate. The pattern obtained in TFE at 50 ℃ comes from degradation byproducts, as confirmed by HPLC analysis.
Table 46 summary of XRPD patterns obtained from short term slurry experiments.
/>
B. Long-term slurry and slow evaporation
Slow evaporation and long term slurry experiments were performed in 10 different solvents/solvent systems, table 47. About 8mg of compound (I) phosphate form a was weighed into a 2mL HPLC vial and 1.5mL of solvent was added while stirring at 35 ℃.
The solids in acetone: water (7:3 volumes) and ACN: water (8:2 volumes) were completely dissolved. These samples were crystallized by slow evaporation. A small gauge needle was inserted into the vial cap and the solvent evaporated while stirring at 35 ℃. The sample in pure solvent is a thin slurry and the solids are not completely dissolved. These samples were used for long term slurries. Water (75. Mu.L) was added to some of the samples (Table 47) and samples from ACN, THF and MeOAc were transferred to a cold plate at 10℃in order to detect possible hydrate formation. After 10 days, the sample in pure solvent was filtered and analyzed by XRPD. Phosphate form a was obtained for all samples. The free base form FB-A was obtained from two slowly evaporating samples.
Table 47 summarizes the results of long-term slurry and slow evaporation.
C. Grinding
Dry and solvent drop milling was performed using a Wig-L-Bug ball mill with 1/4' stainless steel balls as milling media. About 30mg of solid (compound (I) phosphate form a) was weighed into each container and one volume of solvent was added. Milling was performed at 3,800rpm in 3 x 30s increments and the solids on the vessel walls scraped off to minimize agglomeration between milling. The obtained pattern summary is shown in table 48. XRPD analysis showed that the solid obtained from dry milling had amorphous + trace form a.
Table 48 dry milling and solvent drop milling test results.
Am. amorphous form
Example 6: preparation and characterization of the Benzenesulfonate salt of Compound (I)
6.1 preparation of benzenesulfonate form 1-A
386.5mg of compound (I) free base was weighed into a 20mL scintillation vial and a stirring bar was added. 10 volumes (4 mL) of TFE were added to dissolve the solid under stirring at 45 ℃. 1.1 equivalent (4.3 mL) of the stock solution of benzenesulfonic acid was added drop-wise to the solution of the free base with no visible change. The solution was stirred for an additional 1 hour at 45 ℃, then the cover was removed, the temperature was adjusted to 40 ℃, and the solvent was allowed to evaporate for the whole weekend. The vials were placed under active vacuum at 50 ℃ for 3 hours. Once removed, the vial contained a thin orange gel/film on the wall of the vial, with no visible solids present. 20 volumes of EtOH (7.7 mL) were added to the vial and an off-white slurry appeared almost immediately. Some orange outer shell can be seen on the wall. The slurry was allowed to stir at 45 ℃ for 1 hour, then at RT overnight.
The off-white slurry was sampled for XRPD analysis and the benzenesulfonate form 1-a was confirmed. The slurry was filtered, washed with 2x 1.5 volumes (580 μl) of EtOH, and dried under vacuum at 50 ℃. 98.4mg (20% counterion corrected yield) of white solid was collected and a Loss On Drying (LOD) of 71% was observed.
TGA showed a mass loss of 0.9 wt% at up to 120 ℃ and 0.8 wt% at up to 200 ℃ (fig. 4B). Only one endothermic event was observed in the DSC thermogram, with an onset temperature of 189.4 ℃ (fig. 4C). By passing through 1 H NMR confirmed a stoichiometry of about 1:2api: ci. Due to the number of overlapping peaks, it is difficult to obtain accurate stoichiometry. The residual solvent was found to be 0.17 wt% EtOH.
The benzene sulfonate was found to have an amplified purity of 93.12% a/a by HPLC.
6.2 preparation of benzenesulfonate form 1-B
After the benzenesulfonate form 1-a was subjected to humidity testing and re-dried, benzenesulfonate form 1-B was formed.
Example 7: preparation and characterization of the benzoate salt of Compound (I)
7.1 preparation of benzoic acid salt form 2-A
389.8mg of compound (I) free base was weighed into a 20mL scintillation vial and a stirring bar was added. 10 volumes (4 mL) of TFE were added to dissolve the solid under stirring at 45 ℃. 1.1 equivalent (3.5 mL) of a stock solution of benzoic acid was added drop wise to the solution of the free base with no visible change. The solution was stirred for an additional 1 hour at 45 ℃, then the cover was removed, the temperature was adjusted to 40 ℃, and the solvent was allowed to evaporate for the whole weekend. The vials were placed under active vacuum at 50 ℃ for 3 hours. Once removed, the vial contained an off-white solid on the wall of the vial and an orange shell. 20 volumes of EtOAc (7.8 mL) were added to the vial and an off-white slurry appeared almost immediately. Some orange outer shell can be seen on the wall. The slurry was allowed to stir at 45 ℃ for 1 hour, then at RT overnight.
The off-white slurry was sampled for XRPD analysis and the benzoate form 2-a was confirmed. The slurry was filtered, washed with 2x 1.5 volumes (585 μl) of EtOAc, and dried under vacuum at 50 ℃. 347.1mg (counterion corrected yield 72%) of a white solid was collected and 54.7% LOD was observed.
TGA analysis showed a mass loss of 0.2 wt% at up to 120 ℃ and then 5.4 wt% (under melting) at up to 200 ℃, fig. 6B. The DSC thermogram shows two endothermic peaks at 171.3℃and 180.8 ℃and FIG. 6C. By passing through 1 H NMR confirmed the stoichiometry to be 1:1api: ci and found the residual solvent to be 0.83 wt% EtOAc. Microscopic methods reveal the small rods and needles.
The benzoate was found to have a purity of 96.53% a/a by HPLC.
7.2 preparation of benzoic acid salt form 2-B
After moisture testing and re-drying of benzoate form 2-A, benzoate form 2-B is formed.
Example 8: preparation and characterization of sulphates of Compound (I)
A stock solution of sulfuric acid in IPA: water (9:1 volume) was prepared in TFE at a concentration of 85 mg/mL. 352 μl (30 mg) of the free base solution was added to the vial. 1.1 equivalents of sulfuric acid stock solution was added to the vial. The vial was stirred at 45 ℃ for two hours, then the lid was opened and evaporated at 40 ℃ while stirring at atmospheric overnight. The vials were then placed under active vacuum at 50 ℃ for 3 hours to thoroughly dry. Then about 20 volumes (600 μl) of the screening solvent were added to the vials and the samples were heated to 45 ℃ with stirring (450 rpm). The sample was stirred at room temperature. The solids were filtered for characterization. XRPD analysis showed the formation of sulfate form 9-a.
Example 9: amorphous free base of Compound (I)
The solubility of compound (I) free base in various solvents was tested (table 49).
Table 49.
The solubility of the free base of compound (I) in Acetonitrile (ACN) is relatively good in water (7:3 volumes), t-butanol (t-BuOH) and t-BuOH in water (9:1 volumes). These solutions were filtered by syringe filtration (samples 83-1 and 83-2) and then used in lyophilization experiments. The samples were first frozen in liquid nitrogen and then placed in the vacuum system of a freeze dryer (0.045 mbar, -89 ℃ C. Collector temperature). Dried under vacuum overnight at RT, then the sample was removed and prepared for XRPD analysis. XRPD was recorded after one day of storage of the sample in a vial wrapped with parafilm and left on the bench at RT (see fig. 14).
Example 10: preparation of Compound (I) free base
Synthesis of (S) -2- (4- (4- (5- (1-amino-1- (4-fluorophenyl) ethyl) pyrimidin-2-yl) piperazin-1-yl) pyrrolo [1,2-f ] [1,2,4] triazin-6-yl) -1H-pyrazol-1-yl) ethanol
Synthesis of 6-bromo-4-chloropyrrolo [2,1-f ] [1,2,4] triazine:
at 120 ℃, 6-bromopyrrolo [2,1-f][1,2,4]Triazin-4-ol (10.0 g,46.7 mmol), phosphorus oxychloride @A mixture of 13.0mL,140 mmol) and triethylamine (13.0 mL,93.5 mmol) in toluene (100 mL) was stirred for 18h. The reaction mixture was concentrated. The residue was purified by flash chromatography on silica eluting with 1:10 to 1:2 EtOAc/petroleum ether to give the title compound (9.4 g,87% yield) as a pale yellow solid. MS (ES+) C 6 H 3 BrClN 3 The required value: 231, found: 232,234[ M+H ]] +
Synthesis of (S) -1- (2- (4- (6-bromopyrrolo [1,2-f ] [1,2,4] triazin-4-yl) piperazin-1-yl) pyrimidin-5-yl) -1- (4-fluorophenyl) ethylamine:
(S) -1- (4-fluorophenyl) -1- (2- (piperazin-1-yl) pyrimidin-5-yl) ethylamine hydrochloride (2.00 g,6.63 mmol), 6-bromo-4-chloropyrrolo [1, 2-f) at RT][1,2,4]A solution of triazine (2.31 g,9.94 mmol) and triethylamine (2.00 g,19.8 mmol) in dioxane (20 mL) was stirred for 2h. LC-MS showed complete conversion of the reaction. The mixture was purified by flash column chromatography (DCM/meoh=20/1) to give the title compound as a white solid (1.51 g,45% yield). MS (ES+) C 22 H 22 BrFN 8 The required value: 496, found: 480[ M-16 ]] +
Synthesis of (S) -2- (4- (4- (4- (5- (1-amino-1- (4-fluorophenyl) ethyl) pyrimidin-2-yl) piperazin-1-yl) pyrrolo [1,2-f ] [1,2,4] triazin-6-yl) -1H-pyrazol-1-yl) ethanol (compound (I):
(S) -1- (2- (4- (6-bromopyrrolo [1, 2-f)) at 100 DEG C][1,2,4]Triazin-4-yl) piperazin-1-yl) pyrimidin-5-yl) -1- (4-fluorophenyl) ethylamine (500 mg,1.00 mmol), 2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1H-pyrazol-1-yl) ethanol (284 mg,1.20 mmol), pd (dppf) Cl 2 (219 mg, 300. Mu. Mol) and Na 2 CO 3 (317 mg,3.00 mmol) in dioxane/water (20 mL/2 mL) under N 2 Stir overnight. LC-MS showed complete conversion of the reaction. The mixture was purified by flash column chromatography (DCM/meoh=15/1) and preparative HPLC (mobile phase: a=water (0.1% NH 4 HCO 3 ) B=acetonitrile; gradient: b=15-95% in 18 minutes; column: xtimate 10um 150A 21.2X1250 mm) to give the title compound as a white solid (154.0 mg,29% yield). MS (ES+) C 27 H 29 FN 10 O needs value: 528, found 529[ M+H ]] +1 H-NMR(400MHz,6d-DMSO)δppm 8.40(s,1H),8.07(s,1H),8.00(s,1H),7.87(s,1H),7.84(s,1H),7.49-7.44(m,2H),7.24(s,1H),7.14-7.07(m,2H),4.95(t,1H,J=5.2Hz),4.16-4.12(m,2H),4.11-4.07(m,4H),3.92-3.88(m,4H),3.77-3.72(m,2H),2.44(br.S.,2H),1.73(s,3H)。
XRPD spectra (fig. 15) and peaks (table 50) of the obtained products are shown below.
Table 50
Example 11: interconversion between crystalline forms of the free base
The same crystalline form of the free base compound (I) may contain different amounts of water (i.e., channel hydrate) and is therefore characterized by different subtypes, such as form FB-A-0 (monohydrate), form FB-A-1 (metastable hydrate) and form FB-A-2 (dehydrate). The crystalline forms FB-A, FB-A-1 and FB-A-2 can be interconverted with each other. For example, form FB-A-1 can be obtained from form FB-A by slurrying in MeOH or EtOH at 50℃or by slurrying in DCM: meOH (40:60 volumes) and rapid cooling. Form FB-A-1 can be converted to form FB-A under humidified conditions of 40℃and 75% RH.
Form FB-A-2 may be obtained from form FB-A-0 by drying in vacuo at 50℃for three days. When the sample was taken out of the oven, it was purged with nitrogen and immediately covered with an airtight stand and XRPD analysis was performed.
Example 12: phase 1, randomized, double-blind, placebo-controlled, single and multiple dose escalation studies to evaluate compound (I) safety, tolerability and pharmacokinetics
Parts 1 and 2 are random, double blind, placebo controlled studies of SAD (part 1) and MAD (part 2) in healthy adult subjects on oral administration of compound (I). Compound (I) is administered in the form of a monophosphate. Subject screening occurred within 28 days prior to (first) dosing. Subjects participated in only 1 cohort for only 1 study portion.
Part 1 (SAD): six (6) groups were evaluated, with 8 subjects (6 active drugs and 2 placebo) per group. Groups of subjects aged 18 or older received oral placebo (n=2/group) or a single oral dose of compound (I) 15, 25, 50, 100 or 200mg (n=6/group) under fasting conditions. The first group (group S1) included a sentinel group (1 active drug and 1 placebo) that was dosed at least 24 hours before the remaining 6 subjects (5 active drugs and 1 placebo). After safety evaluation of the sentinel group, the remaining 6 subjects in group S1 were dosed.
Plasma samples were collected for Pharmacokinetic (PK) assessment prior to dosing and within 96 hours after administration of compound (I) or placebo. Blood samples were collected prior to dosing on day-1, day 2 and day 5 to measure the concentration of Pharmacodynamic (PD) biomarkers (tryptase). Cardiac kinetic ECG samples were collected prior to dosing and within 24 hours after administration of compound (I).
Once the safety review board (SRC) determines that adequate safety and tolerability have been demonstrated, the next group is dose escalated.
Part 2 (MAD): four (4) groups were evaluated, with 8 subjects (6 active drugs and 2 placebo) per group. In each group, subjects received a daily oral dose of compound (I) or placebo of 25, 50 or 100mg at a 3:1 ratio for 10 consecutive days under fasting conditions.
Plasma samples were collected for PK assessment prior to dosing on days 1-10. Plasma samples were also collected for PK assessment within 24 hours after dosing on day 1 and within 96 hours after dosing on day 10. Blood samples were collected prior to day 1 dosing and prior to days 2, 5 and 9 dosing to measure PD biomarker (tryptase) concentrations. Cardiac kinetic ECG samples were collected prior to dosing and within 24 hours after administration of compound (I) on days 1 and 10. Safety was assessed according to the incidence and severity of adverse events in the recipients of compound (I) compared to placebo.
Compound (I) has good tolerability in five compound (I) SAD groups and three MAD groups. Only grade 1 adverse events were reported, mostly drug independent, including abdominal pain, loss of appetite, fatigue, headache, and nausea. No severe AEs were reported, nor were clinically relevant findings in laboratory or vital sign parameters reported. After administration of compound (I) in a single dose, the median Tmax ranges from 1.5 to 6 hours after administration. The average half-life (t 1/2) of compound (I) ranged from 20h to 28h, indicating that steady state was expected to be reached on day 7, supporting once-daily dosing. The geometric mean cumulative ratio of AUC was 1.6 to 1.8 after repeated oral administration for 10 days of 25 to 100mg of compound (I). The geometric mean Vz/F ranges from 753 to 973L, which indicates a broad tissue distribution. In summary, a dose-proportional increase in the systemic exposure of compound (I) was observed in the SAD and MAD groups. The pharmacokinetics of compound (I) are linear over the dose range in the SAD and MAD groups.
Example 13: compound (I) phosphate form a (referred to as "compound (I)" in this example 13) randomized, double-blind, placebo-controlled phase 2/3 study in inert systemic mastocytosis
This is a randomized, double-blind, placebo-controlled phase 2/3 study that compares the efficacy and safety of compound (I) + optimal supportive care (BSC) with placebo+bsc in ISM patients with symptoms not adequately controlled by BSC. In part 1, the Recommended Dose (RD) of compound (I) is determined in ISM patients with ISM-SAF TSS.gtoreq.28. In part 2, ISM patients, regardless of ISM-SAF TSS, were randomly assigned to rd+bsc of compound (I) identified in part 1 or to matched placebo+bsc. In part M, mcas patients received RD of open label compound (I). In part 3, patients who have completed study part 1 or part 2 are involved in long-term expansion to receive open label therapy for rd+bsc.
Screening (all parts)
After written informed consent is provided, the patient's eligibility will be assessed during the screening period.
The TSS qualification of part 1 is determined by daily average ISM-SAF over a 14 day period. Patients meeting the symptom severity threshold continue to complete ISM-SAF on a daily basis through screening and study participation if deemed eligible. After confirmation of ISM-SAF symptoms in part 1, the remaining screening evaluation was started.
Patients in part 2 were screened daily for ISM-SAF to determine baseline scores, but inclusion in the study was not dependent on the particular TSS. The accrued items are layered with TSS scores (< 28 and > 28) and the number of patients requiring TSS >28 is minimal.
The filter of sections 1, 2 and M includes the following: BM biopsies (archival or fresh samples obtained over the first 12 weeks; BM biopsies over the last 12 months were acceptable for mcas patients) and skin biopsies of both diseased and non-diseased skin (part 1 or part 2 cutaneous mastocytosis patients). BM and skin biopsies were performed and sent to a central pathology laboratory to confirm ISM or mcas diagnosis and to conduct MC quantification. Patients with cutaneous mastocytosis may choose to take a picture of the skin. Other procedures include Magnetic Resonance Imaging (MRI) or computed tomography of the brain, bone densitometry, serotyping, KIT D816V mutation test, allele burden, routine laboratory tests, ECG, and physical examination. The central laboratory requires ISM or mcas diagnostic confirmation.
The random assignment in parts 1 and 2 was performed after patients were considered eligible to participate after screening.
In part 1 of the study, approximately 40 patients with an estimated TSS.gtoreq.28 ISM were equally randomized (1:1:1:1 ratio) to 3 doses of one of compound (I) plus BSC or placebo plus BSC. Compound (I) +bsc and placebo+bsc were tested in parallel at 3 dose levels (25 mg, 50mg or 100 mg). Compound (I) is administered orally once daily (QD) consecutively. Patients were assessed weekly for the first 4 weeks for safety, laboratory monitoring, and quality of life (QoL) assessment. Pharmacokinetic sampling was performed on all patients. After dense PK collection (C1D 1 and C1D 15) was completed for all patients, the pharmacokinetic data were non-blind. ISM-SAF was completed daily. After 12 weeks of treatment, the central pathology laboratory repeatedly performed BM and skin biopsies for MC quantification, and can take skin photographs (optional) of patients with baseline cutaneous mastocytosis. RD was determined based on efficacy, safety and PK data at each dose level. The primary efficacy criteria for selecting RD was the dose of compound (I) that produced the greatest reduction in TSS, as assessed at week 13 compared to baseline using ISM-SAF. Other efficacy metrics (e.g., variation in serum tryptase) are also contemplated. Once the week 13 evaluation is completed, the patients continue to receive the dispensed therapy and dose until RD is determined, at which point all patients in part 1 are blind and transition to part 3, where they receive compound (I) in an open label manner as RD.
In part 2 of the study, up to 303 ISM patients (at least 204 evaluable TSS >28 patients and up to 99 TSS <28 patients) were enrolled. Patients were randomly allocated to treatment at a ratio of 2:1 to receive rd+bsc or matched placebo+bsc of compound (I), respectively. Random assignment was stratified based on TSS scores (< 28 and > 28) and serum tryptase levels (< 20ng/mL vs. 20 ng/mL) measured centrally at screening. In addition, the upper panel limit for patients with TSS.gtoreq.28 is approximately 20% for patients with serum tryptase <20ng/mL and approximately 20% for patients with TSS < 28.
Compound (I) and placebo were administered continuous QD oral administration. Patients were assessed every 4 weeks for safety, laboratory monitoring, and QoL assessments, until week 25. Sparse PK sampling was performed on all patients. For patients with cutaneous mastocytosis to choose to do so, skin photographs are taken every 12 weeks. ISM-SAF was completed daily.
After 24 weeks of treatment is completed, the central pathology laboratory repeatedly performs ISM-SAF, BM and skin biopsies for Mast Cell (MC) quantification, and can take skin photographs (optional) of patients with baseline cutaneous mastocytosis. Each patient completing the 25 th week evaluation will shift to part 3 of the long-term expansion, thereby receiving RD of compound (I) QD in an open-label manner. Week 25 evaluation is part 3 baseline. After all patients in part 2 transferred to part 3, all part 2 treatment assignments were non-blind. At this point, the primary endpoint was analyzed: patient proportion of > 30% TSS decrease from baseline to week 25, as well as other efficacy endpoints.
After all fraction 1 patients completed fraction 1 and RD was determined, or after each patient in fraction 2 completed their study evaluation at week 25, the patient shifted to fraction 3. All patients received open label treatment of RD of compound (I).
In part 3, part 1 patients receiving compound (I) were subjected to study visits every 4 weeks until week 25, then every 8 weeks until week 49. After week 49, the patient visits once every 12 weeks for a total treatment duration of up to 5 years, including part 1 or part 2 (if applicable). Part 1 patients receiving placebo were visited once a week until week 5.
In part 3, patients diverted from part 2 were visited once a week until week 5, and then follow the same schedule as part 1 patients.
Patients in part 1 and part 2 completed ISM-SAF daily, and QoL assessment was performed at study visit at part 3, week 49 or end of treatment (EOT), whichever occurred earlier. For patients with or developing cutaneous mastocytosis during part 1 or part 2, skin photographs (optional) may be taken at part 3 baseline (if not obtained within the first 4 weeks of part 1 or part 2) and other time points. The central pathology laboratory repeatedly performed optional BM and skin biopsies 1 year after part 1 week 13 biopsies or 1 year after part 2 week 25 biopsies for MC quantification. For patients assigned to placebo in part 1 or part 2, the end of part 1 or part 2 study evaluation (including BM and skin biopsies) served as part 3 baseline evaluation.
In part M of the study, about 20 mcas patients received rd+bsc of compound (I) QD in the open label panel. The dosages, formulations and methods of administration are the same as described in section 2. This section does not include placebo. After 24 weeks of treatment was completed, the central pathology department repeatedly performed BM biopsies for MC quantification. The patient continues with the long-term follow-up portion of the mth portion.

Claims (67)

1. A phosphate of a compound (I) represented by the following formula:
wherein the molar ratio between the compound (I) and phosphoric acid is 1:1.
2. The phosphate of claim 1, wherein the phosphate is crystalline.
3. The phosphate of claim 1, wherein the phosphate is in a single crystalline form.
4. A phosphate salt according to any one of claims 1-3, wherein the phosphate salt is unsolvated.
5. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form a, characterized by an X-ray powder diffraction pattern comprising peaks at 4.6 °, 11.8 °, 13.7 °, 18.3 ° and 19.8 ° ± 0.2 in 2Θ.
6. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form a, characterized by an X-ray powder diffraction pattern comprising peaks at 4.6 °, 11.8 °, 13.7 °, 18.3 °, 19.8 ° and 21.8 ° ± 0.2 in 2Θ.
7. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form a, characterized by an X-ray powder diffraction pattern comprising peaks at 4.6 °, 9.2 °, 11.8 °, 13.7 °, 16.3 °, 18.3 °, 19.8 ° and 21.8 ° ± 0.2 in 2Θ.
8. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form a, characterized by an X-ray powder diffraction pattern substantially similar to figure 1A.
9. The phosphate salt of any one of claims 2-8, wherein the crystalline salt is form a, characterized by a thermogravimetric analysis (TGA) substantially similar to figure 1B.
10. The phosphate salt of any one of claims 2-9, wherein the crystalline salt is form a, characterized by a Differential Scanning Calorimetry (DSC) peak phase transition temperature of 161.3±2 ℃ and 191.8±2 ℃.
11. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form G, characterized by an X-ray powder diffraction pattern comprising peaks at 4.1 °, 4.4 °, 7.4 °, 15.6 ° and 23.0 ° ± 0.2 in 2Θ.
12. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form G, characterized by an X-ray powder diffraction pattern comprising peaks, in terms of 2Θ, at 4.1 °, 4.4 °, 7.4 °, 15.0 °, 15.6 °, 18.7 °, 22.5 °, and 23.0 ° ± 0.2.
13. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form G, characterized by an X-ray powder diffraction pattern comprising peaks, in terms of 2Θ, at 4.1 °, 4.4 °, 7.4 °, 8.1 °, 10.6 °, 11.4 °, 15.0 °, 15.6 °, 18.7 °, 20.7 °, 22.5 °, and 23.0 ° ± 0.2.
14. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form G, characterized by an X-ray powder diffraction pattern substantially similar to figure 2A.
15. The phosphate salt of any one of claims 2-4 and 11-14, wherein the crystalline salt is form G, characterized by a Differential Scanning Calorimetry (DSC) peak phase transition temperature of 28.8 ± 2 ℃, 34.7 ± 2 ℃, 99.7 ± 2 ℃, 144.7 ± 2 ℃ and 152.5 ± 2 ℃.
16. The phosphate salt of any one of claims 2-4 and 11-15, wherein the crystalline salt is form G, characterized by a thermogravimetric analysis (TGA) substantially similar to figure 2C.
17. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form O, characterized by an X-ray powder diffraction pattern comprising peaks at 4.2 °, 15.2 °, 21.6 °, 21.9 ° and 22.6 ° ± 0.2 in 2Θ.
18. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form O, characterized by an X-ray powder diffraction pattern comprising at least six or seven peaks selected from 4.2 °, 4.3 °, 15.2 °, 15.4 °, 21.6 °, 21.9 °, 22.6 ° and 24.8 ° ± 0.2 in 2Θ.
19. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form O, characterized by an X-ray powder diffraction pattern comprising peaks, in terms of 2Θ, at 4.2 °, 4.3 °, 15.2 °, 15.4 °, 21.6 °, 21.9 °, 22.6 °, and 24.8 ° ± 0.2.
20. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form O, characterized by an X-ray powder diffraction pattern comprising peaks, in terms of 2Θ, at 4.2 °, 4.3 °, 11.7 °, 15.2 °, 15.4 °, 17.9 °, 20.6 °, 21.6 °, 21.9 °, 22.6 °, 23.4 °, and 24.8 ° ± 0.2.
21. The phosphate salt of any one of claims 2-4, wherein the crystalline salt is form O, characterized by an X-ray powder diffraction pattern substantially similar to figure 3.
22. A benzenesulfonate salt of a compound (I) represented by the following formula:
wherein the molar ratio between the compound (I) and the benzenesulfonic acid is 2:1.
23. The benzenesulfonate of claim 22, wherein the benzenesulfonate is crystalline.
24. The benzenesulfonate salt of claim 23, wherein the crystalline form comprises a form 1 family selected from the group consisting of crystalline forms of form 1-a and form 1-B, characterized by at least one of the following:
(a) An X-ray powder diffraction pattern (XRPD) substantially the same as shown in figure 4A or figure 5;
(b) An X-ray powder diffraction pattern (XRPD) comprising peaks selected from 5.7 °, 7.1 °, 9.7 °, 15.4 ° and 24.8 ° ± 0.2 in 2Θ or 5.5 °, 10.4 °, 14.0 ° and 16.3 ° ± 0.2 in 2Θ;
(c) Thermogravimetric analysis (TGA) is substantially similar to fig. 4B;
(d) The DSC thermogram substantially similar to figure 4C;
(e) The DSC thermogram has an endothermic peak with an onset temperature of 189.4 ℃; or (b)
(f) A combination thereof.
25. The benzenesulfonate salt of any one of claims 22-24, wherein the benzenesulfonate salt is in a single crystalline form.
26. The benzenesulfonate salt of any one of claims 22-25, wherein the benzenesulfonate salt is unsolvated.
27. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-a, characterized by an X-ray powder diffraction pattern comprising peaks at 5.7 °, 7.1 °, 9.7 °, 15.4 °, and 24.8 ° ± 0.2 in 2Θ.
28. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-a, characterized by an X-ray powder diffraction pattern comprising at least four, five, six, or seven peaks selected from 5.7 °, 7.1 °, 8.9 °, 9.7 °, 15.4 °, 19.8 °, 21.9 °, and 24.8 ° ± 0.2 in 2Θ.
29. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-a, characterized by an X-ray powder diffraction pattern comprising peaks, in terms of 2Θ, selected from 5.7 °, 7.1 °, 8.9 °, 9.7 °, 15.4 °, 19.8 °, 21.9 °, and 24.8 ° ± 0.2.
30. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-a, characterized by an X-ray powder diffraction pattern comprising peaks at 5.7 °, 7.1 °, 8.9 °, 9.7 °, 10.6 °, 15.4 °, 17.7 °, 19.8 °, 20.8 °, 21.9 °, 22.9 °, and 24.8 ° ± 0.2 in 2Θ.
31. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-B, characterized by an X-ray powder diffraction pattern comprising peaks at 5.5 °, 10.4 °, 14.0 °, and 16.3 ° ± 0.2 in 2Θ.
32. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-B, characterized by an X-ray powder diffraction pattern comprising at least three peaks selected from 5.5 °, 10.4 °, 14.0 °, 16.3 °, 18.6 °, and 19.0 ° ± 0.2 in 2Θ.
33. The benzenesulfonate salt of any one of claims 22-26, wherein the crystalline salt is form 1-B, characterized by an X-ray powder diffraction pattern comprising peaks, in terms of 2Θ, at 5.5 °, 10.4 °, 10.9 °, 14.0 °, 16.3 °, 18.6 °, 19.0 °, 20.9 °, 21.8 °, and 24.6 ° ± 0.2.
34. A sulfate salt of a compound (I) represented by the following formula:
wherein the molar ratio between the compound (I) and sulfuric acid is 1:1.
35. The sulfate salt of claim 34, wherein the sulfate salt is crystalline.
36. The sulfate salt of claim 34, wherein the sulfate salt is in a single crystalline form.
37. The sulfate salt of any one of claims 34-36, wherein the sulfate salt is unsolvated.
38. The sulfate salt of any one of claims 34-36, wherein the crystalline salt is form 9-a, characterized by an X-ray powder diffraction pattern comprising peaks at 9.0 °, 10.4 °, 13.6 °, 18.1 ° and 21.0 ° ± 0.2 in 2Θ.
39. The sulfate salt of any one of claims 34-36, wherein the crystalline salt is form 9-a, characterized by an X-ray powder diffraction pattern comprising peaks at 9.0 °, 10.4 °, 13.6 °, 18.1 °, 21.0 °, 22.7 °, and 27.7 ° ± 0.2 in 2Θ.
40. The sulfate salt of any one of claims 34-36, wherein the crystalline salt is form 9-a, characterized by an X-ray powder diffraction pattern comprising peaks at 8.6 °, 9.0 °, 10.4 °, 13.6 °, 17.4 °, 18.1 °, 18.9 °, 21.0 °, 22.7 °, 23.9 °, 27.2 °, and 27.7 ° ± 0.2 in 2Θ.
41. The sulphate salt according to any one of claims 34 to 40 wherein the crystalline salt is in form 9-a characterised by an X-ray powder diffraction pattern substantially similar to figure 8A.
42. The sulfate salt of any one of claims 34-41, wherein the crystalline salt is form 9-a, characterized by a Differential Scanning Calorimetry (DSC) peak phase transition temperature of 102.0 ± 2 ℃, 148.7 ± 2 ℃ and 158.2 ± 2 ℃.
43. A benzoate salt of a compound (I) represented by the following formula:
wherein the molar ratio between the compound (I) and the benzoic acid is 1:1.
44. The benzoate according to claim 43, wherein said benzoate salt is crystalline.
45. The benzoate salt according to claim 43, wherein said benzoate salt is in a single crystalline form.
46. The benzoate salt of any one of claims 43-45, wherein the benzoate salt is non-solvated.
47. The benzoate salt of any one of claims 43-46, wherein said crystalline salt is form 2-a, wherein the X-ray powder diffraction pattern includes peaks at 4.1 °, 10.6 °, 16.8 °, 18.1 ° and 22.7 ° ± 0.2 in 2Θ.
48. The benzoate salt of any one of claims 43-46 wherein said crystalline salt is form 2-a, characterized by an X-ray powder diffraction pattern comprising at least four, five, six or seven peaks selected from 4.1 °, 8.3 °, 10.6 °, 16.8 °, 18.1 °, 22.7 ° and 28.5 ° ± 0.2 in 2Θ.
49. The benzoate salt of any one of claims 43-46, wherein said crystalline salt is form 2-a, wherein the X-ray powder diffraction pattern includes peaks, expressed in 2Θ, selected from 4.1 °, 8.3 °, 10.6 °, 16.8 °, 18.1 °, 22.7 ° and 28.5 ° ± 0.2.
50. The benzoate salt of any one of claims 43-46, wherein said crystalline salt is form 2-a, wherein the X-ray powder diffraction pattern includes peaks, expressed in 2Θ, selected from 4.1 °, 8.3 °, 10.6 °, 15.0 °, 15.8 °, 16.8 °, 18.1 °, 20.1 °, 21.0 °, 22.7 °, 25.7 ° and 28.5 ° ± 0.2.
51. Benzoate salt according to any one of claims 43-50 wherein said crystalline salt is in form 2-a, characterized by an X-ray powder diffraction pattern substantially similar to that of figure 6A.
52. The benzoate salt of any one of claims 43-51, wherein said crystalline salt is form 2-a, wherein thermogravimetric analysis (TGA) is substantially similar to figure 6B.
53. The benzoate salt of any one of claims 43-52, wherein said crystalline salt is form 2-a, characterized by a Differential Scanning Calorimetry (DSC) peak phase transition temperature of 171.3 ± 2 ℃ and 180.8 ± 2 ℃.
54. Benzoate salt according to any one of claims 43-46, wherein said crystalline salt is form 2-B, characterized by an X-ray powder diffraction pattern comprising peaks at 5.8 °, 9.7 °, 15.4 ° and 17.8 ° ± 0.2 in 2Θ.
55. Benzoate salt according to any one of claims 43-46 wherein said crystalline salt is form 2-B, characterized by an X-ray powder diffraction pattern comprising at least four peaks selected from the group consisting of 5.8 °, 7.1 °, 9.7 °, 10.7 °, 15.4 °, 17.8 ° and 22.9 ° ± 0.2 in 2Θ.
56. Benzoate salt according to any one of claims 43-46 wherein said crystalline salt is form 2-B, characterized by an X-ray powder diffraction pattern comprising peaks, expressed in 2Θ, selected from 5.8 °, 7.1 °, 9.7 °, 10.7 °, 15.4 °, 17.8 ° and 22.9 ° ± 0.2.
57. The benzoate salt of any one of claims 43-46, wherein said crystalline salt is form 2-B, wherein the X-ray powder diffraction pattern includes peaks, in terms of 2Θ, at 5.8 °, 7.1 °, 9.7 °, 10.7 °, 15.4 °, 16.1 °, 17.8 °, 22.9 °, 24.8 ° and 27.0 ° ± 0.2.
58. An amorphous form of compound (I) represented by the formula:
59. the amorphous form of claim 58 wherein the X-ray powder diffraction pattern is substantially the same as figure 14.
60. A pharmaceutical composition comprising a salt of compound (I) according to any one of claims 1 to 57 or an amorphous form of compound (I) according to claims 58 to 59 and a pharmaceutically acceptable carrier or excipient.
61. A method of treating a disease or condition in a patient in need thereof, wherein the method comprises administering to the patient a salt of compound (I) of any one of claims 1-57, an amorphous form of compound (I) of claims 58-59, or a pharmaceutical composition of claim 60, wherein the disease or condition is selected from systemic mastocytosis, gastrointestinal stromal tumor, acute myelogenous leukemia, melanoma, seminoma, mediastinal B-cell lymphoma, ewing's sarcoma, diffuse large B-cell lymphoma, asexual cell tumor, myelodysplastic syndrome, nasal NK/T-cell lymphoma, chronic granulomonocytic leukemia.
62. A method of treating Inert Systemic Mastocytosis (ISM) or monoclonal mast cell activation syndrome (mcas), comprising orally administering to a patient in need thereof an amount of 15mg to 200mg of compound (I) once daily
Or a pharmaceutically acceptable salt thereof in an amount equivalent to 15mg to 200mg of compound (I).
63. A process for preparing crystalline phosphate form a of compound (I):
comprising forming a phosphate of compound (I) with phosphoric acid in an organic solvent mixture comprising 2-MeTHF/acetone/water.
64. The process of claim 63 wherein the ratio of the organic solvent mixture is 1.0 volumes of 2-MeTHF to 1.0 volumes of acetone to 1.3-2.0 volumes of water.
65. The process of claim 63 or 64 wherein the amount of phosphoric acid is 1.1 equivalent to 1.0 equivalent of compound (I).
66. The method of any one of claims 63-65, further comprising crystallizing the phosphate form a compound (I) by adding acetone.
67. The process of any one of claims 63-66, wherein said acetone is added over a period of 4-8 hours.
CN202280033117.2A 2021-03-10 2022-03-10 Salts and solid forms of kinase inhibitors Pending CN117794935A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US63/159,107 2021-03-10
US202163208641P 2021-06-09 2021-06-09
US63/208,641 2021-06-09
PCT/US2022/019776 WO2022192558A1 (en) 2021-03-10 2022-03-10 Salt and solid forms of a kinase inhibitor

Publications (1)

Publication Number Publication Date
CN117794935A true CN117794935A (en) 2024-03-29

Family

ID=90394948

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202280033117.2A Pending CN117794935A (en) 2021-03-10 2022-03-10 Salts and solid forms of kinase inhibitors

Country Status (1)

Country Link
CN (1) CN117794935A (en)

Similar Documents

Publication Publication Date Title
CN109970745B (en) Substituted pyrrolotriazine compound, pharmaceutical composition and application thereof
AU2014254050B2 (en) Pharmaceutical formulations, processes, solid forms and methods of use relating to 1-ethyl-7-(2-methyl-6-(1h-1,2,4-triazol-3-yl) pyridin-3-yl) -3,4-dihydropyrazino[2,3-b]pyrazin-2(1h)-one
EP3502103B1 (en) Crystal form, salt type of substituted 2-hydro-pyrazole derivative and preparation method therefor
WO2017092413A1 (en) Diaminopyrimidine compounds and composition comprising same
US11713309B2 (en) Solid forms of Cerdulatinib
EP3741753A1 (en) Substituted diamino heterocyclic carboxamide compound and a composition containing the compound and use thereof
WO2022109577A1 (en) Fgfr inhibitors and methods of making and using the same
US9453011B2 (en) Crystal form of dabrafenib mesylate and preparation method thereof
TW202016114A (en) Crystal form of compound for inhibiting the activity of cdk4/6 and use thereof
US9981971B2 (en) Solid forms of 1-ethyl-7-(2-methyl-6-(1H-1,2,4-triazol-3-yl)pyridin-3-yl)-3,4-dihydropyrazino[2,3-b]pyrazin-2(1H)-one as TOR kinase inhibitors
US20230339901A1 (en) Solid forms comprising (s)-2-(2,6-dioxopiperidin-3-yl)-4-((2-fluoro-4-((3-morpholinoazetidin-1-yl)methyl)benzyl)amino)isoindoline-1,3-dione and salts thereof, and compositions comprising and methods of using the same
CN117794935A (en) Salts and solid forms of kinase inhibitors
KR20240013720A (en) Salt and solid forms of kinase inhibitors
CN110256408B (en) Diaryl pyrazole compound, composition containing compound and application of diaryl pyrazole compound
EP3111960B1 (en) Nuclear medicine diagnostic imaging agent
US20190062307A1 (en) Deuterium-substituted quinoline derivatives
US20230399331A1 (en) Solid forms of jak inhibitor and process of preparing the same
WO2023041061A1 (en) Fused bicyclic derivative, pharmaceutically acceptable salt, crystal form thereof and preparation method therefor
TW201125861A (en) CDC7 inhibitor salts
TW202246256A (en) Crystalline forms of a cyclin-dependent kinase inhibitor
CA3232101A1 (en) Crystalline forms of quinazoline derivatives, preparation, composition and use thereof
TW202342041A (en) Pharmaceutical composition and method for preparing active ingredient compound thereof
CN117247382A (en) Crystal forms of pyridopyrimidinone compounds

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination