WO2021113661A1 - Amorphous and polymorphic form of a specific chk1 inhibitor - Google Patents

Amorphous and polymorphic form of a specific chk1 inhibitor Download PDF

Info

Publication number
WO2021113661A1
WO2021113661A1 PCT/US2020/063346 US2020063346W WO2021113661A1 WO 2021113661 A1 WO2021113661 A1 WO 2021113661A1 US 2020063346 W US2020063346 W US 2020063346W WO 2021113661 A1 WO2021113661 A1 WO 2021113661A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
crystalline form
peaks
crystalline
substantially crystalline
Prior art date
Application number
PCT/US2020/063346
Other languages
French (fr)
Inventor
Hui Li
Omid SOLTANI
Tanise SHATTOCK-GORDON
Dayane Vanessa Morais HEWITT
Original Assignee
Seagen Inc.
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 Seagen Inc. filed Critical Seagen Inc.
Publication of WO2021113661A1 publication Critical patent/WO2021113661A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the invention relates to solid-state forms of a Chk1 inhibitor, including polymorphic and amorphous forms, and methods for preparing the solid-state forms.
  • polymorphism The ability of a substance to exist in more than one crystalline form is defined as polymorphism and these different crystalline forms are named "polymorphs".
  • polymorphism is caused by the ability of the molecule of a substance to change its conformation or to form different intermolecular and intramolecular interactions giving different atom arrangements that are reflected in the crystal lattices of different polymorphs.
  • polymorphism is not a universal feature of solids, since some molecules can exist in one or more crystal forms while other molecules cannot.
  • each polymorph typically shows one or more different physical properties in the solid state, such as density, melting point, color, stability, dissolution rate, flowability, compatibility with milling, granulation and compacting and/or uniformity of distribution (see, e.g., P. DiMartino, et al., J. Thermal Anal. 48:447458 (1997)). Therefore, the capacity of any given compound to occur in one or more crystalline forms (i.e. , polymorphs) is unpredictable as are the physical properties of any single crystalline form.
  • Different physical properties of a substance may affect the ability to prepare different pharmaceutical formulations comprising the substance and may also affect the stability, dissolution and bioavailability of a solid-state formulation, which subsequently affects suitability or efficacy of such formulations in treating disease.
  • thermodynamically stable forms can occasionally convert to the more thermodynamically stable form at a given temperature after a sufficient period of time.
  • a thermodynamically unstable form When this transformation is not rapid, such a thermodynamically unstable form is referred to as a "metastable" form.
  • the stable form exhibits the highest melting point, the lowest solubility, and the maximum chemical stability.
  • the metastable form may exhibit sufficient chemical and physical stability under normal storage conditions to permit its use in a commercial form.
  • the metastable form although less thermodynamically stable, may exhibit properties desirable over those of the stable form, such as enhanced solubility or better oral bioavailability.
  • the amorphous form of an active pharmaceutical ingredient may have enhanced solubility in comparison to crystalline material due reduction of crystal lattice forces in the amorphous material that must be overcome to effect dissolution.
  • Chk1 is a serine/threonine kinase that is involved in the induction of cell cycle checkpoints in response to DNA damage and replicative stress. Chk1 inhibition abrogates the intra S and G2/M checkpoints and has been shown to selectively sensitize tumor cells to well-known DNA damaging agents. (See, e.g., McNeely, S. et al. Pharmacology & Therapeutics 2014 (dx.doi.org/10.1016/j.pharmthera.2013.10.005)).
  • Chk1 inhibitors are useful, as an example, in treating tumor cells in which constitutive activation of DNA damage and checkpoint pathways drives genomic instability.
  • WO 2015/120390 describes a series of Chk1 inhibitors, including the compound 5-((5-(4-(4- fluoro-1-methylpiperidin-4-yl)-2-methoxyphenyl)-1H-pyrazol-3-yl)amino)pyrazine-2- carbonitrile (see Example 64).
  • CHK1 inhibitor for use in preparing a composition to treat cancer in a subject will sometimes affect the development of that inhibitor as a medicament.
  • an individual solid-state form of a CHK1 inhibitor having one or more desirable properties would be selected for the development of a pharmaceutical formulation having the desired property(ies).
  • Compound 1 5-((5-(4-(4-fluoro-1-methylpiperidin-4-yl)-2-methoxyphenyl)-1H- pyrazol-3-yl)amino)pyrazine-2-carbonitrile, hereinafter referred to as “Compound 1” or the “compound of the invention”, can form several different crystalline forms as well as in an amorphous form. Accordingly, in a first embodiment (Embodiment 0.1), the invention provides Compound 1, having the formula (1): in an amorphous form.
  • the crystalline form can be, for example, any one of crystalline forms A, B, C and D as defined herein.
  • Compound 1 can be prepared according to the procedure of WO 2015/120390 (Example 64). XRPD analysis of the initially isolated product from this procedure indicates that it is composed of a mixture of two crystalline phases, Form A and Form B. Characterization data suggest Forms A and B are likely anhydrous forms of Compound 1.
  • substantially crystalline refers to forms of the compound of formula (1) in which it is from 50% to 100% crystalline.
  • the compound of formula (1) may be at least 55% crystalline, or at least 60% crystalline, or at least 70% crystalline, or at least 80% crystalline, or at least 90% crystalline, or at least 95% crystalline, or at least 98% crystalline, or at least 99% crystalline, or at least 99.5% crystalline, or at least 99.9% crystalline, for example 100% crystalline.
  • the crystalline form of the Compound 1 is preferably one having a crystalline purity of at least 90%, more preferably at least 95%; i.e. at least 90% (more preferably at least 95%) of the compound is of a single crystalline form (e.g. Form A or Form B).
  • the crystalline forms of the compound of the invention may be solvated (e.g. hydrated) or non-solvated (e.g. anhydrous).
  • anhydrous does not exclude the possibility of the presence of some water on or in the compound (e.g. a crystal of the compound). For example, there may be some water present on the surface of the compound (e.g. compound crystal), or minor amounts within the body of the compound (e.g. crystal).
  • an anhydrous form contains fewer than 0.4 molecules of water per molecule of compound, and more preferably contains fewer than 0.1 molecules of water per molecule of compound, for example 0 molecules of water.
  • the crystalline forms can contain, for example, up to three molecules of water of crystallisation, more usually up to two molecules of water, e.g. one molecule of water or two molecules of water.
  • Non-stoichiometric hydrates may also be formed in which the number of molecules of water present is less than one or is otherwise a non-integer. For example, where there is less than one molecule of water present, there may be 0.4, or 0.5, or 0.6, or 0.7, or 0.8, or 0.9 molecules of water present per molecule of compound (1).
  • the crystalline forms described herein, crystals thereof and their crystal structures form further aspects of the invention.
  • the crystalline forms can be characterised using a number of techniques including, X-ray powder diffraction (XRPD), single crystal X-ray diffraction, differential scanning calorimetry (DSC) and thermogravimetric analysis (TGA).
  • XRPD X-ray powder diffraction
  • DSC differential scanning calorimetry
  • TGA thermogravimetric analysis
  • the behaviour of the crystals under conditions of varying humidity can be analysed by gravimetric vapour sorption studies (such as dynamic vapour sorption (DVS)).
  • XRPD X- ray Powder Diffraction
  • XRPD data for each of the crystalline Forms A, B, C and E are set out below. The relative intensities given should not be strictly interpreted since they may vary depending on the direction of crystal growth, particle sizes and the measurement conditions.
  • the diffraction angles usually mean those that coincide in the range of 2Q ⁇ 0.2°.
  • Form A can be obtained under various experimental conditions, including:
  • the invention provides:
  • Embodiment 1.0 A crystalline form according to Embodiment 1.0 which is of Form A as defined herein.
  • Form A substantially crystalline form of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° ( ⁇ 0.2°).
  • Form A A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three or more, or four or more, five or more, and in particular six diffraction angles (2Q) selected from 10.5°, 16.9°, 17.4°, 20.8°, 26.4° and 28.4° ( ⁇ 0.2°).
  • the X-ray powder diffraction pattern of Form A of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table A-2.
  • the invention provides:
  • Form A substantially crystalline form of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° ( ⁇ 0.2°) (e.g. at least four and more particularly at least five of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 5.2°, 8.5°, 9.8°, 13.9°, 15.8°, 17.1°, 17.7° and 19.4° ( ⁇ 0.2°).
  • Form A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° ( ⁇ 0.2°) (e.g. at least four and more particularly at least five of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 9.8°, 17.1° and 17.7° ( ⁇ 0.2°).
  • Form A substantially crystalline form of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° ( ⁇ 0.2°), and one or more further peaks at diffraction angles (2Q) selected from 9.8°, 17.1° and 17.7° ( ⁇ 0.2°).
  • the invention provides:
  • Crystalline Form A can also be characterised by differential scanning calorimetry (DSC) and has been found to exhibit an endothermic event with an onset temperature of about 224 °C and a peak at about 226 °C as shown in Figure 15 of the accompanying drawings.
  • DSC differential scanning calorimetry
  • Crystalline Form A can also be characterized by Raman Spectroscopy and the Raman spectrum for Form A is shown in Figure 16.
  • the invention provides: 1.16 A substantially crystalline form (Form A) of compound (1) according to any one of Embodiments 1.1 to 1.15 which has a DSC thermogram characterized by an endotherm with an onset at a temperature of about 224 °C and a peak maximum at about 226 °C.
  • composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form A.
  • composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form A.
  • composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form A.
  • 1.23D A composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form A.
  • the invention also provides methods for making crystalline Form A. Accordingly, in further embodiments, the invention provides:
  • Form B can be prepared by slurrying Compound 1 (free base) in THF for 4 weeks at room temperature or 2 weeks at 2-8 °C.
  • the invention provides:
  • Embodiment 1.0 which is of Form B as defined herein.
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5° ( ⁇ 0.2°).
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 7.6° ( ⁇ 0.2°).
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 18.8° ( ⁇ 0.2°).
  • Form B substantially crystalline form of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 21.5° ( ⁇ 0.2°).
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three diffraction angles (2Q) selected from 7.6°, 18.8° and 21.5° ( ⁇ 0.2°).
  • the X-ray powder diffraction pattern of Form B of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table B-2.
  • the invention provides:
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5° ( ⁇ 0.2°) (e.g. at least two and more particularly three of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 12.2°, 14.2°, 15.0°, 15.2°, 22.3°, 27.0° and 29.0° ( ⁇ 0.2°).
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5° ( ⁇ 0.2°) (e.g. at least two and more particularly three of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 15.0°, 15.2° and 29.0° ( ⁇ 0.2°).
  • Form B A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6°, 18.8° and 21.5° ( ⁇ 0.2°), and one or more further peaks at diffraction angles (2Q) selected from 15.0°, 15.2° and 29.0° ( ⁇ 0.2°).
  • the invention provides:
  • the crystalline form of the invention can also be characterised by differential scanning calorimetry (DSC).
  • DSC thermogram of Form B is characterized by endotherms with overlapping peaks at about 222 °C and about 229 °C as shown in Figure 18.
  • Crystalline Form B can also be characterized by Raman Spectroscopy and the Raman spectrum for Form B is shown in Figure 19.
  • the invention provides:
  • 2.23 A substantially crystalline form (Form B) of compound (1) according to any one of Embodiments 2.1 to 2.20 having a crystalline purity of at least 95%.
  • 2.23A A composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form B.
  • composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form B.
  • composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form B.
  • composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form B.
  • Crystalline Form C can be isolated after the addition of water to solutions of Compound 1 in a polar aprotic solvent such as NMP, DMF or DMSO. Based on the experimental data provided herein, Form C is considered to be a solvated/hydrated form of Compound 1 that converts to Form A upon drying and slurrying in water. The results described herein suggest that Form C is likely not a true hydrate or, if it is a hydrate, it is a metastable hydrate.
  • the X-ray diffraction pattern of crystalline Form C of compound (1) exhibits peaks of greatest intensity at the diffraction angles (2Q) set out in Table C-1.
  • the invention provides: 3.1 A crystalline form according to Embodiment 1.0 which is of Form C as defined herein.
  • Form C A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at about the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0° ( ⁇ 0.2°).
  • Form C A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 8.7° ( ⁇ 0.2°).
  • Form C A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 26.5° ( ⁇ 0.2°).
  • Form C A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 27.0° ( ⁇ 0.2°).
  • Form C A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three or more, or four or more, and in particular five diffraction angles (2Q) selected from 5.8°, 8.7°, 26.5° and 27.0° ( ⁇ 0.2°).
  • the X-ray powder diffraction pattern of Form C of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table C-2.
  • the invention provides: 3.8 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0° ( ⁇ 0.2°) (e.g. at least three and more particularly four of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 13.8° and 16.3° ( ⁇ 0.2°).
  • the invention provides:
  • the crystalline form of the invention can also be characterised by differential scanning calorimetry (DSC).
  • DSC thermogram of Form C is characterized by endotherms as shown in Figure 24. Accordingly, the invention also provides:
  • Form C A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.11 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 62°C and a peak maximum at about 67°C.
  • Form C A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.13 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 221 °C and a peak maximum at about 225°C.
  • the crystalline form of the invention can also be characterised by thermogravimetric analysis (TGA).
  • TGA thermogravimetric analysis
  • the TGA plot of Form C is shows a weight loss of 37.2% wt from 49°C to 135°C, as shown in Figure 24. Accordingly, the invention also provides:
  • composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 2.20, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form C.
  • composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 2.20, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form C.
  • composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 3.20, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form C.
  • composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 3.20, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form C.
  • Form D Crystalline Form D of the compound (1) can be formed Crystalline Form D from evaporation of a solution of Compound 1 in acetic acid. Form D is considered to be an acetate solvate of Compound 1. 1 H NMR analysis indicates the presence of 2 moles acetic acid/acetate per mole Compound 1 in the sample, consistent with a 1:2 Compound 1 acetate salt/solvate.
  • the X-ray diffraction pattern of crystalline Form D of compound (1) exhibits peaks of greatest intensity at the diffraction angles (2Q) set out in Table D-1.
  • the invention provides:
  • Embodiment 1.0 A crystalline form according to Embodiment 1.0 which is of Form D as defined herein.
  • Form D A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9°( ⁇ 0.2°).
  • Form D A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 10.7° ( ⁇ 0.2°).
  • Form D A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 16.6° ( ⁇ 0.2°).
  • Form D A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 25.9° ( ⁇ 0.2°).
  • Form D A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three or more, or four or more, and in particular five diffraction angles (2Q) selected from 10.7°, 16.6° and 25.9° ( ⁇ 0.2°).
  • the X-ray powder diffraction pattern of Form D of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table D-2.
  • the invention provides:
  • Form D A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9° ( ⁇ 0.2°), and one or more further peaks at diffraction angles (2Q) selected from 6.5°, 17.7°, 18.9°, 26.1° and 26.9° ( ⁇ 0.2°).
  • the invention provides:
  • Crystalline Form D can also be characterised by differential scanning calorimetry (DSC) and has a thermogram substantially as shown in Figure 12.
  • the crystalline form of the invention can also be characterised by differential scanning calorimetry (DSC).
  • DSC thermogram of Form D is characterized by endotherms as shown in Figure 26. Accordingly, the invention also provides:
  • Form D A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.12 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 62°C and a peak maximum at about 86°C.
  • Form D A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.13 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 161°C and a peak maximum at about 170°C.
  • the crystalline form of the invention can also be characterised by thermogravimetric analysis (TGA).
  • TGA thermogravimetric analysis
  • the TGA plot of Form D is shows a weight loss of 8.2% wt from 40°C to 114°C, as shown in Figure 26. Accordingly, the invention also provides:
  • Form D A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.15 which has a TGA plot characterized by a weight loss of about 8.2% wt between 40°C and 114°C.
  • composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form D.
  • composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form D.
  • composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form D.
  • the crystalline forms of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or the amorphous form as defined in Embodiment 0.1 may contain one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element.
  • a reference to hydrogen includes within its scope 1 H, 2 H (D), and 3 H (T).
  • references to carbon and oxygen include within their scope respectively 12 C, 13 C and 14 C and 16 0 and 18 0.
  • the isotopes may be radioactive or non-radioactive.
  • the crystalline/amorphous form of the compound of formula (1) as defined in any one of Embodiments 0.1 to 4.20 contains no radioactive isotopes. Such compounds are preferred for therapeutic use.
  • the crystalline/amorphous form of the compound of formula (1) as defined in any one of Embodiments 0.1 to 4.20 may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.
  • Bioloqical Activity Crystalline/amorphous forms of the compound of formula (1) are potent inhibitors of Chk-1 and consequently are expected to be beneficial alone or in combination with various chemotherapeutic agents or radiation for treating a wide spectrum of proliferative disorders.
  • the invention provides:
  • Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use as a Chk-1 kinase inhibitor.
  • a method for the prophylaxis or treatment of a proliferative disease such as cancer comprises administering to a patient in combination with radiotherapy or chemotherapy a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
  • a method for the prophylaxis or treatment of a proliferative disease such as cancer comprises administering to a patient a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
  • carcinomas for example carcinomas of the bladder, breast, colon, kidney, epidermis, liver, lung, oesophagus, gall bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, gastrointestinal system, or skin, hematopoieitic tumours such as leukaemia, B-cell lymphoma, T-cell lymphoma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, hairy cell lymphoma, or Burkett's lymphoma; hematopoieitic tumours of myeloid lineage, for example acute and chronic myelogenous leukaemias, myelodysplastic syndrome, or promyelocytic leukaemia; thyroid follicular cancer; tumours of mesenchymal origin, for example fibrosarcoma or habdomy
  • the Chk-1 inhibitors of the invention may be useful in treating tumours in which there is a defective DNA repair mechanism or a defective cell cycle, for example a cancer in which mutations (e.g. in p53) have led to the G1/S DNA damage checkpoint being lost (see the introductory section of this application).
  • the Chk-1 inhibitors of the invention may also be useful in treating RAD17 mutant tumours, ATM-deficient RAD50-mutant tumours and fanconi anaemia. Accordingly in further embodiments (Embodiments 6.11 to 6.24), the invention provides:
  • MYC oncogene-driven cancer is a B-cell lymphoma, leukemia, neuroblastoma, breast cancer or lung cancer.
  • a p53 negative or mutated tumour e.g. a cancer selected from breast cancer, colon cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, glioma, and leukemia
  • a chemotherapeutic agent selected from cytarabine, etoposide, gemcitabine and SN-38.
  • Embodiment 6.18 The use according to Embodiment 6.17 wherein the cancer is a p53 negative or mutated tumour.
  • a method for the treatment of a patient e.g. a human patient suffering from a cancer which is characterised by a defective DNA repair mechanism or defective cell cycle, which method comprises administering to the patient a therapeutically effective amount of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24. 6.20 A method according to Embodiment 6.19 wherein the cancer is a p53 negative or mutated tumour.
  • Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in the treatment of Fanconi anaemia.
  • a method of treating Fanconi anaemia in a subject comprises administering to the subject a therapeutically effective amount of crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
  • Chk-1 inhibitor crystalline/amorphous forms of the compound of the invention may be used alone or they may be used in combination with DNA-damaging anti-cancer drugs and/or radiation therapy to treat subjects with multi-drug resistant cancers.
  • a cancer is considered to be resistant to a drug when it resumes a normal rate of tumour growth while undergoing treatment with the drug after the tumour had initially responded to the drug.
  • a tumour is considered to "respond to a drug" when it exhibits a decrease in tumor mass or a decrease in the rate of tumour growth.
  • Compound 1 can be prepared as described WO 2015/120390 (see Example 64).
  • the active compound While it is possible for the active compound to be administered alone, it is preferable to present it as a pharmaceutical composition (e.g. formulation) comprising at least one active compound of the invention together with one or more pharmaceutically acceptable excipients such as carriers, adjuvants, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art, and optionally other therapeutic or prophylactic agents.
  • a pharmaceutical composition e.g. formulation
  • pharmaceutically acceptable excipients such as carriers, adjuvants, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art, and optionally other therapeutic or prophylactic agents.
  • pharmaceutically acceptable refers to 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 a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problems or complication, commensurate with a reasonable benefit/risk ratio.
  • a subject e.g. human
  • Each excipient must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
  • the invention provides:
  • a pharmaceutical composition comprising a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-
  • a pharmaceutical composition according to Embodiment 7.1 which comprises from approximately 1% (w/w) to approximately 95% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-
  • a pharmaceutical composition according to Embodiment 7.2 which comprises from approximately 5% (w/w) to approximately 90% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-
  • a pharmaceutical composition according to Embodiment 7.3 which comprises from approximately 10% (w/w) to approximately 90% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1), or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, as defined in Embodiment 0.1 and from 90% (w/w) to 10% of a pharmaceutically excipient or combination of excipients.
  • a pharmaceutical composition according to Embodiment 7.4 which comprises from approximately 20% (w/w) to approximately 90% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21- 4.24, and from 80% (w/w) to 10% of a pharmaceutically excipient or combination of excipients.
  • a pharmaceutical composition according to Embodiment 4.5 which comprises from approximately 25% (w/w) to approximately 80% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21- 4.24, and from 75% (w/w) to 20% of a pharmaceutically excipient or combination of excipients.
  • compositions comprising a crystalline form of the compound of formula (1) will typically be solid compositions such as tablets, capsules, caplets, pills, lozenges, sprays, powders, granules, sublingual tablets, wafers or patches and buccal patches, or liquid compositions such as suspensions where the active compound is in solid form.
  • the invention provides:
  • Embodiment 7.8 A pharmaceutical composition according to Embodiment 7.7 which is selected from tablets, capsules, caplets, pills, lozenges, powders, granules, suspensions, sublingual tablets, wafers or patches and buccal patches. 7.9 A pharmaceutical composition according to Embodiment 7.8 which is selected from tablets and capsules.
  • compositions containing a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, can be formulated in accordance with known techniques, see for example, Remington’s Pharmaceutical Sciences, Mack Publishing Company, Easton, PA, USA.
  • tablet compositions can contain a unit dosage of active compound together with an inert diluent or carrier such as a sugar or sugar alcohol, e.g.; lactose, sucrose, sorbitol or mannitol; and/or a non-sugar derived diluent such as sodium carbonate, calcium phosphate, talc, calcium carbonate, or a cellulose or derivative thereof such as methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch.
  • Tablets may also contain such standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants (e.g.
  • swellable crosslinked polymers such as crosslinked carboxymethylcellulose
  • lubricating agents e.g. stearates
  • preservatives e.g. parabens
  • antioxidants e.g. BHT
  • buffering agents for example phosphate or citrate buffers
  • effervescent agents such as citrate/bicarbonate mixtures.
  • Capsule formulations may be of the hard gelatin or soft gelatin variety and can contain the active component in solid, semi-solid, or liquid form.
  • Gelatin capsules can be formed from animal gelatin or synthetic or plant derived equivalents thereof.
  • the solid dosage forms can be coated or un-coated, but typically have a coating, for example a protective film coating (e.g. a wax or varnish) or a release controlling coating.
  • a protective film coating e.g. a wax or varnish
  • the coating e.g. a Eudragit TM type polymer
  • the coating can be designed to release the active component at a desired location within the gastro-intestinal tract.
  • the coating can be selected so as to degrade under certain pH conditions within the gastrointestinal tract, thereby selectively release the compound in the stomach or in the ileum or duodenum.
  • the drug can be presented in a solid matrix comprising a release controlling agent, for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract.
  • a release controlling agent for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract.
  • the matrix material or release retarding coating can take the form of an erodible polymer (e.g. a maleic anhydride polymer) which is substantially continuously eroded as the dosage form passes through the gastrointestinal tract.
  • compositions for topical use include ointments, creams, sprays, patches, gels, liquid drops and inserts (for example intraocular inserts). Such compositions can be formulated in accordance with known methods.
  • compositions for parenteral administration are typically presented as sterile aqueous or oily fine suspensions, or may be provided in finely divided sterile powder form for making up extemporaneously with sterile water for injection.
  • formulations for rectal or intra-vaginal administration include pessaries and suppositories which may be, for example, formed from a shaped mouldable or waxy material containing the active compound.
  • compositions for administration by inhalation may take the form of inhalable powder compositions or powder sprays, and can be administrated in standard form using powder inhaler devices or aerosol dispensing devices. Such devices are well known.
  • the powdered formulations typically comprise the active compound together with an inert solid powdered diluent such as lactose.
  • crystalline forms of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1 will generally be presented in unit dosage form and, as such, will typically contain sufficient compound to provide a desired level of biological activity.
  • a composition intended for oral administration may contain from 2 milligrams to 200 milligrams of active ingredient, more usually from 10 milligrams to 100 milligrams, for example, 12.5 milligrams, 25 milligrams and 50 milligrams.
  • Forms of Compound 1, whether administered alone, or in combination with DNA damaging agents and other anti-cancer agents and therapies, are generally administered to a subject in need of such administration, for example a human or animal patient, preferably a human.
  • another chemotherapeutic agent for example an anticancer drug.
  • chemotherapeutic agents that may be co-administered with the crystalline/amorphous forms of Compound 1 or compositions of matter as defined in any one of Embodiments 0.1 to 4.24 include:
  • hypoxia triggered DNA damaging agents e.g. Tirapazamine, TH-302
  • chemotherapeutic agents that may be administered in combination with the crystalline/amorphous forms of Compound 1 as defined in any one of Embodiments 0.1 to 4.20 include: nitrogen mustards such as mechlorethamine, cyclophosphamide, ifosfamide, melphalan and chlorambucil; nitrosoureas such as carmustine, lomustine and semustine; ethyleneimine/methylmelamine compounds such as triethylenemelamine, triethylene thiophosphoramide and hexamethylmelamine; alkyl sulphonates such as busulfan; triazines such as dacarbazine
  • Antimetabolites such as folates, methotrexate, trimetrexate, 5-fluorouracil, fluorodeoxyuridine, gemcitabine, cytosine arabinoside, 5-azacytidine, 2, 2'- difluorodeoxycytidine, 6-mercaptopurine, 6-thioguanine, azathioprine, 2'-deoxycoformycin, erythrohydroxynonyl-adenine, fludarabine phosphate and 2-chlorodeoxyadenosine; type I topoisomerase inhibitors such as camptothecin, topotecan and irinotecan; type II topoisomerase inhibitors such as the epipodophylotoxins (e.g.
  • antimitotic drugs such as paclitaxel, Taxotere, Vinca alkaloids (e.g. vinblastine, vincristine, vinorelbine) and estramustine (e.g.
  • estramustine phosphate antibiotics such as actimomycin D, daunomycin (rubidomycin), doxorubicin (adriamycin), mitoxantrone, idarubicine, bleomycin, mithramycin, mitomycin C and dactinomycin enzymes such as L-asparaginase; cytokines and biological response modifiers such as interferon (a, b,g), interleukin-2G-CSF and GM-CSF: retinoids such as retinoic acid derivatives (e.g.
  • radiosensitisers such as metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, nicotinamide, 5-bromodeoxyuridine, 5- iododeoxyuridine and bromodeoxycytidine; platinum compounds such as cisplatin, carboplatin, spiroplatin, iproplatin, onnaplatin, tetraplatin and oxaliplatin; anthracenediones such as mitoxantrone; ureas such as hydroxyurea; hydrazine derivatives such as N-methylhydrazine and procarbazine; adrenocortical suppressants such as mitotane and aminoglutethimide; adrenocorticosteroids and antagonists such as prednisone, dexamethasone and aminoglutethimide; progestins such as hydroxy
  • hydroxyprogesterone caproate medroxyprogesterone (e.g. medroxyprogesterone acetate) and megestrol (e.g. megestrol acetate); oestrogens such as diethylstilbestrol and ethynyl estradiol; anti-oestrogens such as tamoxifen; androgens such as testosterone (e.g. testosterone propionate) and fluoxymesterone; anti-androgens such as flutamide and leuprolide; nonsteroidal anti-androgens such as flutamide; and signal transduction inhibitors such as PARP inhibitors [e.g. as disclosed in Cancer Res.
  • chemotherapeutic agents examples include the chemotherapeutic agents described in Blasina etal., Mol. Cancer Ther., 2008, 7(8), 2394- 2404, Ashwell et al., Clin. Cancer Res., 2008, 14(13), 4032-4037, Ashwell et al., Expert Opin. Investig. Drugs, 2008, 17(9), 1331-1340, Trends in Molecular Medicine February 2011, Vol. 17, No. 2 and Clin Cancer Res; 16(2) January 15, 2010.
  • chemotherapeutic agents that may be used in combination with the Chk-1 inhibitor compounds of the invention as defined herein include antimetabolites (such as capecitabine, cytarabine, fludarabine, gemcitabine and pemetrexed), Topoisomerase-I inhibitors (such as SN38, topotecan, irinotecan), platinum compounds (such as carboplatin, oxaloplatin and cisplatin), Topoisomerase-I I inhibitors (such as daunorubicin, doxorubicin and etoposide), thymidylate synthase inhibitors (such as 5-fluoruracil), mitotic inhibitors (such as docetaxel, paclitaxel, vincristine and vinorelbine, ) and alkylating agents (such as mitomycin C).
  • antimetabolites such as capecitabine, cytarabine, fludarabine, gemcitabine and pemetrexed
  • a further set of chemotherapeutic agents that may be used in combination with the Chk-1 inhibitor compounds of the invention as defined herein includes agents that induce stalled replication forks (see Ashwell et al., Clin. Cancer Res., above), and examples of such compounds include gemcitabine, 5-fluorouracil and hydroxyurea.
  • the compounds of the invention and combinations with chemotherapeutic agents or radiation therapies as described above may be administered over a prolonged term to maintain beneficial therapeutic effects or may be administered for a short period only. Alternatively they may be administered in a pulsatile or continuous manner.
  • the compounds of the invention will be administered in an effective amount, i.e. an amount which is effective to bring about the desired therapeutic effect either alone (in monotherapy) or in combination with one or more chemotherapeutic agents or radiation therapy.
  • an effective amount can be a quantity of compound which, when administered alone or together with a DNA-damaging drug or other anti-cancer drug to a subject suffering from cancer, slows tumour growth, ameliorates the symptoms of the disease and/or increases longevity.
  • an effective amount of the Chk-1 inhibitor of the invention is the quantity in which a greater response is achieved when the Chk-1 inhibitor is co-administered with the DNA damaging anti-cancer drug and/or radiation therapy compared with when the DNA damaging anti-cancer drug and/or radiation therapy is administered alone.
  • an "effective amount" of the DNA damaging drug and/or an "effective” radiation dose are administered to the subject, which is a quantity in which anti-cancer effects are normally achieved.
  • the Chk-1 inhibitors of the invention and the DNA damaging anti-cancer drug can be co-administered to the subject as part of the same pharmaceutical composition or, alternatively, as separate pharmaceutical compositions.
  • the Chk-1 inhibitor of the invention and the DNA-damaging anti-cancer drug (and/or radiation therapy) can be administered simultaneously or at different times, provided that the enhancing effect of the Chk-1 inhibitor is retained.
  • a compound of the invention as defined herein is administered before (e.g by up to 8 hours or up to 12 hours or up to one day before) administration of the DNA- damaging anticancer drug.
  • a compound of the inventin as defined herein is administered after (e.g by up to 8 hours or up to 12 hours or up to 24 hours or up to 30 hours or up to 48 hours after) administration of the DNA-damaging anticancer drug.
  • a first dose of a compound of the invention as described herein is administered one day after administration of the DNA-damaging anticancer drug and a second dose of the said compound is administered two days after administration of the DNA-damaging anticancer drug.
  • a first dose of a compound of the invention as defined herein is administered one day after administration of the DNA-damaging anticancer drug, a second dose of the said compound is administered two days after administration of the DNA- damaging anticancer drug, and third dose of the said compound is administered three days after administration of the DNA-damaging anticancer drug.
  • Particular dosage regimes comprising the administration of a compound of the invention as defined herein and a DNA-damaging anticancer drug may be as set out in WO2010/118390 (Array Biopharma), the contents of which are incorporated herein by reference.
  • Chk-1 inhibitor compound of the invention and (in the case of combination therapy) the DNA damaging anti-cancer drug and radiation dose administered to the subject will depend on the nature and potency of the DNA damaging anti-cancer drug, the type and severity of the disease or condition and on the characteristics of the subject, such as general health, age, sex, body weight and tolerance to drugs. The skilled person will be able to determine appropriate dosages depending on these and other factors. Effective dosages for commonly used anti-cancer drugs and radiation therapy are well known to the skilled person.
  • a typical daily dose of Compound 1 can be in the range from 100 picograms to 100 milligrams per kilogram of body weight, more typically 5 nanograms to 25 milligrams per kilogram of bodyweight, and more usually 10 nanograms to 15 milligrams per kilogram (e.g. 10 nanograms to 10 milligrams, and more typically 1 microgram per kilogram to 20 milligrams per kilogram, for example 1 microgram to 10 milligrams per kilogram) per kilogram of bodyweight although higher or lower doses may be administered where required.
  • the compound can be administered on a daily basis or on a repeat basis every 2, or 3, or 4, or 5, or 6, or 7, or 10 or 14, or 21, or 28 days for example.
  • the quantity of compound administered and the type of composition used will be commensurate with the nature of the disease or physiological condition being treated and will be at the discretion of the physician.
  • a patient Prior to administration of a compound of the invention, a patient may be screened to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Chk1.
  • the invention provides:
  • a method for the diagnosis and treatment of a disease state or condition mediated by Chk1 kinase comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against the kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient an effective Chk1 inhibiting amount of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
  • a subject e.g. patient
  • a diagnostic test to detect a marker indicative of the presence of a disease or condition in which Chk1 is implicated, or a marker indicative of susceptibility to the said disease or condition.
  • subjects may be screened for genetic markers indicative of a susceptibility to develop an autoimmune or inflammatory disease.
  • a patient may be screened to determine whether a cancer from which the patient is or may be suffering is one which is characterised by a defective DNA repair mechanism or a defective cell cycle, for example a defective cell cycle due to a p53 mutation or is a p53 negative cancer.
  • Cancers which are characterised by p53 mutations or the absence of p53 can be identified, for example, by the methods described in Allred et al., J. Nat. Cancer Institute, Vol. 85, No.
  • p53 protein may be detected by immuno-histochemical methods such as immuno-staining.
  • the diagnostic tests are typically conducted on a biological sample selected from tumour biopsy samples, blood samples (isolation and enrichment of shed tumour cells), stool biopsies, sputum, chromosome analysis, pleural fluid, peritoneal fluid, or urine.
  • mutations to other DNA repair factors such as RAD17, RAD50, and members of the Fanconi’s anaemia complementation group may be predictive of response to Chk1 inhibitors alone, or in combination with chemotherapy.
  • Cancers which contain mutations in these DNA repair pathways may be identified by DNA sequence analysis of tumor biopsy tissue or circulating tumor DNA (ctDNA) or, in the case of Fanconi’s anaemia, by evaluating DNA foci formation in tumor biopsy specimens using an antibody to FANCD2, as described in Duan et al., Frontiers in Oncology vol.4, 1-8 (2014).
  • the crystalline forms of the compound of the invention described herein may be used to treat members of a sub-population of patients who have been screened (for example by testing one or more biological samples taken from the said patients) and have been found to be suffering from a cancer characterised by p53 mutation or a p53 negative cancer, or a cancer containing a RAD17 or RAD50 mutation, or a mutation in a member of the Fanconi’s anaemia complementation group.
  • Figure 1 shows the chemical structure of Compound 1.
  • Figure 2 shows a comparison of XRPD Patterns of Compound 1 (free base) with Forms A and B.
  • Figure 3 shows a Thermal Analysis of Compound 1 (free base).
  • Figures 4A and 4B are Hot Stage Microscopy Images for Compound 1.
  • Figure 5 shows a comparison of XRPD Patterns of Compound 1 (free base) with Forms A
  • Figure 6 is an overlay of XRPD Pattern of Solids Isolated from 1,4-Dioxane.
  • Figure 7 shows the XRPD Patterns from THF Slurries at 2-8 °C, RT and 50 °C.
  • Figure 8 shows the XRPD Pattern for Form A of Compound 1 collected with Cu-Ka radiation.
  • Figure 8A shows the XPRD Pattern for Form A of Compound 1 but with the main peaks labelled.
  • Figure 9 is an atomic displacement ellipsoid diagram of Form A in which non-hydrogen atoms are represented by 50% probability anisotropic thermal ellipsoids.
  • Figure 10 is a packing diagram of Compound 1 Form A viewed down the crystallographic a axis.
  • Figure 11 is a packing diagram of Compound 1 Form A viewed down the crystallographic b axis.
  • Figure 12 is a packing diagram of Compound 1 Form A viewed down the crystallographic c axis.
  • Figure 13 indicates hydrogen bonding between two molecules of Compound 1 observed for Form A.
  • Figure 14 is an overlay of Calculated Pattern and Experimental Pattern for Form A.
  • Figure 15 shows a thermal analysis of Compound 1 Form A.
  • Figure 16 shows the Raman Spectrum for Compound 1 Form A.
  • Figure 17 shows the XRPD Pattern for Form B of Compound 1 collected with Cu-Ka radiation.
  • Figure 17A shows the XRPD pattern for Form B of Compound 1 but with observed peaks collected with Cu-Ka labelled.
  • Figure 18 shows a thermal analysis of Compound 1 Form B.
  • Figure 19 shows the Raman Spectrum for Compound 1 Form B.
  • Figure 20 is an overlay of the XRPD Patterns of Form A and Form B.
  • Figure 21 is a comparison of the Raman Spectra of Form A and Form B.
  • Figure 22 is an overlay of the XRPD Patterns of Form C isolated from different solvent systems.
  • Figure 23 shows the XRPD Pattern for Form C of Compound 1 collected with Cu-Ka radiation.
  • Figure 24 shows a thermal analysis of Form C (Isolated from NMP/H2O)
  • Figure 25 shows the XRPD Pattern for Form C of Compound 1 collected with Cu-Ka radiation.
  • Figure 26 shows a thermal analysis of Form D
  • Figure 27 shows the XRPD Pattern for amorphous Compound 1.
  • Figure 28 shows a thermal analysis for amorphous Compound 1.
  • Solubility was estimated based on the total volume of solvent used to provide complete dissolution. The actual solubility may be greater than the value calculated due to the incremental addition of solvent and kinetics of dissolution of the material. The solubility is expressed as “less than” if dissolution did not occur during the experiment. The solubility is expressed as “greater than” if dissolution occurred after the addition of first aliquot.
  • a solution of Compound 1 (free base) was prepared in a given solvent. The solution was filtered into a clean vial and an anti-solvent was added in order to precipitate solids. Conversely, a solution of Compound 1 (free base) was filtered directly into an anti-solvent solution in selected experiments.
  • Solutions of Compound 1 (free base) were prepared at elevated temperatures in given solvent or solvent mixture. The resulting solution was hot filtered into a pre-warmed vial. The vial was capped and placed on a bench top at room temperature to quickly cool.
  • Solutions of Compound 1 (free base) were generated at ambient temperature in a given solvent or solvent mixture. The solutions were allowed to evaporate partially or to dryness from an uncapped vial at ambient conditions.
  • a solid sample of Compound 1 (free base) was placed in an agate jar with a ⁇ 4mm agate ball. 20 pL of specified solvent was added and then the sample placed in a Retsch mill at 30 Hz for 10 minutes. The walls of the agate jar were scraped and 20 pL of designated solvent added. Sample was ground in the Retsch mill for another 10 minutes at 30 Hz. Solids were collected.
  • a solution of Compound 1 (free base) was prepared at elevated temperature in a given solvent or solvent mixture.
  • the resulting solution was hot filtered in a pre-warmed vial.
  • the vial was capped and left in a heating block at elevated temperature. The heater then was turned off for the sample to cool down naturally to room temperature.
  • Solutions of Compound 1 (free base) were generated at ambient temperature in a given solvent or solvent mixture. The solutions were filtered and allowed to evaporate partially from a loosely capped vial at ambient conditions.
  • Hot stage microscopy was performed using a Linkam hot stage (FTIR 600) mounted on a Leica DM LP microscope equipped with a SPOT InsightTM color digital camera. Temperature calibrations were performed using USP melting point standards. Samples were placed on a cover glass, and a second cover glass was placed on top of the sample. As the stage was heated, each sample was visually observed using a 20x objective with crossed polarizers and a first order red compensator. Images were captured using SPOT software (v. 4.5.9).
  • Solution 1 H NMR spectra were acquired with an Avance 600 MHz NMR Spectrometer. Samples were prepared by dissolving in DMSO-d 6 containing TMS. The data acquisition parameters are displayed on the first page of the spectrum in the Data section of this report.
  • Raman spectrum were acquired on a FT-Raman module interfaced to a Nexus 670 FT-IR spectrophotometer (Thermo Nicolet) equipped with an indium gallium arsenide (InGaAs) detector. Wavelength verification was performed using sulfur and cyclohexane.
  • the sample was prepared for analysis by placing the sample into a glass tube and positioning the tube in a gold- coated tube holder. Approximately 0.3 W of Nd:YVC>4 laser power (1064 nm excitation wavelength) was used to irradiate the sample. The spectrum represents 256 co-added scans collected at a spectral resolution of 4 cm -1 .
  • the structure was solved by direct methods using SHELXT [6] The remaining atoms were located in succeeding difference Fourier syntheses.
  • the structure was refined using SHELXL- 2014 [7, 6] Hydrogen atoms were refined independently.
  • the structure was refined in full- matrix least-squares by minimizing the function:
  • the atomic displacement ellipsoid diagram was prepared using MERCURY. Atoms are represented by 50% probability anisotropic thermal ellipsoids.
  • TGA/DSC Thermogravimetric Analysis/Differential Scanning Calorimetry (TGA/DSC) TGA/DSC analysis was performed using a Mettler-Toledo TGA/DSC3+ analyzer. Temperature and enthalpy adjustments were performed using indium, tin, and zinc, and then verified with indium. The balance was verified with calcium oxalate.
  • the sample was placed in an open aluminum pan. The pan was hermetically sealed, the lid pierced, then inserted into the TG furnace. A weighed aluminum pan configured as the sample pan was placed on the reference platform. The furnace was heated under nitrogen. The data acquisition parameters for the thermogram are displayed in the image in the Data section of this report.
  • XRPD patterns were collected with a PANalytical X'Pert PRO MPD or PANalytical Empyrean diffractometer using an incident beam of Cu radiation produced using a long, fine-focus source.
  • An elliptically graded multilayer mirror was used to focus Cu Ka X-rays through the specimen and onto the detector.
  • a silicon specimen NIST SRM 640e was analyzed to verify the observed position of the Si 111 peak is consistent with the NIST- certified position.
  • a specimen of the sample was sandwiched between 3-pm-thick films and analyzed in transmission geometry. A beam-stop, short antiscatter extension, and antiscatter knife edge were used to minimize the background generated by air.
  • Soller slits for the incident and diffracted beams were used to minimize broadening and asymmetry from axial divergence. Diffraction patterns were collected using a scanning position-sensitive detector (X'Celerator) located 240 mm from the specimen and Data Collector software v. 5.5.
  • X'Celerator scanning position-sensitive detector
  • Indexing is the process of determining the size and shape of the crystallographic unit cell given the peak positions in a diffraction pattern. The term gets its name from the assignment of Miller index labels to individual peaks.
  • XRPD indexing serves several purposes. If all of the peaks in a pattern are indexed by a single unit cell, this is strong evidence that the sample contains a single crystalline phase. Given the indexing solution, the unit cell volume may be calculated directly and can be useful to determine their solvation states. Indexing is also a robust description of a crystalline form and provides a concise summary of all available peak positions for that phase at a particular thermodynamic state point. Indexing of XRPD pattern was done using proprietary SSCI software. Space groups consistent with the assigned extinction symbol, unit cell parameters, and derived quantities are tabulated in the respective figures providing the indexing solution for each form.
  • XRPD patterns were indexed using proprietary software 1 or X'Pert High-Score Plus 2.2a (2.2.1). Indexing and structure refinement are computational studies. Agreement between the allowed peak positions, marked with red bars, and the observed peaks indicates a consistent unit cell determination. Successful indexing of the pattern indicates that the sample is composed primarily of a single crystalline phase. Space groups consistent with the assigned extinction symbol, unit cell parameters, and derived quantities are tabulated below each figure showing tentative indexing solution. To confirm the tentative indexing solution, the molecular packing motifs within the crystallographic unit cells must be determined. No attempts at molecular packing were performed.
  • Solids that display unique XRPD patterns in comparison to the starting material or are of unknown composition or crystalline phase content are designated as "Material” followed by a sequential capital letter (e.g. Material A, Material B, etc.) applied sequentially.
  • a solid is designated as a "Form” (e.g. Compound 1 Form A) only if its chemical composition is known (e.g. 1 H NMR) and it is demonstrated to be a single crystalline phase (e.g. indexing or single crystal data).
  • Compound 1 (free base) is composed of a mixture of two crystalline phases, Form A and Form B ( Figure 2). Form A and Form B were isolated as single crystalline phases during the course of this study.
  • the 1 H NMR spectrum of Compound 1 (free base) is overall consistent with the structure of Compound 1. Water is observed in the spectrum based on the peak at 3.3 ppm. The water content was determined by KF and found to be 0.2% which suggests the loss observed by TGA is likely due to unbound water on the sample.
  • Solubility estimates were conducted using an incremental solvent addition method to aid in experimental design. Solubility estimates are presented in Table 2. Table 2 - Approximate Kinetic Solubility Estimates of Compound 1 a Solubilities are calculated based on the total solvent used to give a solution; actual solubilities may be greater because of the volume of the solvent portions utilized or a slow rate of dissolution. Solubilities are rounded to the nearest mg/mL.
  • Compound 1 (free base) exhibited low solubility ( ⁇ 1 mg/mL) at RT in the majority of solvents tested. Intermediate to good solubility was observed in THF, acetic acid, formic acid, dimethyl acetamide (DMA), dimethyl formamide (DMF), dimethyl sulfoxide (DMSO), N-Methyl-2- pyrrolidone (NMP), 1,1,1,3,3,3-hexafluoro-2- propanol (HFIPA) and 2,2,2-trifluoroethanol (TFE).
  • DMA dimethyl acetamide
  • DMF dimethyl formamide
  • DMSO dimethyl sulfoxide
  • NMP N-Methyl-2- pyrrolidone
  • HFIPA 1,1,1,3,3,3-hexafluoro-2- propanol
  • TFE 2,2,2-trifluoroethanol
  • a preliminary polymorph and stable form screen of Compound 1 was conducted in an effort to determine the stable form at RT and to evaluate the propensity of the compound to exist in various crystalline forms, including polymorphs, solvates, and hydrates.
  • Approximately thirty-six (36) experiments were performed using different crystallization techniques to vary conditions of nucleation and growth investigating both kinetic and thermodynamic conditions. Crystallization techniques employed included cooling, evaporation, relative humidity stressing, solvent/antisolvent addition, slurry, or a combination of techniques (Table 3 and Table 4).
  • Solids isolated from experiments were typically inspected by polarized light microscopy (PLM) and analyzed by XRPD. XRPD patterns were compared with known forms of Compound 1 and with each other.
  • PLM polarized light microscopy
  • Table 4 Stable Form Screen of Compound 1 a. Approx. 70 mg starting material and 1 mL solvent used for each experiment, unless otherwise indicated b. Samples were initially heated to 50 °C for ⁇ 3 hours prior to slurrying at RT for the stated time b. Times are approximate c. Approx. 100 mg starting material used
  • Form A as a single crystalline phase was observed from a number of polymorph screening experiments.
  • Form B was only observed as a single crystalline phase during the stable form screen from slurries in THF at RT and 2-8 °C.
  • Form A and Form B are crystalline, anhydrous forms of Compound 1 that are likely enantiotropically related.
  • Form C is likely solvated/hydrated and converts to Form A on drying.
  • Form D is a potential diacetate solvate/salt based on pKa considerations (the calculated pKas for Compound 1 are 11.99 and 7.58 ⁇ 0.1 (ACD11)[1], and the pKa for acetic acid is 4.756) and 1 H NMR data.
  • Compound 1 (free base), which is composed of a physical mixture of Form A and Form B, was used as the starting material for slurry experiments. Samples were slurried in various solvents and solvent mixtures at RT in an effort to identify the stable form at RT and evaluate the tendency to form hydrates. The rate of transformation between forms tends to be related to the solubility in that solvent system such that faster transformation kinetics are typically observed in higher solubility solvent systems. Given the low solubility of Compound 1 (free base) observed in the majority of solvents, longer slurry times were used in an effort to overcome potentially slow transformation rates.
  • Form B at RT was unclear possibly due to the similarity in energy of the two forms at room temperature.
  • Form B Compound 1 (free base) was slurried in THF at 2-8 °C and at 50 °C for 2 weeks. Solids isolated after slurrying in THF at 2-8 °C were consistent with Form B, whereas solids isolated from the 50 °C slurry were composed of Form A based on XRPD ( Figure 7). Results suggest Form A and Form B are likely enantiotropically related with Form B likely the stable form at 2-8 °C and Form A likely the stable form at 50 °C. Further investigation would be needed to confirm the thermodynamic relationship and the transition temperature or transition temperature range between the two forms.
  • Form A was obtained under various experimental conditions, including evaporation of an acetonitrile solution, precipitation upon stirring solutions produced by the addition of antisolvents to solutions of Compound 1 (free base), specifically DMA/MTBE, HFIPA/toluene and TFE/IPA solvent/antisolvent additions (Table 3).
  • Form A was also prepared by rotary evaporation of a DMSO/acetone solution and after slurrying Compound (free base) in THF at 50 °C for 2 weeks.
  • Form A, prepared from DMA/MTBE was characterized by XRPD with indexing, 1H NMR and TGA/DSC analyses (Table 5).
  • Form B was prepared by slurrying Compound 1 (free base) in THF for 4 weeks at RT or 2 weeks at 2-8 °C.
  • the XRPD pattern of Form B was successfully indexed ( Figure 17) indicating the sample is composed primarily or exclusively of a single crystalline phase.
  • the indexed unit cell volume 992.8 A3/cell suggests the material is anhydrous/unsolvated based on considerations of molecular volume.
  • the TGA thermogram exhibited a weight loss of 0.1% upon heating the sample to 151 °C (Figure 18). Overlapping endotherms at 222 °C and 229 °C (peak maxima) are observed in the DSC data and are associated with melt/decomposition of the sample based on the TGA data.
  • Crystalline Form C was isolated after the addition of water to solutions of NMP, DMF, or DMSO ( Figure 22). Due to sample limitation, analyses were performed on Form C samples generated from different solvent systems.
  • Form C was slurried in water for 4 weeks at RT and resulted in Form A.
  • the result suggests Form C is likely not a true hydrate or if it is a hydrate, it is a metastable hydrate.
  • Form C is likely a solvated/hydrated form of Compound 1 that converts to Form A upon drying and slurrying in water.
  • Crystalline Form D was prepared from evaporation of an acetic acid solution.
  • the XRPD pattern of the isolated solids was successfully indexed ( Figure 25).
  • the indexed unit cell volume (2689.3 A3/cell) indicates quite a bit of extra space in addition to the compound 1 molecule, consistent with a 1:2 acetate solvate/salt.
  • 1 H NMR analysis confirms the presence of 2 moles acetic acid/acetate per mole Compound 1 in the sample, consistent with a 1 :2 Compound 1 acetate salt/solvate.
  • Form D is likely a diacetate salt/solvate of Compound 1.
  • Amorphous material was prepared from evaporation of a TFE solution ( Figure 27). An XRPD pattern showing no reflections was also obtained for solids isolated after evaporation of a HFIPA solution. The sample isolated from TFE was characterized by XRPD, 1H NMR and TGA/DSC analyses.
  • the 1 H NMR spectrum is consistent with the structure of the API and contains approx. 1 mole TFE per mole API.
  • Thermal analysis is presented in Figure 28.
  • the sample exhibits a continuous weight loss upon heating between 45 and 170 °C in the TGA data, likely attributed to the loss of volatiles.
  • the weight loss observed in the TGA data is associated with a stepwise feature at ⁇ 70 °C and a broad endotherm at 162 °C (peak max) based on the DSC data. Further investigation of the amorphous material, including modulated DSC analysis to determine the glass transition temperature, would be needed to better understand the thermal events observed.
  • Form A was obtained under various experimental conditions including evaporation of an acetonitrile solution, precipitation upon stirring solutions produced by the addition of antisolvents to solutions of the API specifically DMA/MTBE, HFIPA/toluene and TFE/IPA solvent/antisolvent additions, rotary evaporation of a DMSO/acetone solution and after slurrying Compound 1 free base in THF at ⁇ 50 °C for 2 weeks.
  • Compound 1 - Free Base Form B Form B was prepared by slurrying Compound 1 free base in tetrahydrofuran (THF) for 4 weeks at RT or 2 weeks at 2-8 °C.
  • the Raman spectrum was acquired on a FT-Raman module interfaced to a Nexus 670 FT- IR spectrophotometer (Thermo Nicolet) equipped with an indium gallium arsenide (InGaAs) detector. Wavelength verification was performed using sulfur and cyclohexane.
  • the sample was prepared for analysis by placing the sample into a glass tube and positioning the tube in a gold-coated tube holder. Approximately 0.301 W of Nd:YVC>4 laser power (1064 nm excitation wavelength) was used to irradiate the sample. The spectrum represents 256 co added scans collected at a spectral resolution of 4 cm -1 .
  • XRPD X-ray Powder Diffraction
  • XRPD patterns were collected with a PANalytical Empyrean diffractometer using an incident beam of Cu radiation produced using a long, fine-focus source.
  • An elliptically graded multilayer mirror was used to focus Cu Ka X-rays through the specimen and onto the detector.
  • a silicon specimen NIST SRM 640e was analyzed to verify the observed position of the Si 111 peak is consistent with the NIST-certified position.
  • a specimen of the sample was sandwiched between 3-pm-thick films and analyzed in transmission geometry. A beam-stop, short antiscatter extension, and antiscatter knife edge were used to minimize the background generated by air.
  • Soller slits for the incident and diffracted beams were used to minimize broadening and asymmetry from axial divergence. Diffraction patterns were collected using a scanning position-sensitive detector (X'Celerator) located 240 mm from the specimen and Data Collector software v. 5.5. iii) X-ray Powder Diffraction Peak Identification Process
  • the range of data collected is typically provided in the form in which the data were initially reported, and is instrument dependent. Under most circumstances, peaks within the range of up to about 30° 2Q were selected. Rounding algorithms were used to round each peak to the nearest 0.1° or 0.01° 2Q, depending upon the instrument used to collect the data and/or the inherent peak resolution. The location of the peaks along the x-axis (° 2Q) in both the figures and the tables were determined using proprietary software (TRIADSTM v2.1) and rounded to one or two significant figures after the decimal point based upon the above criteria.
  • Peak position variabilities are given to within ⁇ 0.2° 2Q based upon recommendations outlined in the USP discussion of variability in x-ray powder diffraction (United States Pharmacopeia, USP 41-NF 36, through S2, ⁇ 941 >, official from 3/1/2018). The accuracy and precision associated with any particular measurement reported herein has not been determined. Moreover, third party measurements on independently prepared samples on different instruments may lead to variability which is greater than ⁇ 0.2° 2Q. For d-space listings, the wavelength used to calculate d-spacings was 1.5405929A, the Cu-K ci wavelength ( Phys . Rev. A56 (6) 4554-4568 (1997)). Variability associated with d-spacing estimates was calculated from the USP recommendation, at each d-spacing, and provided in the respective data tables.
  • variable hydrates and solvates may display peak variances greater than 0.2° 2Q and therefore peak variances of 0.2° 2Q are not applicable to these materials.
  • peak tables contain data identified only as "Prominent Peaks". These peaks are a subset of the entire observed peak list. Prominent peaks are selected from observed peaks by identifying preferably non overlapping, low-angle peaks, with strong intensity.
  • assessments of particle statistics (PS) and/or preferred orientation (PO) are possible. Reproducibility among XRPD patterns from multiple samples analyzed on a single diffractometer indicates that the particle statistics are adequate. Consistency of relative intensity among XRPD patterns from multiple diffractometers indicates good orientation statistics. Alternatively, the observed XRPD pattern may be compared with a calculated XRPD pattern based upon a single crystal structure, if available. Two-dimensional scattering patterns using area detectors can also be used to evaluate PS/PO. If the effects of both PS and PO are determined to be negligible, then the XRPD pattern is representative of the powder average intensity for the sample and prominent peaks may be identified as “Representative Peaks”.
  • Characteristic peaks are a subset of Representative Peaks and are used to differentiate one crystalline polymorph from another crystalline polymorph (polymorphs being crystalline forms having the same chemical composition). Characteristic peaks are determined by evaluating which representative peaks, if any, are present in one crystalline polymorph of a compound against all other known crystalline polymorphs of that compound to within ⁇ 0.2 °2Q. Not all crystalline polymorphs of a compound necessarily have at least one characteristic peak.
  • Compound 1 (free base), which is composed of a physical mixture of Form A and Form B, was used as the starting material for the study.
  • Form A and Form B are likely anhydrous forms of Compound 1 that were isolated as single crystalline phases during this study and characterized.
  • Form C and Form D two unique crystalline phases, Form C and Form D, as well as X-ray amorphous material, were discovered during screening.
  • Form C is a potentially solvated/hydrated form of Compound 1 that converts to Form A upon drying and slurrying in water.
  • Form D is a diacetate solvate/salt.

Abstract

The invention relates to solid state forms, including amorphous and crystalline forms, of the CHK1 inhibitor compound 5-((5-(4-(4-fluoro-1-methylpiperidin-4-yl)-2-methoxyphenyl)-1H-pyrazol-3-yl)amino)pyrazine-2-carbonitrile and methods of preparing specific amorphous and polymorphic forms of the compound.

Description

AMORPHOUS AND POLYMORPHIC FORM OF A SPECIFIC CHK1 INHIBITOR
This application claims priority from US provisional patent applications numbers 62/944,139 (filed on 5 December 2019) and 62/946,223 (filed 10 December 2019), the contents of each of which are incorporated herein by reference in their entirety.
The invention relates to solid-state forms of a Chk1 inhibitor, including polymorphic and amorphous forms, and methods for preparing the solid-state forms.
Background of the Invention
The ability of a substance to exist in more than one crystalline form is defined as polymorphism and these different crystalline forms are named "polymorphs". In general, polymorphism is caused by the ability of the molecule of a substance to change its conformation or to form different intermolecular and intramolecular interactions giving different atom arrangements that are reflected in the crystal lattices of different polymorphs. However, polymorphism is not a universal feature of solids, since some molecules can exist in one or more crystal forms while other molecules cannot.
The different polymorphs of a substance possess different energies of the crystal lattice and thus each polymorph typically shows one or more different physical properties in the solid state, such as density, melting point, color, stability, dissolution rate, flowability, compatibility with milling, granulation and compacting and/or uniformity of distribution (see, e.g., P. DiMartino, et al., J. Thermal Anal. 48:447458 (1997)). Therefore, the capacity of any given compound to occur in one or more crystalline forms (i.e. , polymorphs) is unpredictable as are the physical properties of any single crystalline form. Different physical properties of a substance may affect the ability to prepare different pharmaceutical formulations comprising the substance and may also affect the stability, dissolution and bioavailability of a solid-state formulation, which subsequently affects suitability or efficacy of such formulations in treating disease.
In the case of a chemical substance that exists in more than one polymorphic form, the less thermodynamically stable forms can occasionally convert to the more thermodynamically stable form at a given temperature after a sufficient period of time. When this transformation is not rapid, such a thermodynamically unstable form is referred to as a "metastable" form. In some instances, the stable form exhibits the highest melting point, the lowest solubility, and the maximum chemical stability. In other cases, the metastable form may exhibit sufficient chemical and physical stability under normal storage conditions to permit its use in a commercial form. In that case, the metastable form, although less thermodynamically stable, may exhibit properties desirable over those of the stable form, such as enhanced solubility or better oral bioavailability. Likewise, the amorphous form of an active pharmaceutical ingredient may have enhanced solubility in comparison to crystalline material due reduction of crystal lattice forces in the amorphous material that must be overcome to effect dissolution.
Chk1 is a serine/threonine kinase that is involved in the induction of cell cycle checkpoints in response to DNA damage and replicative stress. Chk1 inhibition abrogates the intra S and G2/M checkpoints and has been shown to selectively sensitize tumor cells to well-known DNA damaging agents. (See, e.g., McNeely, S. et al. Pharmacology & Therapeutics 2014 (dx.doi.org/10.1016/j.pharmthera.2013.10.005)).
Resistance to chemotherapy and radiotherapy, a clinical problem for conventional therapy, has been associated with activation of the DNA damage response in which Chk1 has been implicated (Nature 2006; 444(7): 756-760) and the inhibition of Chk1 sensitizes lung cancer brain metastases to radiotherapy ( Biochem . Biophys. Res. Commun. 2011; 406(1):53-8). Chk1 inhibitors, either as single agents or in combination, are useful, as an example, in treating tumor cells in which constitutive activation of DNA damage and checkpoint pathways drives genomic instability.
WO 2015/120390 describes a series of Chk1 inhibitors, including the compound 5-((5-(4-(4- fluoro-1-methylpiperidin-4-yl)-2-methoxyphenyl)-1H-pyrazol-3-yl)amino)pyrazine-2- carbonitrile (see Example 64).
Knowledge of polymorphism of a CHK1 inhibitor for use in preparing a composition to treat cancer in a subject will sometimes affect the development of that inhibitor as a medicament. On the basis of that knowledge, an individual solid-state form of a CHK1 inhibitor having one or more desirable properties would be selected for the development of a pharmaceutical formulation having the desired property(ies).
The Invention
It has now been found that 5-((5-(4-(4-fluoro-1-methylpiperidin-4-yl)-2-methoxyphenyl)-1H- pyrazol-3-yl)amino)pyrazine-2-carbonitrile, hereinafter referred to as “Compound 1” or the “compound of the invention”, can form several different crystalline forms as well as in an amorphous form. Accordingly, in a first embodiment (Embodiment 0.1), the invention provides Compound 1, having the formula (1):
Figure imgf000005_0001
in an amorphous form.
In a second embodiment (Embodiment 1.0), the invention provides a substantially crystalline form of Compound 1, having the formula (1):
Figure imgf000005_0002
The crystalline form can be, for example, any one of crystalline forms A, B, C and D as defined herein.
Compound 1 can be prepared according to the procedure of WO 2015/120390 (Example 64). XRPD analysis of the initially isolated product from this procedure indicates that it is composed of a mixture of two crystalline phases, Form A and Form B. Characterization data suggest Forms A and B are likely anhydrous forms of Compound 1.
In an amorphous solid, the three dimensional structure that normally exists in a crystalline form does not exist and the positions of the molecules relative to one another in the amorphous form are essentially random, see for example Hancock et al. J. Pharm. Sci. (1997), 86, 1).
The term “substantially crystalline” as used herein refers to forms of the compound of formula (1) in which it is from 50% to 100% crystalline. Within this range, the compound of formula (1) may be at least 55% crystalline, or at least 60% crystalline, or at least 70% crystalline, or at least 80% crystalline, or at least 90% crystalline, or at least 95% crystalline, or at least 98% crystalline, or at least 99% crystalline, or at least 99.5% crystalline, or at least 99.9% crystalline, for example 100% crystalline.
The crystalline form of the Compound 1 is preferably one having a crystalline purity of at least 90%, more preferably at least 95%; i.e. at least 90% (more preferably at least 95%) of the compound is of a single crystalline form (e.g. Form A or Form B).
The crystalline forms of the compound of the invention may be solvated (e.g. hydrated) or non-solvated (e.g. anhydrous).
The term "anhydrous" as used herein does not exclude the possibility of the presence of some water on or in the compound (e.g. a crystal of the compound). For example, there may be some water present on the surface of the compound (e.g. compound crystal), or minor amounts within the body of the compound (e.g. crystal). Typically, an anhydrous form contains fewer than 0.4 molecules of water per molecule of compound, and more preferably contains fewer than 0.1 molecules of water per molecule of compound, for example 0 molecules of water.
Where the crystalline forms are hydrated, they can contain, for example, up to three molecules of water of crystallisation, more usually up to two molecules of water, e.g. one molecule of water or two molecules of water. Non-stoichiometric hydrates may also be formed in which the number of molecules of water present is less than one or is otherwise a non-integer. For example, where there is less than one molecule of water present, there may be 0.4, or 0.5, or 0.6, or 0.7, or 0.8, or 0.9 molecules of water present per molecule of compound (1). The crystalline forms described herein, crystals thereof and their crystal structures form further aspects of the invention.
The crystalline forms can be characterised using a number of techniques including, X-ray powder diffraction (XRPD), single crystal X-ray diffraction, differential scanning calorimetry (DSC) and thermogravimetric analysis (TGA). The behaviour of the crystals under conditions of varying humidity can be analysed by gravimetric vapour sorption studies (such as dynamic vapour sorption (DVS)).
The crystalline structure of a compound can be analysed by the solid-state technique of X- ray Powder Diffraction (XRPD). XRPD can be carried out according to conventional methods such as those described herein (see the Examples below) and in Introduction to X- ray Powder Diffraction, Ron Jenkins and Robert L. Snyder (John Wiley & Sons, New York, 1996). The presence of defined peaks (as opposed to random background noise) in an XRPD diffractogram indicates that the compound has a degree of crystallinity.
A compound’s X-ray powder pattern is characterised by the diffraction angle (2Q) and interplanar spacing (d) parameters of an X-ray diffraction spectrum. These are related by Bragg's equation, nA=2d Sin Q, (where n=1; A=wavelength of the X-ray radiation; d=interplanar spacing; and 9=diffraction angle). XRPD data for each of the crystalline Forms A, B, C and E are set out below. The relative intensities given should not be strictly interpreted since they may vary depending on the direction of crystal growth, particle sizes and the measurement conditions. In addition, the diffraction angles usually mean those that coincide in the range of 2Q ±0.2°.
Form A
Form A can be obtained under various experimental conditions, including:
• evaporation of an acetonitrile solution;
• slurrying Form C in water;
• precipitation upon stirring solutions produced by the addition of antisolvents to solutions of Compound 1 (free base), specifically DMA/MTBE, HFIPA/toluene and TFE/IPA solvent/antisolvent additions; and
• evaporation (e.g. rotary evaporation) of a DMSO/acetone solution and after slurrying Compound 1 (free base) in THF at 50 °C for 2 weeks.
Further methods of preparing crystalline Form A are set out below and in the Examples. The XRPD diffractogram for Form A is shown in Figure 8.
The X-ray diffraction pattern of crystalline Form A of compound (1) exhibits peaks of greatest intensity at the diffraction angles (2Q) set out in Table A-1
Figure imgf000008_0001
Accordingly, in further embodiments, the invention provides:
1.1 A crystalline form according to Embodiment 1.0 which is of Form A as defined herein.
1.2 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° (±0.2°).
1.3 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 10.5° (±0.2°).
1.4 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 16.9° (±0.2°).
1.5 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 17.4° (±0.2°).
1.6 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 20.8° (±0.2°). 1.7 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 26.4° (±0.2°).
1.8 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 28.4° (±0.2°)
1.9 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three or more, or four or more, five or more, and in particular six diffraction angles (2Q) selected from 10.5°, 16.9°, 17.4°, 20.8°, 26.4° and 28.4° (±0.2°).
The X-ray powder diffraction pattern of Form A of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table A-2.
Figure imgf000009_0001
Therefore, in further embodiments, the invention provides:
1.10 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° (±0.2°) (e.g. at least four and more particularly at least five of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 5.2°, 8.5°, 9.8°, 13.9°, 15.8°, 17.1°, 17.7° and 19.4° (±0.2°).
1.11 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° (±0.2°) (e.g. at least four and more particularly at least five of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 9.8°, 17.1° and 17.7° (±0.2°).
1.12 A substantially crystalline form (Form A) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° (±0.2°), and one or more further peaks at diffraction angles (2Q) selected from 9.8°, 17.1° and 17.7° (±0.2°).
In further embodiments, the invention provides:
1.13 A substantially crystalline form (Form A) of compound (1) which exhibits peaks at the diffraction angles set forth in Table 7 or Table 8 provided below which have a relative intensity of at least 15%.
1.14 A substantially crystalline form (Form A) of compound (1) which exhibits peaks at the diffraction angles corresponding to those of the X-ray powder diffraction pattern shown in Figure 8.
1.15 A substantially crystalline form (Form A) of compound (1) having an X-ray powder diffraction pattern substantially as shown in Figure 8.
Crystalline Form A can also be characterised by differential scanning calorimetry (DSC) and has been found to exhibit an endothermic event with an onset temperature of about 224 °C and a peak at about 226 °C as shown in Figure 15 of the accompanying drawings.
Crystalline Form A can also be characterized by Raman Spectroscopy and the Raman spectrum for Form A is shown in Figure 16.
Therefore, in a further embodiment, the invention provides: 1.16 A substantially crystalline form (Form A) of compound (1) according to any one of Embodiments 1.1 to 1.15 which has a DSC thermogram characterized by an endotherm with an onset at a temperature of about 224 °C and a peak maximum at about 226 °C.
1.17 A substantially crystalline form (Form A) of compound (1) according to any one of Embodiments 1.1 to 1.15 which has a DSC thermogram substantially as shown in Figure 15 of the drawings appended hereto.
1.18 A substantially crystalline form (Form A) of compound (1) which exhibits peaks in its Raman spectra at the Raman shifts set forth in Table 9 provided below which have a Raman intensity of at least 150, for example at least 250, preferably at least 350.
1.19 A substantially crystalline form (Form A) of compound (1) which exhibits peaks in its Raman spectra at the Raman shifts corresponding to those of the Raman spectrum shown in Figure 16.
1.20 A substantially crystalline form (Form A) of compound (1) having a Raman spectrum substantially as shown in Figure 16.
1.21 A substantially crystalline form (Form A) of compound (1) according to any one of Embodiments 1.1 to 1.20 having a crystalline purity of at least 80%.
1.22 A substantially crystalline form (Form A) of compound (1) according to any one of Embodiments 1.1 to 1.20 having a crystalline purity of at least 90%.
1.23 A substantially crystalline form (Form A) of compound (1) according to any one of Embodiments 1.1 to 1.20 having a crystalline purity of at least 95%.
1.23A A composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form A.
1.23B A composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form A.
1.23C A composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form A. 1.23D A composition of matter comprising a substantially crystalline form (Form A) of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form A.
The invention also provides methods for making crystalline Form A. Accordingly, in further embodiments, the invention provides:
1.24 A method of preparing crystalline Form A of Compound 1 as defined in any one of Embodiments 1.1 to 1.23, which method comprises:
(i) forming a solution of Compound 1 free base in a polar aprotic solvent such as acetonitrile, dimethylsulphoxide or acetone, or mixtures thereof, and then evaporating the solvent (for example by rotary evaporation);
(ii) forming a slurry of Compound 1 free base (e.g. Form C) in water and stirring the slurry until the slurry consists predominantly of Form A (for example until the slurry consists of greater than 80% Form A, preferably greater than 90% Form A, for example greater than 95% Form A); or
(iii) forming a solution of Compound 1 free base in a polar solvent such as DMA, TFE or HFIPA or mixtures of two or more thereof and adding an anti-solvent (e.g. an antisolvent selected from MTBE, toluene and I PA and mixtures of two or more thereof) to precipitate Compound 1 Form A.
1.25 A method according to Embodiment 1.24 which wherein the solution of Compound 1 free base is formed in acetonitrile and the solution is then evaporated.
1.26 A method according to Embodiment 1.24 wherein the solution of Compound 1 free base is formed in a mixture of dimethylsulphoxide and acetone and then evaporating the solvent (for example by rotary evaporation).
Form B
Form B can be prepared by slurrying Compound 1 (free base) in THF for 4 weeks at room temperature or 2 weeks at 2-8 °C.
The XRPD diffractogram for Form B is shown in Figure 17.
The X-ray diffraction pattern of crystalline Form B of compound (1) exhibits peaks of greatest intensity at the diffraction angles (2Q) set out in Table B-1
Figure imgf000013_0001
Accordingly, in further embodiments, the invention provides:
2.1 A crystalline form according to Embodiment 1.0 which is of Form B as defined herein.
2.2 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5° (±0.2°).
2.3 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 7.6° (±0.2°).
2.4 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 18.8° (±0.2°).
2.5 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 21.5° (±0.2°).
2.6 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three diffraction angles (2Q) selected from 7.6°, 18.8° and 21.5° (±0.2°).
The X-ray powder diffraction pattern of Form B of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table B-2.
Figure imgf000013_0002
Figure imgf000014_0001
Therefore, in further embodiments, the invention provides:
2.7 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5° (±0.2°) (e.g. at least two and more particularly three of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 12.2°, 14.2°, 15.0°, 15.2°, 22.3°, 27.0° and 29.0° (±0.2°).
2.8 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5° (±0.2°) (e.g. at least two and more particularly three of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 15.0°, 15.2° and 29.0° (±0.2°).
2.9 A substantially crystalline form (Form B) of the compound of formula (1 ) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6°, 18.8° and 21.5° (±0.2°), and one or more further peaks at diffraction angles (2Q) selected from 15.0°, 15.2° and 29.0° (±0.2°).
In further embodiments, the invention provides:
2.10 A substantially crystalline form (Form B) of compound (1) which exhibits peaks at the diffraction angles set forth in Table 10 or Table 11 which have a relative intensity of at least 15%.
2.11 A substantially crystalline form (Form B) of compound (1) which exhibits peaks at the diffraction angles corresponding to those of the X-ray powder diffraction pattern shown in Figure 17. 2.12 A substantially crystalline form (Form B) of compound (1) having an X-ray powder diffraction pattern substantially as shown in Figure 17.
The crystalline form of the invention can also be characterised by differential scanning calorimetry (DSC). The DSC thermogram of Form B is characterized by endotherms with overlapping peaks at about 222 °C and about 229 °C as shown in Figure 18. Crystalline Form B can also be characterized by Raman Spectroscopy and the Raman spectrum for Form B is shown in Figure 19.
Therefore, in a further embodiment, the invention provides:
2.16 A substantially crystalline form (Form B) of compound (1) according to any one of Embodiments 2.1 to 2.15 which has a DSC thermogram characterized by endotherms with peaks maxima at about 222 °C and/or about 229 °C
2.17 A substantially crystalline form (Form B) of compound (1) according to any one of Embodiments 2.1 to 2.15 which has a DSC thermogram substantially as shown in Figure 18 of the drawings appended hereto.
2.18 A substantially crystalline form (Form B) of compound (1) which exhibits peaks in its Raman spectra at the Raman shifts set forth in Table 12 provided below which have a Raman intensity of at least 150, for example at least 250, preferably at least 350.
2.19 A substantially crystalline form (Form B) of compound (1) which exhibits peaks in its Raman spectra at the Raman shifts corresponding to those of the Raman spectrum shown in Figure 19.
2.20 A substantially crystalline form (Form B) of compound (1) having a Raman spectrum substantially as shown in Figure 19.
2.21 A substantially crystalline form (Form B) of compound (1) according to any one of Embodiments 2.1 to 2.20 having a crystalline purity of at least 80%.
2.22 A substantially crystalline form (Form B) of compound (1) according to any one of Embodiments 2.1 to 2.20 having a crystalline purity of at least 90%.
2.23 A substantially crystalline form (Form B) of compound (1) according to any one of Embodiments 2.1 to 2.20 having a crystalline purity of at least 95%. 2.23A A composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form B.
2.23B A composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form B.
2.23C A composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form B.
2.23D A composition of matter comprising a substantially crystalline form (Form B) of Compound 1 as defined in any one of Embodiments 2.1 to 2.23, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form B.
Form C
Crystalline Form C can be isolated after the addition of water to solutions of Compound 1 in a polar aprotic solvent such as NMP, DMF or DMSO. Based on the experimental data provided herein, Form C is considered to be a solvated/hydrated form of Compound 1 that converts to Form A upon drying and slurrying in water. The results described herein suggest that Form C is likely not a true hydrate or, if it is a hydrate, it is a metastable hydrate.
The XRPD diffractogram for Form C is shown in Figure 23.
The X-ray diffraction pattern of crystalline Form C of compound (1) exhibits peaks of greatest intensity at the diffraction angles (2Q) set out in Table C-1.
Figure imgf000016_0001
Accordingly, in further embodiments, the invention provides: 3.1 A crystalline form according to Embodiment 1.0 which is of Form C as defined herein.
3.2 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at about the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0° (±0.2°).
3.3 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 5.8° (±0.2°).
3.4 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 8.7° (±0.2°).
3.5 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 26.5° (±0.2°).
3.6 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 27.0° (±0.2°).
3.7 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three or more, or four or more, and in particular five diffraction angles (2Q) selected from 5.8°, 8.7°, 26.5° and 27.0° (±0.2°).
The X-ray powder diffraction pattern of Form C of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table C-2.
Figure imgf000017_0001
Therefore, in further embodiments, the invention provides: 3.8 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0° (±0.2°) (e.g. at least three and more particularly four of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 13.8° and 16.3° (±0.2°).
3.9 A substantially crystalline form (Form C) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0° (±0.2°), and one or more further peaks at diffraction angles (2Q) selected from about 13.8° and about 16.3°(±0.2°).
In further embodiments, the invention provides:
3.10 A substantially crystalline form (Form C) of compound (1) which exhibits peaks at the diffraction angles corresponding to those of the X-ray powder diffraction pattern shown in Figure 23.
3.11 A substantially crystalline form (Form C) of compound (1) having an X-ray powder diffraction pattern substantially as shown in Figure 23.
The crystalline form of the invention can also be characterised by differential scanning calorimetry (DSC). The DSC thermogram of Form C is characterized by endotherms as shown in Figure 24. Accordingly, the invention also provides:
3.12 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.11 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 62°C and a peak maximum at about 67°C.
3.13 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.12 which has a DSC thermogram characterized by an endotherm having a peak maximum at about 107°C.
3.14 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.13 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 221 °C and a peak maximum at about 225°C.
3.15 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.14 which has a DSC thermogram substantially as shown in Figure 24 of the drawings appended hereto. The crystalline form of the invention can also be characterised by thermogravimetric analysis (TGA). The TGA plot of Form C is shows a weight loss of 37.2% wt from 49°C to 135°C, as shown in Figure 24. Accordingly, the invention also provides:
3.16 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.15 which has a TGA plot characterized by a weight loss of about 37.2% wt between 49°C and 135°C.
3.17 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.16 which has a TGA plot substantially as shown in Figure 24 of the drawings appended hereto.
3.18 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.17 having a crystalline purity of at least 80%.
3.19 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.17 having a crystalline purity of at least 90%.
3.20 A substantially crystalline form (Form C) of compound (1) according to any one of Embodiments 3.1 to 3.17 having a crystalline purity of at least 95%.
3.21 A composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 2.20, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form C.
3.22 A composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 2.20, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form C.
3.23 A composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 3.20, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form C.
3.24 A composition of matter comprising a substantially crystalline form (Form C) of Compound 1 as defined in any one of Embodiments 3.1 to 3.20, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form C.
Form D Crystalline Form D of the compound (1) can be formed Crystalline Form D from evaporation of a solution of Compound 1 in acetic acid. Form D is considered to be an acetate solvate of Compound 1. 1H NMR analysis indicates the presence of 2 moles acetic acid/acetate per mole Compound 1 in the sample, consistent with a 1:2 Compound 1 acetate salt/solvate.
The XRPD diffractogram for Form D is shown in Figure 25.
The X-ray diffraction pattern of crystalline Form D of compound (1) exhibits peaks of greatest intensity at the diffraction angles (2Q) set out in Table D-1.
Figure imgf000020_0001
Accordingly, in further embodiments, the invention provides:
4.1 A crystalline form according to Embodiment 1.0 which is of Form D as defined herein.
4.2 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9°(±0.2°).
4.3 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 10.7° (±0.2°).
4.4 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 16.6° (±0.2°).
4.5 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of a major peak at the diffraction angle (2Q) 25.9° (±0.2°).
4.8 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at two or more, e.g. three or more, or four or more, and in particular five diffraction angles (2Q) selected from 10.7°, 16.6° and 25.9° (±0.2°).
The X-ray powder diffraction pattern of Form D of compound (1) may also have lesser peaks present at the diffraction angles (2Q) set out in Table D-2.
Figure imgf000021_0001
Therefore, in further embodiments, the invention provides:
4.9 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9 °(±0.2°) (e.g. at least two and more particularly three of the diffraction angles), and optionally one or more further peaks at diffraction angles (2Q) selected from 6.5°, 17.7°, 18.9°, 26.1° and 26.9° (±0.2°).
4.10 A substantially crystalline form (Form D) of the compound of formula (1) having an X- ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9° (±0.2°), and one or more further peaks at diffraction angles (2Q) selected from 6.5°, 17.7°, 18.9°, 26.1° and 26.9° (±0.2°).
In further embodiments, the invention provides:
4.11 A substantially crystalline form (Form D) of compound (1) which exhibits peaks at the diffraction angles corresponding to those of the X-ray powder diffraction pattern shown in Figure 25.
4.12 A substantially crystalline form (Form D) of compound (1) having an X-ray powder diffraction pattern substantially as shown in Figure 25. Crystalline Form D can also be characterised by differential scanning calorimetry (DSC) and has a thermogram substantially as shown in Figure 12.
The crystalline form of the invention can also be characterised by differential scanning calorimetry (DSC). The DSC thermogram of Form D is characterized by endotherms as shown in Figure 26. Accordingly, the invention also provides:
4.13 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.12 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 62°C and a peak maximum at about 86°C.
4.14 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.13 which has a DSC thermogram characterized by an endotherm having an onset temperature at about 161°C and a peak maximum at about 170°C.
4.15 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.14 which has a DSC thermogram substantially as shown in Figure 26 of the drawings appended hereto.
The crystalline form of the invention can also be characterised by thermogravimetric analysis (TGA). The TGA plot of Form D is shows a weight loss of 8.2% wt from 40°C to 114°C, as shown in Figure 26. Accordingly, the invention also provides:
4.16 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.15 which has a TGA plot characterized by a weight loss of about 8.2% wt between 40°C and 114°C.
4.17 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.16 which has a TGA plot substantially as shown in Figure 26 of the drawings appended hereto.
4.18 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.17 having a crystalline purity of at least 80%.
4.19 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.17 having a crystalline purity of at least 90%.
4.20 A substantially crystalline form (Form D) of compound (1) according to any one of Embodiments 4.1 to 4.17 having a crystalline purity of at least 95%. 4.21 A composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 80% by weight of the Compound 1 in the composition is in crystalline Form D.
4.22 A composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 90% by weight of the Compound 1 in the composition is in crystalline Form D.
4.23 A composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 95% by weight of the Compound 1 in the composition is in crystalline Form D.
4.24 A composition of matter comprising a substantially crystalline form (Form D) of Compound 1 as defined in any one of Embodiments 4.1 to 4.20, wherein at least 99% by weight of the Compound 1 in the composition is in crystalline Form D.
Isotopes
The crystalline forms of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or the amorphous form as defined in Embodiment 0.1 may contain one or more isotopic substitutions, and a reference to a particular element includes within its scope all isotopes of the element. For example, a reference to hydrogen includes within its scope 1H, 2H (D), and 3H (T). Similarly, references to carbon and oxygen include within their scope respectively 12C, 13C and 14C and 160 and 180.
In an analogous manner, a reference to a particular functional group also includes within its scope isotopic variations, unless the context indicates otherwise.
The isotopes may be radioactive or non-radioactive. In one general embodiment of the invention (Embodiment 5.1), the crystalline/amorphous form of the compound of formula (1) as defined in any one of Embodiments 0.1 to 4.20 contains no radioactive isotopes. Such compounds are preferred for therapeutic use. In another embodiment (Embodiment 5.2), however, the crystalline/amorphous form of the compound of formula (1) as defined in any one of Embodiments 0.1 to 4.20 may contain one or more radioisotopes. Compounds containing such radioisotopes may be useful in a diagnostic context.
Bioloqical Activity Crystalline/amorphous forms of the compound of formula (1) are potent inhibitors of Chk-1 and consequently are expected to be beneficial alone or in combination with various chemotherapeutic agents or radiation for treating a wide spectrum of proliferative disorders.
Accordingly, in further embodiments (Embodiments 6.1 to 6.10), the invention provides:
6.1 A crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in medicine or therapy.
6.2 A crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use as a Chk-1 kinase inhibitor.
6.3 A crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in enhancing a therapeutic effect of radiation therapy or chemotherapy in the treatment of a proliferative disease such as cancer.
6.4 A crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in the treatment of a proliferative disease such as cancer.
6.5 The use of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for the manufacture of a medicament for enhancing a therapeutic effect of radiation therapy or chemotherapy in the treatment of a proliferative disease such as cancer.
6.6 The use of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for the manufacture of a medicament for the treatment of a proliferative disease such as cancer.
6.7 A method for the prophylaxis or treatment of a proliferative disease such as cancer, which method comprises administering to a patient in combination with radiotherapy or chemotherapy a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
6.8 A method for the prophylaxis or treatment of a proliferative disease such as cancer, which method comprises administering to a patient a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
6.9 A compound for use, use or method as defined in any one of Embodiments 6.3 to 6.8 wherein the cancer is selected from carcinomas, for example carcinomas of the bladder, breast, colon, kidney, epidermis, liver, lung, oesophagus, gall bladder, ovary, pancreas, stomach, cervix, thyroid, prostate, gastrointestinal system, or skin, hematopoieitic tumours such as leukaemia, B-cell lymphoma, T-cell lymphoma, Hodgkin’s lymphoma, non-Hodgkin’s lymphoma, hairy cell lymphoma, or Burkett's lymphoma; hematopoieitic tumours of myeloid lineage, for example acute and chronic myelogenous leukaemias, myelodysplastic syndrome, or promyelocytic leukaemia; thyroid follicular cancer; tumours of mesenchymal origin, for example fibrosarcoma or habdomyosarcoma; tumours of the central or peripheral nervous system, for example astrocytoma, neuroblastoma, glioma or schwannoma; melanoma; seminoma; teratocarcinoma; osteosarcoma; xeroderma pigmentosum; keratoctanthoma; thyroid follicular cancer; Ewing’s sarcoma or Kaposi's sarcoma.
6.10 A compound for use, use or method according to Embodiment 6.9 wherein the cancer is selected from breast cancer, colon cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, glioma, Ewing’s sarcoma and leukemia.
It is also envisaged that the Chk-1 inhibitors of the invention may be useful in treating tumours in which there is a defective DNA repair mechanism or a defective cell cycle, for example a cancer in which mutations (e.g. in p53) have led to the G1/S DNA damage checkpoint being lost (see the introductory section of this application). The Chk-1 inhibitors of the invention may also be useful in treating RAD17 mutant tumours, ATM-deficient RAD50-mutant tumours and fanconi anaemia. Accordingly in further embodiments (Embodiments 6.11 to 6.24), the invention provides:
6.11 A crystalline form or an amorphous form or composition of matter for use, use or method as defined in any one of Embodiments 6.3 to 6.10 wherein the cancer is one which is characterized by a defective DNA repair mechanism or defective cell cycle.
6.12 A crystalline form or an amorphous form or a composition of matter for use, use or method according to Embodiment 6.11 wherein the cancer is a p53 negative or mutated tumour.
6.13 A crystalline form or an amorphous form or a composition of matter for use, use or method as defined in any one of Embodiments 6.3 to 6.10 wherein the cancer is an MYC oncogene-driven cancer.
6.14 A crystalline form or an amorphous form or a composition of matter for use, use or method according to Embodiment 6.13 wherein the MYC oncogene-driven cancer is a B-cell lymphoma, leukemia, neuroblastoma, breast cancer or lung cancer.
6.15 A crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in the treatment of a patient suffering from a p53 negative or mutated tumour (e.g. a cancer selected from breast cancer, colon cancer, lung cancer, ovarian cancer, pancreatic cancer, prostate cancer, glioma, and leukemia) in combination with radiotherapy or chemotherapy.
6.16 A crystalline form or an amorphous form for use according to any one of Embodiments 6.3 to 6.15 wherein, in addition to administration of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, the treatment comprises administration to a patient of a chemotherapeutic agent selected from cytarabine, etoposide, gemcitabine and SN-38.
6.17 The use of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for the manufacture of a medicament for the treatment of a patient suffering from a cancer which is characterised by a defective DNA repair mechanism or defective cell cycle.
6.18 The use according to Embodiment 6.17 wherein the cancer is a p53 negative or mutated tumour.
6.19 A method for the treatment of a patient (e.g. a human patient) suffering from a cancer which is characterised by a defective DNA repair mechanism or defective cell cycle, which method comprises administering to the patient a therapeutically effective amount of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24. 6.20 A method according to Embodiment 6.19 wherein the cancer is a p53 negative or mutated tumour.
6.21 A crystalline form or an amorphous form for use, use or method as defined in any one of Embodiments 6.3 to 6.10 wherein the cancer is a RAD17-mutant tumour or an ATM- deficient RAD50-mutant tumour.
6.22 A crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in the treatment of Fanconi anaemia.
6.23 The use of a crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for the manufacture of a medicament for the treatment of Fanconi anaemia.
6.24 A method of treating Fanconi anaemia in a subject (e.g. a human subject) in need thereof, which method comprises administering to the subject a therapeutically effective amount of crystalline form according to any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
The Chk-1 inhibitor crystalline/amorphous forms of the compound of the invention may be used alone or they may be used in combination with DNA-damaging anti-cancer drugs and/or radiation therapy to treat subjects with multi-drug resistant cancers. A cancer is considered to be resistant to a drug when it resumes a normal rate of tumour growth while undergoing treatment with the drug after the tumour had initially responded to the drug. A tumour is considered to "respond to a drug" when it exhibits a decrease in tumor mass or a decrease in the rate of tumour growth.
Methods for the Preparation of the Compound of Formula (1)
Compound 1 can be prepared as described WO 2015/120390 (see Example 64).
Details of how to prepare crystalline Forms A, B, C and D and amorphous forms of Compound 1 are described in the Examples section below.
Pharmaceutical Formulations
While it is possible for the active compound to be administered alone, it is preferable to present it as a pharmaceutical composition (e.g. formulation) comprising at least one active compound of the invention together with one or more pharmaceutically acceptable excipients such as carriers, adjuvants, diluents, fillers, buffers, stabilisers, preservatives, lubricants, or other materials well known to those skilled in the art, and optionally other therapeutic or prophylactic agents.
The term “pharmaceutically acceptable” as used herein refers to 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 a subject (e.g. human) without excessive toxicity, irritation, allergic response, or other problems or complication, commensurate with a reasonable benefit/risk ratio. Each excipient must also be “acceptable” in the sense of being compatible with the other ingredients of the formulation.
Accordingly, in further embodiments, the invention provides:
7.1 A pharmaceutical composition comprising a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-
4.24, and a pharmaceutically acceptable excipient.
In further embodiments, there are provided:
7.2 A pharmaceutical composition according to Embodiment 7.1 which comprises from approximately 1% (w/w) to approximately 95% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-
4.24, and from 99% (w/w) to 5% (w/w) of a pharmaceutically acceptable excipient or combination of excipients and optionally one or more further therapeutically active ingredients.
7.3 A pharmaceutical composition according to Embodiment 7.2 which comprises from approximately 5% (w/w) to approximately 90% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-
4.24, and from 95% (w/w) to 10% of a pharmaceutically excipient or combination of excipients and optionally one or more further therapeutically active ingredients. 7.4 A pharmaceutical composition according to Embodiment 7.3 which comprises from approximately 10% (w/w) to approximately 90% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1), or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, as defined in Embodiment 0.1 and from 90% (w/w) to 10% of a pharmaceutically excipient or combination of excipients.
7.5 A pharmaceutical composition according to Embodiment 7.4 which comprises from approximately 20% (w/w) to approximately 90% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21- 4.24, and from 80% (w/w) to 10% of a pharmaceutically excipient or combination of excipients.
7.6 A pharmaceutical composition according to Embodiment 4.5 which comprises from approximately 25% (w/w) to approximately 80% (w/w) of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21- 4.24, and from 75% (w/w) to 20% of a pharmaceutically excipient or combination of excipients.
It will be appreciated that pharmaceutical compositions comprising a crystalline form of the compound of formula (1) will typically be solid compositions such as tablets, capsules, caplets, pills, lozenges, sprays, powders, granules, sublingual tablets, wafers or patches and buccal patches, or liquid compositions such as suspensions where the active compound is in solid form..
Accordingly, in further embodiments, the invention provides:
7.7 A pharmaceutical composition according to any one of Embodiments 7.1 to 7.6 which is suitable for oral administration.
7.8 A pharmaceutical composition according to Embodiment 7.7 which is selected from tablets, capsules, caplets, pills, lozenges, powders, granules, suspensions, sublingual tablets, wafers or patches and buccal patches. 7.9 A pharmaceutical composition according to Embodiment 7.8 which is selected from tablets and capsules.
7.10 A pharmaceutical composition according to any one of Embodiments 7.1 to 7.6 which is suitable for parenteral administration and is in the form of a suspension for injection or infusion.
Pharmaceutical compositions (e.g. as defined in any one of Embodiments 7.1 to 7.10) containing a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, can be formulated in accordance with known techniques, see for example, Remington’s Pharmaceutical Sciences, Mack Publishing Company, Easton, PA, USA.
Thus, tablet compositions (as in Embodiment 7.9) can contain a unit dosage of active compound together with an inert diluent or carrier such as a sugar or sugar alcohol, e.g.; lactose, sucrose, sorbitol or mannitol; and/or a non-sugar derived diluent such as sodium carbonate, calcium phosphate, talc, calcium carbonate, or a cellulose or derivative thereof such as methyl cellulose, ethyl cellulose, hydroxypropyl methyl cellulose, and starches such as corn starch. Tablets may also contain such standard ingredients as binding and granulating agents such as polyvinylpyrrolidone, disintegrants (e.g. swellable crosslinked polymers such as crosslinked carboxymethylcellulose), lubricating agents (e.g. stearates), preservatives (e.g. parabens), antioxidants (e.g. BHT), buffering agents (for example phosphate or citrate buffers), and effervescent agents such as citrate/bicarbonate mixtures. Such excipients are well known and do not need to be discussed in detail here.
Capsule formulations (as in Embodiment 7.9) may be of the hard gelatin or soft gelatin variety and can contain the active component in solid, semi-solid, or liquid form. Gelatin capsules can be formed from animal gelatin or synthetic or plant derived equivalents thereof.
The solid dosage forms (e.g.; tablets, capsules etc.) can be coated or un-coated, but typically have a coating, for example a protective film coating (e.g. a wax or varnish) or a release controlling coating. The coating (e.g. a Eudragit ™ type polymer) can be designed to release the active component at a desired location within the gastro-intestinal tract. Thus, the coating can be selected so as to degrade under certain pH conditions within the gastrointestinal tract, thereby selectively release the compound in the stomach or in the ileum or duodenum. Instead of, or in addition to, a coating, the drug can be presented in a solid matrix comprising a release controlling agent, for example a release delaying agent which may be adapted to selectively release the compound under conditions of varying acidity or alkalinity in the gastrointestinal tract. Alternatively, the matrix material or release retarding coating can take the form of an erodible polymer (e.g. a maleic anhydride polymer) which is substantially continuously eroded as the dosage form passes through the gastrointestinal tract.
Compositions for topical use include ointments, creams, sprays, patches, gels, liquid drops and inserts (for example intraocular inserts). Such compositions can be formulated in accordance with known methods.
Compositions for parenteral administration (as in Embodiments 7.11 to 7.12) are typically presented as sterile aqueous or oily fine suspensions, or may be provided in finely divided sterile powder form for making up extemporaneously with sterile water for injection.
Examples of formulations for rectal or intra-vaginal administration include pessaries and suppositories which may be, for example, formed from a shaped mouldable or waxy material containing the active compound.
Compositions for administration by inhalation may take the form of inhalable powder compositions or powder sprays, and can be administrated in standard form using powder inhaler devices or aerosol dispensing devices. Such devices are well known. For administration by inhalation, the powdered formulations typically comprise the active compound together with an inert solid powdered diluent such as lactose.
The crystalline forms of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form of the compound for formula (1) as defined in Embodiment 0.1 will generally be presented in unit dosage form and, as such, will typically contain sufficient compound to provide a desired level of biological activity. For example, according to any one of Embodiments 7.1 to 7.9), a composition intended for oral administration may contain from 2 milligrams to 200 milligrams of active ingredient, more usually from 10 milligrams to 100 milligrams, for example, 12.5 milligrams, 25 milligrams and 50 milligrams.
Methods of Treatment
It is envisaged that the crystalline/amorphous forms of Compound 1 or compositions of matter as defined in any one of Embodiments 0.1 to 4.24 herein will be useful either alone or in combination therapy with chemotherapeutic agents (particularly DNA-damaging agents) or radiation therapy in the prophylaxis or treatment of a range of proliferative disease states or conditions. Examples of such disease states and conditions are set out above.
Forms of Compound 1, whether administered alone, or in combination with DNA damaging agents and other anti-cancer agents and therapies, are generally administered to a subject in need of such administration, for example a human or animal patient, preferably a human.
According to another embodiment of the invention, there is provided a combination of an the crystalline/amorphous forms of Compound 1 or compositions of matter as defined in any one of Embodiments 0.1 to 4.24 together with another chemotherapeutic agent, for example an anticancer drug.
Examples of chemotherapeutic agents that may be co-administered with the crystalline/amorphous forms of Compound 1 or compositions of matter as defined in any one of Embodiments 0.1 to 4.24 include:
• Topoisomerase I inhibitors
• Antimetabolites
• Tubulin targeting agents
• DNA binder and topoisomerase II inhibitors
• Alkylating Agents
• Monoclonal Antibodies.
• Anti-Hormones
• Signal Transduction Inhibitors
• Proteasome Inhibitors
• DNA methyl transferases
• Cytokines and retinoids
• Hypoxia triggered DNA damaging agents (e.g. Tirapazamine, TH-302)
Particular examples of chemotherapeutic agents that may be administered in combination with the crystalline/amorphous forms of Compound 1 as defined in any one of Embodiments 0.1 to 4.20 include: nitrogen mustards such as mechlorethamine, cyclophosphamide, ifosfamide, melphalan and chlorambucil; nitrosoureas such as carmustine, lomustine and semustine; ethyleneimine/methylmelamine compounds such as triethylenemelamine, triethylene thiophosphoramide and hexamethylmelamine; alkyl sulphonates such as busulfan; triazines such as dacarbazine
Antimetabolites such as folates, methotrexate, trimetrexate, 5-fluorouracil, fluorodeoxyuridine, gemcitabine, cytosine arabinoside, 5-azacytidine, 2, 2'- difluorodeoxycytidine, 6-mercaptopurine, 6-thioguanine, azathioprine, 2'-deoxycoformycin, erythrohydroxynonyl-adenine, fludarabine phosphate and 2-chlorodeoxyadenosine; type I topoisomerase inhibitors such as camptothecin, topotecan and irinotecan; type II topoisomerase inhibitors such as the epipodophylotoxins (e.g. etoposide and teniposide); antimitotic drugs such as paclitaxel, Taxotere, Vinca alkaloids (e.g. vinblastine, vincristine, vinorelbine) and estramustine (e.g. estramustine phosphate); antibiotics such as actimomycin D, daunomycin (rubidomycin), doxorubicin (adriamycin), mitoxantrone, idarubicine, bleomycin, mithramycin, mitomycin C and dactinomycin enzymes such as L-asparaginase; cytokines and biological response modifiers such as interferon (a, b,g), interleukin-2G-CSF and GM-CSF: retinoids such as retinoic acid derivatives (e.g. bexarotene); radiosensitisers such as metronidazole, misonidazole, desmethylmisonidazole, pimonidazole, etanidazole, nimorazole, nicotinamide, 5-bromodeoxyuridine, 5- iododeoxyuridine and bromodeoxycytidine; platinum compounds such as cisplatin, carboplatin, spiroplatin, iproplatin, onnaplatin, tetraplatin and oxaliplatin; anthracenediones such as mitoxantrone; ureas such as hydroxyurea; hydrazine derivatives such as N-methylhydrazine and procarbazine; adrenocortical suppressants such as mitotane and aminoglutethimide; adrenocorticosteroids and antagonists such as prednisone, dexamethasone and aminoglutethimide; progestins such as hydroxyprogesterone (e.g. hydroxyprogesterone caproate), medroxyprogesterone (e.g. medroxyprogesterone acetate) and megestrol (e.g. megestrol acetate); oestrogens such as diethylstilbestrol and ethynyl estradiol; anti-oestrogens such as tamoxifen; androgens such as testosterone (e.g. testosterone propionate) and fluoxymesterone; anti-androgens such as flutamide and leuprolide; nonsteroidal anti-androgens such as flutamide; and signal transduction inhibitors such as PARP inhibitors [e.g. as disclosed in Cancer Res. ] 66: (16)], Mek inhibitors [e.g as disclosed in Blood. 2008 September 15; 112(6): 2439-2449], farnesyltransferase inhibitors [e.g. as disclosed in Blood. 2005 Feb 15; 105(4): 1706- 16], wee1 inhibitors [e.g. as disclosed in Haematologica 2013.093187 (epub ahead of print)], rapamycin and Src inhibitors [e.g as disclosed in Blood. 2011 Feb 10; 117(6): 1947-57]
Examples of the chemotherapeutic agents than may be used in combination with the Chk-1 inhibitor compounds of Embodiments 0.1 to 4.20 as defined herein include the chemotherapeutic agents described in Blasina etal., Mol. Cancer Ther., 2008, 7(8), 2394- 2404, Ashwell et al., Clin. Cancer Res., 2008, 14(13), 4032-4037, Ashwell et al., Expert Opin. Investig. Drugs, 2008, 17(9), 1331-1340, Trends in Molecular Medicine February 2011, Vol. 17, No. 2 and Clin Cancer Res; 16(2) January 15, 2010.
Particular examples of chemotherapeutic agents that may be used in combination with the Chk-1 inhibitor compounds of the invention as defined herein include antimetabolites (such as capecitabine, cytarabine, fludarabine, gemcitabine and pemetrexed), Topoisomerase-I inhibitors (such as SN38, topotecan, irinotecan), platinum compounds (such as carboplatin, oxaloplatin and cisplatin), Topoisomerase-I I inhibitors (such as daunorubicin, doxorubicin and etoposide), thymidylate synthase inhibitors (such as 5-fluoruracil), mitotic inhibitors (such as docetaxel, paclitaxel, vincristine and vinorelbine, ) and alkylating agents (such as mitomycin C).
A further set of chemotherapeutic agents that may be used in combination with the Chk-1 inhibitor compounds of the invention as defined herein includes agents that induce stalled replication forks (see Ashwell et al., Clin. Cancer Res., above), and examples of such compounds include gemcitabine, 5-fluorouracil and hydroxyurea.
The compounds of the invention and combinations with chemotherapeutic agents or radiation therapies as described above may be administered over a prolonged term to maintain beneficial therapeutic effects or may be administered for a short period only. Alternatively they may be administered in a pulsatile or continuous manner.
The compounds of the invention will be administered in an effective amount, i.e. an amount which is effective to bring about the desired therapeutic effect either alone (in monotherapy) or in combination with one or more chemotherapeutic agents or radiation therapy. For example, the "effective amount" can be a quantity of compound which, when administered alone or together with a DNA-damaging drug or other anti-cancer drug to a subject suffering from cancer, slows tumour growth, ameliorates the symptoms of the disease and/or increases longevity. More particularly, when used in combination with radiation therapy, with a DNA-damaging drug or other anti-cancer drug, an effective amount of the Chk-1 inhibitor of the invention is the quantity in which a greater response is achieved when the Chk-1 inhibitor is co-administered with the DNA damaging anti-cancer drug and/or radiation therapy compared with when the DNA damaging anti-cancer drug and/or radiation therapy is administered alone. When used as a combination therapy, an "effective amount" of the DNA damaging drug and/or an "effective" radiation dose are administered to the subject, which is a quantity in which anti-cancer effects are normally achieved. The Chk-1 inhibitors of the invention and the DNA damaging anti-cancer drug can be co-administered to the subject as part of the same pharmaceutical composition or, alternatively, as separate pharmaceutical compositions.
When administered as separate pharmaceutical compositions, the Chk-1 inhibitor of the invention and the DNA-damaging anti-cancer drug (and/or radiation therapy) can be administered simultaneously or at different times, provided that the enhancing effect of the Chk-1 inhibitor is retained.
In one embodiment, a compound of the invention as defined herein is administered before (e.g by up to 8 hours or up to 12 hours or up to one day before) administration of the DNA- damaging anticancer drug.
In another embodiment, a compound of the inventin as defined herein is administered after (e.g by up to 8 hours or up to 12 hours or up to 24 hours or up to 30 hours or up to 48 hours after) administration of the DNA-damaging anticancer drug. In another embodiment, a first dose of a compound of the invention as described herein is administered one day after administration of the DNA-damaging anticancer drug and a second dose of the said compound is administered two days after administration of the DNA-damaging anticancer drug.
In a further embodiment, a first dose of a compound of the invention as defined herein is administered one day after administration of the DNA-damaging anticancer drug, a second dose of the said compound is administered two days after administration of the DNA- damaging anticancer drug, and third dose of the said compound is administered three days after administration of the DNA-damaging anticancer drug.
Particular dosage regimes comprising the administration of a compound of the invention as defined herein and a DNA-damaging anticancer drug may be as set out in WO2010/118390 (Array Biopharma), the contents of which are incorporated herein by reference.
The amount of Chk-1 inhibitor compound of the invention and (in the case of combination therapy) the DNA damaging anti-cancer drug and radiation dose administered to the subject will depend on the nature and potency of the DNA damaging anti-cancer drug, the type and severity of the disease or condition and on the characteristics of the subject, such as general health, age, sex, body weight and tolerance to drugs. The skilled person will be able to determine appropriate dosages depending on these and other factors. Effective dosages for commonly used anti-cancer drugs and radiation therapy are well known to the skilled person.
A typical daily dose of Compound 1 , whether administered on its own in monotherapy or administered in combination with a DNA damaging anticancer drug, can be in the range from 100 picograms to 100 milligrams per kilogram of body weight, more typically 5 nanograms to 25 milligrams per kilogram of bodyweight, and more usually 10 nanograms to 15 milligrams per kilogram (e.g. 10 nanograms to 10 milligrams, and more typically 1 microgram per kilogram to 20 milligrams per kilogram, for example 1 microgram to 10 milligrams per kilogram) per kilogram of bodyweight although higher or lower doses may be administered where required. The compound can be administered on a daily basis or on a repeat basis every 2, or 3, or 4, or 5, or 6, or 7, or 10 or 14, or 21, or 28 days for example.
Ultimately, however, the quantity of compound administered and the type of composition used will be commensurate with the nature of the disease or physiological condition being treated and will be at the discretion of the physician.
Methods of Diagnosis
Prior to administration of a compound of the invention, a patient may be screened to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against Chk1.
Accordingly, in further embodiments (8.1 to 8.3), the invention provides:
8.1 A crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for use in the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against a Chk1 kinase.
8.2 The use of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1 , or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24, for the manufacture of a medicament for the treatment or prophylaxis of a disease state or condition in a patient who has been screened and has been determined as suffering from, or being at risk of suffering from, a disease or condition which would be susceptible to treatment with a compound having activity against Chk1 kinase.
8.3 A method for the diagnosis and treatment of a disease state or condition mediated by Chk1 kinase, which method comprises (i) screening a patient to determine whether a disease or condition from which the patient is or may be suffering is one which would be susceptible to treatment with a compound having activity against the kinase; and (ii) where it is indicated that the disease or condition from which the patient is thus susceptible, thereafter administering to the patient an effective Chk1 inhibiting amount of a crystalline form of the compound of formula (1) as defined in any one of Embodiments 1.0 to 4.20 or an amorphous form according to Embodiment 0.1, or a composition of matter as defined in any one of Embodiments 1.23A to1.23D, 2.23A to 2.23D, 3.21-3.24 and 4.21-4.24.
A subject (e.g. patient) may be subjected to a diagnostic test to detect a marker indicative of the presence of a disease or condition in which Chk1 is implicated, or a marker indicative of susceptibility to the said disease or condition. For example, subjects may be screened for genetic markers indicative of a susceptibility to develop an autoimmune or inflammatory disease.
More particularly, a patient may be screened to determine whether a cancer from which the patient is or may be suffering is one which is characterised by a defective DNA repair mechanism or a defective cell cycle, for example a defective cell cycle due to a p53 mutation or is a p53 negative cancer.
Cancers which are characterised by p53 mutations or the absence of p53 can be identified, for example, by the methods described in Allred et al., J. Nat. Cancer Institute, Vol. 85, No.
3, 200-206 (1993) and the methods described in the articles listed in the introductory part of this application. For example, p53 protein may be detected by immuno-histochemical methods such as immuno-staining.
The diagnostic tests are typically conducted on a biological sample selected from tumour biopsy samples, blood samples (isolation and enrichment of shed tumour cells), stool biopsies, sputum, chromosome analysis, pleural fluid, peritoneal fluid, or urine.
In addition to p53, mutations to other DNA repair factors such as RAD17, RAD50, and members of the Fanconi’s anaemia complementation group may be predictive of response to Chk1 inhibitors alone, or in combination with chemotherapy. Cancers which contain mutations in these DNA repair pathways may be identified by DNA sequence analysis of tumor biopsy tissue or circulating tumor DNA (ctDNA) or, in the case of Fanconi’s anaemia, by evaluating DNA foci formation in tumor biopsy specimens using an antibody to FANCD2, as described in Duan et al., Frontiers in Oncology vol.4, 1-8 (2014).
Thus, the crystalline forms of the compound of the invention described herein may be used to treat members of a sub-population of patients who have been screened (for example by testing one or more biological samples taken from the said patients) and have been found to be suffering from a cancer characterised by p53 mutation or a p53 negative cancer, or a cancer containing a RAD17 or RAD50 mutation, or a mutation in a member of the Fanconi’s anaemia complementation group.
Methods of indentifying genetic markers such as single nucleotide polymorphisms are well known. Examples of suitable methods for identifying such markers are described in Ban et al. and Sato et al. above.
Brief Description of the Drawings
Figure 1 shows the chemical structure of Compound 1.
Figure 2 shows a comparison of XRPD Patterns of Compound 1 (free base) with Forms A and B.
Figure 3 shows a Thermal Analysis of Compound 1 (free base).
Figures 4A and 4B are Hot Stage Microscopy Images for Compound 1.
Figure 5 shows a comparison of XRPD Patterns of Compound 1 (free base) with Forms A,
B, C and D and amorphous forms of the compound.
Figure 6 is an overlay of XRPD Pattern of Solids Isolated from 1,4-Dioxane. Figure 7 shows the XRPD Patterns from THF Slurries at 2-8 °C, RT and 50 °C.
Figure 8 shows the XRPD Pattern for Form A of Compound 1 collected with Cu-Ka radiation.
Figure 8A shows the XPRD Pattern for Form A of Compound 1 but with the main peaks labelled.
Figure 9 is an atomic displacement ellipsoid diagram of Form A in which non-hydrogen atoms are represented by 50% probability anisotropic thermal ellipsoids.
Figure 10 is a packing diagram of Compound 1 Form A viewed down the crystallographic a axis.
Figure 11 is a packing diagram of Compound 1 Form A viewed down the crystallographic b axis.
Figure 12 is a packing diagram of Compound 1 Form A viewed down the crystallographic c axis.
Figure 13 indicates hydrogen bonding between two molecules of Compound 1 observed for Form A.
Figure 14 is an overlay of Calculated Pattern and Experimental Pattern for Form A.
Figure 15 shows a thermal analysis of Compound 1 Form A.
Figure 16 shows the Raman Spectrum for Compound 1 Form A.
Figure 17 shows the XRPD Pattern for Form B of Compound 1 collected with Cu-Ka radiation.
Figure 17A shows the XRPD pattern for Form B of Compound 1 but with observed peaks collected with Cu-Ka labelled.
Figure 18 shows a thermal analysis of Compound 1 Form B.
Figure 19 shows the Raman Spectrum for Compound 1 Form B.
Figure 20 is an overlay of the XRPD Patterns of Form A and Form B.
Figure 21 is a comparison of the Raman Spectra of Form A and Form B. Figure 22 is an overlay of the XRPD Patterns of Form C isolated from different solvent systems.
Figure 23 shows the XRPD Pattern for Form C of Compound 1 collected with Cu-Ka radiation.
Figure 24 shows a thermal analysis of Form C (Isolated from NMP/H2O)
Figure 25 shows the XRPD Pattern for Form C of Compound 1 collected with Cu-Ka radiation.
Figure 26 shows a thermal analysis of Form D
Figure 27 shows the XRPD Pattern for amorphous Compound 1.
Figure 28 shows a thermal analysis for amorphous Compound 1.
EXAMPLES
The following non-limiting examples illustrate the synthesis and properties of solid forms of Compound 1.
A. Experimental Methods
1. Estimation of Approximate Solubility
Aliquots of various solvents were added to weighed amounts of Compound 1 (free base) with agitation (typically sonication) at ambient temperature until complete dissolution was achieved, as judged by visual observation. Solubility was estimated based on the total volume of solvent used to provide complete dissolution. The actual solubility may be greater than the value calculated due to the incremental addition of solvent and kinetics of dissolution of the material. The solubility is expressed as “less than” if dissolution did not occur during the experiment. The solubility is expressed as “greater than” if dissolution occurred after the addition of first aliquot.
Figure imgf000040_0001
Figure imgf000041_0001
2. Antisolvent Addition
A solution of Compound 1 (free base) was prepared in a given solvent. The solution was filtered into a clean vial and an anti-solvent was added in order to precipitate solids. Conversely, a solution of Compound 1 (free base) was filtered directly into an anti-solvent solution in selected experiments.
3. Fast Cool
Solutions of Compound 1 (free base) were prepared at elevated temperatures in given solvent or solvent mixture. The resulting solution was hot filtered into a pre-warmed vial. The vial was capped and placed on a bench top at room temperature to quickly cool.
4. Fast Evaporation
Solutions of Compound 1 (free base) were generated at ambient temperature in a given solvent or solvent mixture. The solutions were allowed to evaporate partially or to dryness from an uncapped vial at ambient conditions.
5. Filtration
Unless otherwise specified filtration of samples in non-halogen solvent systems was carried out using positive pressure with a 0.2 pm Nylon filter. Halogenated solvent containing samples were positive pressure filtered using 0.2 pm PTFE filters.
6. Dry Milling
A solid sample of Compound 1 (free base) was placed in an agate jar with a ~4mm agate ball. 20 pL of specified solvent was added and then the sample placed in a Retsch mill at 30 Hz for 10 minutes. The walls of the agate jar were scraped and 20 pL of designated solvent added. Sample was ground in the Retsch mill for another 10 minutes at 30 Hz. Solids were collected.
7. Relative Humidity Tests
Solids were placed in a 1 dram vial and the opening covered with a KimWipe™. The vial was placed inside a 94% relative humidity jar at ambient conditions. Saturated salt solutions were used for maintaining specified relative humidities (Saturated Salt Solutions for Maintaining Specified Relative Humidities" Nyqvist, H. E. Int. J. Pharma. Technol. Prod. Manuf. 1983, 4, 47-48.) 8. Slow Cool
A solution of Compound 1 (free base) was prepared at elevated temperature in a given solvent or solvent mixture. The resulting solution was hot filtered in a pre-warmed vial. The vial was capped and left in a heating block at elevated temperature. The heater then was turned off for the sample to cool down naturally to room temperature.
9. Slow Evaporation
Solutions of Compound 1 (free base) were generated at ambient temperature in a given solvent or solvent mixture. The solutions were filtered and allowed to evaporate partially from a loosely capped vial at ambient conditions.
10. Slurry
Slurry experiments were carried out by making saturated solutions containing excess solid. The suspensions were agitated at a given temperature (ambient, sub-ambient, or elevated) for a specified amount of time. The solids present were recovered by vacuum filtration or positive pressure filtration.
B. Instrumental Techniques
1. Hot Stage Microscopy
Hot stage microscopy was performed using a Linkam hot stage (FTIR 600) mounted on a Leica DM LP microscope equipped with a SPOT Insight™ color digital camera. Temperature calibrations were performed using USP melting point standards. Samples were placed on a cover glass, and a second cover glass was placed on top of the sample. As the stage was heated, each sample was visually observed using a 20x objective with crossed polarizers and a first order red compensator. Images were captured using SPOT software (v. 4.5.9).
2. Karl Fischer (KF)
Coulometric Karl Fischer analysis with Stromboli oven for water determination was performed using a Mettler Toledo DL39 Karl Fischer titrator with a Stromboli oven attachment. A NIST- traceable water standard (Hydranal Water Standard 1.0) was analyzed to check the operation of the coulometer. Additionally, a qualified standard (Apura Water Standard Oven 1%) was analyzed to check the operation of the coulometer/oven system. Approximately 100 mg of sample was weighed in a pre-dried Stromboli vial and sealed. Two samples were weighed and placed into the drying oven set at a pre-selected temperature (e.g. 250 °C). The drying oven was purged into the titrator vessel with dry nitrogen. The samples were then titrated by means of a generator electrode, which produces iodine by electrochemical oxidation: 2I I2 + 2e~.
3. Polarized Light Microscopy (PLM)
Light microscopy was performed using a Leica MZ12.5 stereomicroscope. Samples were observed using 0.8-1 Ox objectives with crossed polarizers and a first order red compensator.
4. Proton Nuclear Magnetic Resonance Spectroscopy (1H NMR)
Solution 1H NMR spectra were acquired with an Avance 600 MHz NMR Spectrometer. Samples were prepared by dissolving in DMSO-d6 containing TMS. The data acquisition parameters are displayed on the first page of the spectrum in the Data section of this report.
5. Raman Spectroscopy
Raman spectrum were acquired on a FT-Raman module interfaced to a Nexus 670 FT-IR spectrophotometer (Thermo Nicolet) equipped with an indium gallium arsenide (InGaAs) detector. Wavelength verification was performed using sulfur and cyclohexane. The sample was prepared for analysis by placing the sample into a glass tube and positioning the tube in a gold- coated tube holder. Approximately 0.3 W of Nd:YVC>4 laser power (1064 nm excitation wavelength) was used to irradiate the sample. The spectrum represents 256 co-added scans collected at a spectral resolution of 4 cm-1.
6. Single Crystal X-ray Diffraction (SCXRD) a. Preparation of the Single Crystal Sample
Single crystals of Form A were obtained from evaporation of an acetonitrile solution. b. Data Collection
A colorless block having approximate dimensions of 0.34 c 0.12 c 0.05 mm3, was mounted on a polymer loop in random orientation. Preliminary examination and data collection were performed on a Rigaku SuperNova diffractometer, equipped with a copper anode microfocus sealed X-ray tube (Cu Ka l = 1.54184 A) and a Dectris Pilatus3 R 200K hybrid pixel array detector. Cell constants and an orientation matrix for data collection were obtained from least- squares refinement using the setting angles of 5634 reflections in the range 5.2460° < Q < 77.3540°. The space group was determined by the program CRYSALISPRO [5] to be P1 (international tables no. 2). The data were collected to a maximum diffraction angle ( 2Q ) of 155.146° at room temperature. c. Data Reduction Frames were integrated with CRYSALISPRO. A total of 8561 reflections were collected, of which 4063 were unique. Lorentz and polarization corrections were applied to the data. The linear absorption coefficient is 0.787 mm-1 for Cu Ka radiation. An empirical absorption correction using CRYSALISPRO was applied. Transmission coefficients ranged from 0.907 to 1.000. A secondary extinction correction was applied. The final coefficient, refined in least- squares, was 0.0042(6) (in absolute units). Intensities of equivalent reflections were averaged. The agreement factor for the averaging was 2% based on intensity. d. Structure Solution and Refinement
The structure was solved by direct methods using SHELXT [6] The remaining atoms were located in succeeding difference Fourier syntheses. The structure was refined using SHELXL- 2014 [7, 6] Hydrogen atoms were refined independently. The structure was refined in full- matrix least-squares by minimizing the function:
S T-N'ί where the weight, w, is defined as M[cF{F0 2) + (0.0628 P)2 +(0.1333 F ], where P= (F0 2 +2Fc 2)/3. Scattering factors were taken from the “International Tables for Crystallography” [8] Of the 4063 reflections used in the refinements, only the reflections with intensities larger than twice their uncertainty [ / > 2s(/) ], 3405, were used in calculating the fit residual, R. The final cycle of refinement included 360 variable parameters, 0 restraints, and converged with respective unweighted and weighted agreement factors of:
Figure imgf000044_0001
The standard deviation of an observation of unit weight (goodness of fit) was 1.08. The highest peak in the final difference Fourier had an electron density of 0.188 e/A3. The minimum negative peak had a value of -0.240 e/A3. e. Calculated X-ray Powder Diffraction (XRPD) Pattern
A calculated XRPD pattern was generated for Cu radiation using MERCURY [7] and the atomic coordinates, space group, and unit cell parameters from the single crystal structure. f. Atomic Displacement Ellipsoid and Packing Diagrams
The atomic displacement ellipsoid diagram was prepared using MERCURY. Atoms are represented by 50% probability anisotropic thermal ellipsoids.
7. Thermogravimetric Analysis/Differential Scanning Calorimetry (TGA/DSC) TGA/DSC analysis was performed using a Mettler-Toledo TGA/DSC3+ analyzer. Temperature and enthalpy adjustments were performed using indium, tin, and zinc, and then verified with indium. The balance was verified with calcium oxalate. The sample was placed in an open aluminum pan. The pan was hermetically sealed, the lid pierced, then inserted into the TG furnace. A weighed aluminum pan configured as the sample pan was placed on the reference platform. The furnace was heated under nitrogen. The data acquisition parameters for the thermogram are displayed in the image in the Data section of this report.
8. X-Ray Powder Diffraction (XRPD)
XRPD patterns were collected with a PANalytical X'Pert PRO MPD or PANalytical Empyrean diffractometer using an incident beam of Cu radiation produced using a long, fine-focus source. An elliptically graded multilayer mirror was used to focus Cu Ka X-rays through the specimen and onto the detector. Prior to the analysis, a silicon specimen (NIST SRM 640e) was analyzed to verify the observed position of the Si 111 peak is consistent with the NIST- certified position. A specimen of the sample was sandwiched between 3-pm-thick films and analyzed in transmission geometry. A beam-stop, short antiscatter extension, and antiscatter knife edge were used to minimize the background generated by air. Soller slits for the incident and diffracted beams were used to minimize broadening and asymmetry from axial divergence. Diffraction patterns were collected using a scanning position-sensitive detector (X'Celerator) located 240 mm from the specimen and Data Collector software v. 5.5.
C. Computational Software and Activities
1. Indexing
Indexing is the process of determining the size and shape of the crystallographic unit cell given the peak positions in a diffraction pattern. The term gets its name from the assignment of Miller index labels to individual peaks. XRPD indexing serves several purposes. If all of the peaks in a pattern are indexed by a single unit cell, this is strong evidence that the sample contains a single crystalline phase. Given the indexing solution, the unit cell volume may be calculated directly and can be useful to determine their solvation states. Indexing is also a robust description of a crystalline form and provides a concise summary of all available peak positions for that phase at a particular thermodynamic state point. Indexing of XRPD pattern was done using proprietary SSCI software. Space groups consistent with the assigned extinction symbol, unit cell parameters, and derived quantities are tabulated in the respective figures providing the indexing solution for each form.
XRPD patterns were indexed using proprietary software1 or X'Pert High-Score Plus 2.2a (2.2.1). Indexing and structure refinement are computational studies. Agreement between the allowed peak positions, marked with red bars, and the observed peaks indicates a consistent unit cell determination. Successful indexing of the pattern indicates that the sample is composed primarily of a single crystalline phase. Space groups consistent with the assigned extinction symbol, unit cell parameters, and derived quantities are tabulated below each figure showing tentative indexing solution. To confirm the tentative indexing solution, the molecular packing motifs within the crystallographic unit cells must be determined. No attempts at molecular packing were performed.
2. Pattern Match
XRPD figures provided were generated using SSCI Pattern Match 3.0.4 software.
NOMENCLATURE, ACRONYMS AND ABBREVIATIONS A. Nomenclature
The following nomenclature was applied: solids that display unique XRPD patterns in comparison to the starting material or are of unknown composition or crystalline phase content are designated as "Material" followed by a sequential capital letter (e.g. Material A, Material B, etc.) applied sequentially. A solid is designated as a "Form" (e.g. Compound 1 Form A) only if its chemical composition is known (e.g. 1H NMR) and it is demonstrated to be a single crystalline phase (e.g. indexing or single crystal data).
B. Acronyms and Abbreviations
Figure imgf000046_0001
Figure imgf000047_0001
Figure imgf000048_0001
RESULTS AND DISCUSSION
A. Characterization of Compound 1 (free base) Compound 1 (free base), 97% purity, as prepared according to the procedure described for Example 64 in WO 2015/120390, was the starting material for screening experiments (Table 1). The starting material was characterized by X-ray powder diffraction (XRPD), solution proton nuclear magnetic spectroscopy (1H NMR), TGA/DSC, hot stage microscopy, and Karl Fischer (KF) analyses. Table 1 - Characterization of Compound 1 (free base), 97% purity
Figure imgf000048_0002
Figure imgf000049_0001
The XRPD pattern showed resolution of peaks consistent with a crystalline material. Compound 1 (free base) is composed of a mixture of two crystalline phases, Form A and Form B (Figure 2). Form A and Form B were isolated as single crystalline phases during the course of this study.
Thermal analysis of Compound 1 (free base) is presented in Figure 3. A weight loss of -0.3% is observed in the TGA data upon heating the sample up to 170 °C. The DSC thermogram exhibits a single endotherm at - 228 °C (peak max), likely due to melt/decomposition based on TGA data. Hot stage microscopy was performed on Compound 1 (free base) (Figure 4). No changes were observed upon heating the sample from room temperature (RT) to 192 °C. At 226.3°C the first sign of melting was observed. Melting was complete at 227.8 °C. The sample did not recrystallize upon cooling of the melt.
The 1H NMR spectrum of Compound 1 (free base) is overall consistent with the structure of Compound 1. Water is observed in the spectrum based on the peak at 3.3 ppm. The water content was determined by KF and found to be 0.2% which suggests the loss observed by TGA is likely due to unbound water on the sample.
Solubility estimates were conducted using an incremental solvent addition method to aid in experimental design. Solubility estimates are presented in Table 2. Table 2 - Approximate Kinetic Solubility Estimates of Compound 1
Figure imgf000049_0002
Figure imgf000050_0001
a Solubilities are calculated based on the total solvent used to give a solution; actual solubilities may be greater because of the volume of the solvent portions utilized or a slow rate of dissolution. Solubilities are rounded to the nearest mg/mL.
Compound 1 (free base) exhibited low solubility (<1 mg/mL) at RT in the majority of solvents tested. Intermediate to good solubility was observed in THF, acetic acid, formic acid, dimethyl acetamide (DMA), dimethyl formamide (DMF), dimethyl sulfoxide (DMSO), N-Methyl-2- pyrrolidone (NMP), 1,1,1,3,3,3-hexafluoro-2- propanol (HFIPA) and 2,2,2-trifluoroethanol (TFE).
B. Preliminary Polymorph and Stable Form Screen Results
A preliminary polymorph and stable form screen of Compound 1 was conducted in an effort to determine the stable form at RT and to evaluate the propensity of the compound to exist in various crystalline forms, including polymorphs, solvates, and hydrates. Approximately thirty-six (36) experiments were performed using different crystallization techniques to vary conditions of nucleation and growth investigating both kinetic and thermodynamic conditions. Crystallization techniques employed included cooling, evaporation, relative humidity stressing, solvent/antisolvent addition, slurry, or a combination of techniques (Table 3 and Table 4). Solids isolated from experiments were typically inspected by polarized light microscopy (PLM) and analyzed by XRPD. XRPD patterns were compared with known forms of Compound 1 and with each other.
Table 3 - Polymorph Screen of Compound 1
Figure imgf000051_0001
Figure imgf000052_0001
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
a. Approx. 70 mg Compound 1 (free base) used as starting material for each experiment unless otherwise note b. Times are approximate
Table 4 - Stable Form Screen of Compound 1
Figure imgf000057_0001
Figure imgf000058_0001
a. Approx. 70 mg starting material and 1 mL solvent used for each experiment, unless otherwise indicated b. Samples were initially heated to 50 °C for ~3 hours prior to slurrying at RT for the stated time b. Times are approximate c. Approx. 100 mg starting material used
1. Preliminary Polymorph Screen
In addition to Form A and Form B, two additional unique crystalline phases, Form C and Form D, as well as X-ray amorphous material were observed during this study. An overlay of Compound 1 materials identified during screening is presented in Figure 5.
Form A as a single crystalline phase was observed from a number of polymorph screening experiments. Form B was only observed as a single crystalline phase during the stable form screen from slurries in THF at RT and 2-8 °C. Form A and Form B are crystalline, anhydrous forms of Compound 1 that are likely enantiotropically related. Form C is likely solvated/hydrated and converts to Form A on drying. Form D is a potential diacetate solvate/salt based on pKa considerations (the calculated pKas for Compound 1 are 11.99 and 7.58 ± 0.1 (ACD11)[1], and the pKa for acetic acid is 4.756) and 1H NMR data.
Solids isolated from slurrying Compound 1 (free base) in dioxane at 70 °C were composed of primarily Form A and contained additional peaks (Figure 6) suggesting the presence of an unknown secondary phase. This sample was solely analyzed by XRPD so further investigation would be needed to confirm the nature of this potential secondary phase.
2. Stable Form Screen
Compound 1 (free base), which is composed of a physical mixture of Form A and Form B, was used as the starting material for slurry experiments. Samples were slurried in various solvents and solvent mixtures at RT in an effort to identify the stable form at RT and evaluate the tendency to form hydrates. The rate of transformation between forms tends to be related to the solubility in that solvent system such that faster transformation kinetics are typically observed in higher solubility solvent systems. Given the low solubility of Compound 1 (free base) observed in the majority of solvents, longer slurry times were used in an effort to overcome potentially slow transformation rates.
The majority of slurry experiments at RT resulted in physical mixtures of Form A and Form
B, with one exception. Solids isolated from slurrying Compound 1 (free base) in THF for 4 weeks at RT resulted in Form B. Based on these results, the relative stability of Form A and
Form B at RT was unclear possibly due to the similarity in energy of the two forms at room temperature. To further investigate the thermodynamic relationship between Form A and
Form B, Compound 1 (free base) was slurried in THF at 2-8 °C and at 50 °C for 2 weeks. Solids isolated after slurrying in THF at 2-8 °C were consistent with Form B, whereas solids isolated from the 50 °C slurry were composed of Form A based on XRPD (Figure 7). Results suggest Form A and Form B are likely enantiotropically related with Form B likely the stable form at 2-8 °C and Form A likely the stable form at 50 °C. Further investigation would be needed to confirm the thermodynamic relationship and the transition temperature or transition temperature range between the two forms.
C. Characterization of Forms and Materials
A summary of the physical characteristics of Forms A-D of Compound 1 and amorphous Compound 1 is provided in Table 5 below. Table 5 - Physical Characterisation of Compound 1 Forms
Figure imgf000060_0001
Figure imgf000061_0001
1. Form A
Form A was obtained under various experimental conditions, including evaporation of an acetonitrile solution, precipitation upon stirring solutions produced by the addition of antisolvents to solutions of Compound 1 (free base), specifically DMA/MTBE, HFIPA/toluene and TFE/IPA solvent/antisolvent additions (Table 3). Form A was also prepared by rotary evaporation of a DMSO/acetone solution and after slurrying Compound (free base) in THF at 50 °C for 2 weeks. Form A, prepared from DMA/MTBE, was characterized by XRPD with indexing, 1H NMR and TGA/DSC analyses (Table 5).
The XRPD pattern of Form A was successfully indexed (Figure 8) indicating the sample is composed primarily or exclusively of a single crystalline phase. Based on considerations of molecular volume, the unit cell volume (998.6 A3/cell) is consistent with the molecule suggesting Form A is likely anhydrous. The anhydrous nature of Form A was confirmed by single crystal structure determination, and the indexing solution showed good agreement with crystallographic parameters for single crystal data of Form A acquired during this study (Figure 8, Table 6).
Table 6 - Crystal Data and Data Collection Parameters for Compound 1 Form A
Empirical formula C21 H22FN7O Formula weight (g mol-1) 407.45
Temperature (K) 299.51(10)
Wavelength (A) 1.54184
Crystal system triclinic Space group P~ Unit cell parameters a = 5.71579(13) A a = 91.5336(19)° b = 10.5063(2) A b = 97.5428(19)° c = 17.0060(4) A Y = 100.2508(19)° Unit cell volume (A3) 994.92(4)
Cell formula units, Z 2 Calculated density (g cm-3) 1.360
Absorption coefficient (mm-1) 0.787
F(000) 428
Crystal size (mm3) 0.34 0.12 0.05
Reflections used for cell measurement 5634 Q range for cell measurement 5.2460°-77.3540° Total reflections collected 8561 Index ranges -7 £ /7 £ 7; -7 £ /C £ 13; -21 £ / £ 21
Q range for data collection 0min = 4.282°, 0max = 77.573° Completeness to @max 95.8% Completeness to QM = 67.684° 99.8% Absorption correction multi-scan Transmission coefficient range 0.907-1.000 Refinement method full matrix least-squares on F2 Independent reflections 4063 [Rim = 0.0200, Ra = 0.0266] Reflections [ />2o(/) ] 3405 Reflections / restraints / parameters 4063 / 0 / 360 Goodness-of-fit on F2 S = 1.08 Final residuals [ />2o(/) ] R = 0.0430, Rw = 0.1207
Final residuals [ all reflections ] R = 0.0496, Rw = 0.1262 Largest diff. peak and hole (e A-3) 0.188, -0.240 Max/mean shift/standard uncertainty 0.001 / 0.000
Single crystals of Form A were obtained from evaporation of an acetonitrile solution and the single crystal structure was successfully determined. The crystal system is triclinic, and the space group is P1- The cell parameters and calculated volume are: a = 5.71579(13) A, b = 10.5063(2) A, c = 17.0060(4) A, a = 91.5336(19)°, b = 97.5428(19)°, g = 100.2508(19)°, V = 994.92(4) A3. The molecular weight is 407.45 g mol-1 with Z = 2, resulting in a calculated density of 1.360 g cm-3. Standard uncertainty in this report is written in crystallographic parenthesis notation, e.g. 0.123(4) is equivalent to 0.123 ± 0.004. The asymmetric unit shown in Figure 9 contains one Compound 1 molecule. Further details of the crystal data and crystallographic data collection parameters are summarized in Table 6.
The quality of the structure obtained is high, as indicated by the fit residual, R, of 0.0430 (4.3%). R-factors in the range 2%-6% are quoted to be the most reliably determined structures. Packing diagrams and hydrogen bonding observed for Form A are presented in Figure 10 - Figure 13. An overlay of the experimental XRPD pattern of Form A with the calculated XRPD pattern is shown in Figure 14 and shows good agreement.
Thermal analysis of Form A is presented in Figure 15. A single endotherm at 226 °C (peak max) is observed in the DSC thermogram, likely due to melt/decomposition based on TGA data. A weight loss of 0.3% from 36 °C to 196 °C is observed in the TGA thermogram.
The 1H NMR spectrum is consistent with the structure of Compound 1. Water is observed in the spectrum based on the presence of the singlet at 3.3 ppm.
The Raman spectrum of Form A was collected as reference (Figure 16).
2. Form B
Form B was prepared by slurrying Compound 1 (free base) in THF for 4 weeks at RT or 2 weeks at 2-8 °C. The XRPD pattern of Form B was successfully indexed (Figure 17) indicating the sample is composed primarily or exclusively of a single crystalline phase. The indexed unit cell volume (992.8 A3/cell) suggests the material is anhydrous/unsolvated based on considerations of molecular volume.
The TGA thermogram exhibited a weight loss of 0.1% upon heating the sample to 151 °C (Figure 18). Overlapping endotherms at 222 °C and 229 °C (peak maxima) are observed in the DSC data and are associated with melt/decomposition of the sample based on the TGA data.
1H NMR data are consistent with the chemical structure of Compound 1. Water is observed in the spectrum based on the presence of the singlet at 3.3 ppm.
The Raman spectrum of Form B was collected as reference (Figure 19). No further characterization of Form B was performed.
Data suggest Form B is a likely anhydrous form of Compound 1.
Overlays of XRPD patterns and Raman spectra for Form A and Form B are presented in Figure 20 and Figure 21.
3. Form C
Crystalline Form C was isolated after the addition of water to solutions of NMP, DMF, or DMSO (Figure 22). Due to sample limitation, analyses were performed on Form C samples generated from different solvent systems.
Solids that precipitated upon addition of water to an NMP solution were characterized by XRPD with indexing, 1H NMR and TGA/DSC analyses. The XRPD pattern of the sample was successfully indexed (Figure 23) and based on considerations of molecular volume, the indexed unit cell volume (1266.8 A3/cell) suggest Form C is likely solvated/hydrated.
The 1H NMR spectrum of the damp sample was overall consistent with the chemical structure of Compound 1. Water and trace amounts of NMP was also observed in the spectrum.
A stepwise weight loss of 37.2% is observed in the TGA data (Figure 24), associated with two broad endotherms at 67 °C and 107 °C (peak maxima), likely attributable to desolvation/dehydration. This is followed by a third endothermic event likely due to the melting of the neat form. To further investigate the thermal behavior observed, Form C isolated from DMSO/water, was dried at 160 °C and analyzed by XRPD. The dried solids were consistent with Form A. The wet sample isolated from DMF/water was also analyzed by 1H NMR spectroscopy. The 1H NMR spectrum was consistent with the chemical structure of Compound 1 and contained approximately 1 mole DMF based on the peak at 7.95 ppm.
Form C was slurried in water for 4 weeks at RT and resulted in Form A. The result suggests Form C is likely not a true hydrate or if it is a hydrate, it is a metastable hydrate.
Form C is likely a solvated/hydrated form of Compound 1 that converts to Form A upon drying and slurrying in water.
4. Form D
Crystalline Form D was prepared from evaporation of an acetic acid solution. The XRPD pattern of the isolated solids was successfully indexed (Figure 25). The indexed unit cell volume (2689.3 A3/cell) indicates quite a bit of extra space in addition to the compound 1 molecule, consistent with a 1:2 acetate solvate/salt. 1H NMR analysis confirms the presence of 2 moles acetic acid/acetate per mole Compound 1 in the sample, consistent with a 1 :2 Compound 1 acetate salt/solvate.
Two broad endotherms at 86 °C and 170 ° C (peak maxima) are observed in the DSC data (Figure 26). The initial endotherm at 86 °C is associated with a stepwise weight loss of 8.2% in the TGA data. Further investigation such as hot stage microscopy, evaluation of the sample upon drying and or TGI R analyses would be needed to better understand the thermal events observed by TGA/DSC. No further analysis of Form D was performed during this study.
Form D is likely a diacetate salt/solvate of Compound 1.
5. X-rav Amorphous
Amorphous material was prepared from evaporation of a TFE solution (Figure 27). An XRPD pattern showing no reflections was also obtained for solids isolated after evaporation of a HFIPA solution. The sample isolated from TFE was characterized by XRPD, 1H NMR and TGA/DSC analyses.
The 1H NMR spectrum is consistent with the structure of the API and contains approx. 1 mole TFE per mole API.
Thermal analysis is presented in Figure 28. The sample exhibits a continuous weight loss upon heating between 45 and 170 °C in the TGA data, likely attributed to the loss of volatiles. The weight loss observed in the TGA data is associated with a stepwise feature at ~70 °C and a broad endotherm at 162 °C (peak max) based on the DSC data. Further investigation of the amorphous material, including modulated DSC analysis to determine the glass transition temperature, would be needed to better understand the thermal events observed.
FUTHER CHARACTERISING DATA OF FORMS A AND B
COMPOUND 1 Free Base Form A
Form A was obtained under various experimental conditions including evaporation of an acetonitrile solution, precipitation upon stirring solutions produced by the addition of antisolvents to solutions of the API specifically DMA/MTBE, HFIPA/toluene and TFE/IPA solvent/antisolvent additions, rotary evaporation of a DMSO/acetone solution and after slurrying Compound 1 free base in THF at ~50 °C for 2 weeks.
Examples of Preparation
1. 7 ml_ acetonitrile was added to 70.06 mg Compound 1 free base resulting in a white suspension. The sample was slurried overnight at ~70 °C. The temperature was then increased to ~80 °C for ~1 hour then decreased back to ~70 °C for ~1 hour. The sample was filtered hot using a 0.2 urn NYL filter into a pre-warmed vial. The clear filtrate was fast cooled to RT and the vial was left open for evaporation. Yellow needle-like agglomerates were produced after 7 days.
2. 70.45 mg Compound 1 free base was dissolved in 0.4 ml_ dimethyl acetamide (DMA) and resulting yellow solution was filtered into a clean vial. Approximately 40 ml_ methyl tert-butyl ether (MTBE) was added to the API solution producing a clear solution. The sample was stirred at room temperature (RT) for approximately 1 day producing a white suspension. The sample was filtered and solids were collected for analysis.
3. 1 ml_ THF was added to 202.6 mg Compound 1 free base resulting in an off white suspension. The sample was slurried at ~50 °C for ~2 weeks. The sample was filtered hot and the isolated solids were collected for analysis.
Compound 1 - Free Base Form B Form B was prepared by slurrying Compound 1 free base in tetrahydrofuran (THF) for 4 weeks at RT or 2 weeks at 2-8 °C.
Examples of Preparation
1. 1 ml_ THF was added to 100.88 mg Compound 1 free base producing an off-white suspension. The sample was slurried at RT for ~13 days then solids were isolated by filtration and analyzed by XRPD. 0.6 ml_ THF was added to the post-analysis solids and the yellow suspension produced was further slurried at RT for ~16 days. The suspension was filtered and the yellow solids collected for analysis.
2. 1 ml_ THF was added to 100.9 mg Compound 1 free base producing a yellow suspension. The sample was slurried at 2-8 °C for ~2 weeks. The sample was filtered cold and the isolated solids were collected for analysis
Instrumental Techniques i) Raman Spectroscopy
The Raman spectrum was acquired on a FT-Raman module interfaced to a Nexus 670 FT- IR spectrophotometer (Thermo Nicolet) equipped with an indium gallium arsenide (InGaAs) detector. Wavelength verification was performed using sulfur and cyclohexane. The sample was prepared for analysis by placing the sample into a glass tube and positioning the tube in a gold-coated tube holder. Approximately 0.301 W of Nd:YVC>4 laser power (1064 nm excitation wavelength) was used to irradiate the sample. The spectrum represents 256 co added scans collected at a spectral resolution of 4 cm-1. ii) X-ray Powder Diffraction (XRPD)
Figures of X-ray powder diffraction patterns were generated using unvalidated software PatternMatch v3.0.4 and are non-cGMP representations.
XRPD patterns were collected with a PANalytical Empyrean diffractometer using an incident beam of Cu radiation produced using a long, fine-focus source. An elliptically graded multilayer mirror was used to focus Cu Ka X-rays through the specimen and onto the detector. Prior to the analysis, a silicon specimen (NIST SRM 640e) was analyzed to verify the observed position of the Si 111 peak is consistent with the NIST-certified position. A specimen of the sample was sandwiched between 3-pm-thick films and analyzed in transmission geometry. A beam-stop, short antiscatter extension, and antiscatter knife edge were used to minimize the background generated by air. Soller slits for the incident and diffracted beams were used to minimize broadening and asymmetry from axial divergence. Diffraction patterns were collected using a scanning position-sensitive detector (X'Celerator) located 240 mm from the specimen and Data Collector software v. 5.5. iii) X-ray Powder Diffraction Peak Identification Process
The range of data collected is typically provided in the form in which the data were initially reported, and is instrument dependent. Under most circumstances, peaks within the range of up to about 30° 2Q were selected. Rounding algorithms were used to round each peak to the nearest 0.1° or 0.01° 2Q, depending upon the instrument used to collect the data and/or the inherent peak resolution. The location of the peaks along the x-axis (° 2Q) in both the figures and the tables were determined using proprietary software (TRIADS™ v2.1) and rounded to one or two significant figures after the decimal point based upon the above criteria. Peak position variabilities are given to within ±0.2° 2Q based upon recommendations outlined in the USP discussion of variability in x-ray powder diffraction (United States Pharmacopeia, USP 41-NF 36, through S2, <941 >, official from 3/1/2018). The accuracy and precision associated with any particular measurement reported herein has not been determined. Moreover, third party measurements on independently prepared samples on different instruments may lead to variability which is greater than ±0.2° 2Q. For d-space listings, the wavelength used to calculate d-spacings was 1.5405929A, the Cu-Kci wavelength ( Phys . Rev. A56 (6) 4554-4568 (1997)). Variability associated with d-spacing estimates was calculated from the USP recommendation, at each d-spacing, and provided in the respective data tables.
Per USP guidelines, variable hydrates and solvates may display peak variances greater than 0.2° 2Q and therefore peak variances of 0.2° 2Q are not applicable to these materials.
For samples with only one XRPD pattern and no other means to evaluate whether the sample provides a good approximation of the powder average, peak tables contain data identified only as "Prominent Peaks". These peaks are a subset of the entire observed peak list. Prominent peaks are selected from observed peaks by identifying preferably non overlapping, low-angle peaks, with strong intensity.
If multiple diffraction patterns are available, then assessments of particle statistics (PS) and/or preferred orientation (PO) are possible. Reproducibility among XRPD patterns from multiple samples analyzed on a single diffractometer indicates that the particle statistics are adequate. Consistency of relative intensity among XRPD patterns from multiple diffractometers indicates good orientation statistics. Alternatively, the observed XRPD pattern may be compared with a calculated XRPD pattern based upon a single crystal structure, if available. Two-dimensional scattering patterns using area detectors can also be used to evaluate PS/PO. If the effects of both PS and PO are determined to be negligible, then the XRPD pattern is representative of the powder average intensity for the sample and prominent peaks may be identified as “Representative Peaks”. In general, the more data collected to determine Representative Peaks, the more confident one can be of the classification of those peaks. “Characteristic peaks”, to the extent they exist, are a subset of Representative Peaks and are used to differentiate one crystalline polymorph from another crystalline polymorph (polymorphs being crystalline forms having the same chemical composition). Characteristic peaks are determined by evaluating which representative peaks, if any, are present in one crystalline polymorph of a compound against all other known crystalline polymorphs of that compound to within ±0.2 °2Q. Not all crystalline polymorphs of a compound necessarily have at least one characteristic peak.
Peak Data for Forms A and B
Table 7 - Observed Peaks for COMPOUND 1 Free Base Form A
Figure imgf000069_0001
Figure imgf000069_0002
Figure imgf000070_0003
Figure imgf000070_0002
Table 8 - Representative Peaks for COMPOUND 1 Free Base Form A
Figure imgf000070_0001
Figure imgf000070_0004
Figure imgf000071_0004
Figure imgf000071_0003
Table 9 - List of Observed Peaks in Raman Spectrum of Compound 1 Free Base Form A
Figure imgf000071_0001
Figure imgf000071_0002
Figure imgf000072_0001
Figure imgf000072_0002
Table 10 - Observed Peaks for COMPOUND 1 Free Base Form B
Figure imgf000072_0003
Figure imgf000072_0004
Figure imgf000073_0005
Figure imgf000073_0006
Table 11 - Prominent Peaks for Compound 1 Free Base Form B
Figure imgf000073_0007
Figure imgf000073_0001
Figure imgf000073_0002
Table 12 - List of Observed Peaks in Raman Spectrum of Compound 1 Free Base Form B
Figure imgf000073_0003
Figure imgf000073_0004
Figure imgf000074_0001
Figure imgf000074_0002
Further characterizing data for the amorphous form and Forms A, B, C and D can be found in US provisional patent applications numbers 62/944,139 (filed on 5 December 2019) and 62/946,223 (filed 10 December 2019), from which the present application claims priority, see for example the following data disclosed in US provisional application number 62/944,139: • The Raman spectra for free base Form A for the ranges 3600-2400 cm-1, 2400-1200 cm-1 and 1200-140 cm-1 on pages 11 to 13;
• The Raman spectra for free base Form B for the ranges 3600-2400 cm-1, 2400-1200 cm-1 and 1200-100 cm 1 on pages 21 to 23; and
• The spectra in Section VIII spanning pages 71 to 192.
Each of the aforementioned disclosures is incorporated herein by reference.
CONCLUSIONS
A stable form and preliminary polymorph screen were conducted on Compound 1.
Compound 1 (free base), which is composed of a physical mixture of Form A and Form B, was used as the starting material for the study. Form A and Form B are likely anhydrous forms of Compound 1 that were isolated as single crystalline phases during this study and characterized. In addition to Form A and Form B, two unique crystalline phases, Form C and Form D, as well as X-ray amorphous material, were discovered during screening. Form C is a potentially solvated/hydrated form of Compound 1 that converts to Form A upon drying and slurrying in water. Form D is a diacetate solvate/salt.
The majority of slurry experiments conducted in an effort to identify the stable form at RT resulted in mixtures of Form A and Form B, with the exception of the slurry in THF, which resulted in Form B. Based on these results the relative stability of Form A and Form B at RT was unclear possibly due to the similarity in energy of the two forms at or about room temperature. To further investigate the thermodynamic relationship between Form A and Form B, additional 2 weeks slurry experiments were performed in THF at 2-8 °C and 50 °C. Solids isolated from the slurry at 2-8 °C were consistent with Form B and solids isolated from the 50 °C slurry were composed of Form A suggesting these forms are likely enantiotropically related.
Equivalents
It will readily be apparent that numerous modifications and alterations may be made to the specific embodiments of the invention described above without departing from the principles underlying the invention. All such modifications and alterations are intended to be embraced by this application.

Claims

1. An amorphous form of a compound having the formula (1):
Figure imgf000076_0001
2. A substantially crystalline form of a compound having the formula (1):
Figure imgf000076_0002
and having a crystalline purity of at least 90%.
3. A substantially crystalline form (Form A) according to claim 2 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4°.
4. A substantially crystalline form (Form A) according to claim 3 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.5° and/or 16.9° and/or 17.4° and/or 20.8° and/or 26.4° and/or 28.4° (±0.2°), and one or more further peaks at diffraction angles (2Q) selected from 9.8°, 17.1° and 17.6° (±0.2°).
5. A substantially crystalline form (Form A) according to any one of claims 2 to 4 which exhibits peaks at the diffraction angles corresponding to those of the X-ray powder diffraction pattern shown in Figure 8.
6. A substantially crystalline form (Form A) according to any one of claims 2 to 5 having an X-ray powder diffraction pattern substantially as shown in Figure 8.
7. A substantially crystalline form (Form A) according to any one of claims 2 to 6 (which has a DSC thermogram characterized by an endotherm with an onset at a temperature of about 224 °C and a peak maximum at about 226 °C.
8. A substantially crystalline form (Form B) according to claim 2 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6° and/or 18.8° and/or 21.5°
9. A substantially crystalline form (Form B) according to claim 8 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 7.6°, 18.8° and 21.5° (±0.2°), and one or more further peaks at diffraction angles (2Q) selected from 15.0°, 15.2° and 29.0° (±0.2°).
10. A substantially crystalline form (Form B) according to any one of claims 2, 8 or 9 which exhibits peaks at the diffraction angles corresponding to those of the X-ray powder diffraction pattern shown in Figure 17.
11. A substantially crystalline form (Form B) according to any one of claims 2 or 8 to 10 having an X-ray powder diffraction pattern substantially as shown in Figure 17.
12. A substantially crystalline form (Form B) according to any one of claims 2 or 8 to 11 which has a DSC thermogram characterized by endotherms with peaks maxima at about 222 °C and/or about 229 °C
13. A substantially crystalline form (Form C) according to claim 2 having an X-ray powder diffraction pattern characterised by the presence of major peaks at about the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0°.
14. A substantially crystalline form (Form C) according to claim 13 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 5.8° and/or 8.7° and/or 26.5° and/or 27.0°, and one or more further peaks at diffraction angles (2Q) selected from about 13.8° and about 16.3°.
15. A substantially crystalline form (Form D) according to claim 2 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9°.
16. A substantially crystalline form (Form D) according to claim 15 having an X-ray powder diffraction pattern characterised by the presence of major peaks at the diffraction angles (2Q) 10.7° and/or 16.6° and/or 25.9°, and one or more further peaks at diffraction angles (2Q) selected from 6.5°, 17.7°, 18.9°, 26.1° and 26.9° (±0.2°).
17. An invention as defined in any one of Embodiment 0.1, 1.0 to 1.26, 2.1 to 2.23D, 3.1 to 3.24, 4.1 to 4.24, 5.1 to 5.2, 6.1 to 6.24, 7.1 to 7.12 or 8.1 to 8.3.
PCT/US2020/063346 2019-12-05 2020-12-04 Amorphous and polymorphic form of a specific chk1 inhibitor WO2021113661A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201962944139P 2019-12-05 2019-12-05
US62/944,139 2019-12-05
US201962946223P 2019-12-10 2019-12-10
US62/946,223 2019-12-10

Publications (1)

Publication Number Publication Date
WO2021113661A1 true WO2021113661A1 (en) 2021-06-10

Family

ID=74104191

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2020/063346 WO2021113661A1 (en) 2019-12-05 2020-12-04 Amorphous and polymorphic form of a specific chk1 inhibitor

Country Status (1)

Country Link
WO (1) WO2021113661A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023226658A1 (en) * 2022-05-25 2023-11-30 Sperogenix Therapeutics Limited Nitrogen-containing five-membered heterocyclic derivatives as checkpoint kinase 1 inhibitor and uses thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010118390A1 (en) 2009-04-11 2010-10-14 Array Biopharma Inc. Checkpoint kinase 1 inhibitors for potentiating dna damaging agents
WO2015120390A1 (en) 2014-02-10 2015-08-13 Oncothyreon Inc. Pharmaceutical compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010118390A1 (en) 2009-04-11 2010-10-14 Array Biopharma Inc. Checkpoint kinase 1 inhibitors for potentiating dna damaging agents
WO2015120390A1 (en) 2014-02-10 2015-08-13 Oncothyreon Inc. Pharmaceutical compounds

Non-Patent Citations (21)

* Cited by examiner, † Cited by third party
Title
ALLRED, J. NAT. CANCER INSTITUTE, vol. 85, no. 3, 1993, pages 200 - 206
ASHWELL ET AL., CLIN. CANCER RES.
ASHWELL ET AL., CLIN. CANCER RES., vol. 14, no. 13, 2008, pages 4032 - 4037
ASHWELL ET AL., EXPERT OPIN. INVESTIG. DRUGS, vol. 17, no. 9, 2008, pages 1331 - 1340
BIOCHEM. BIOPHYS. RES. COMMUN., vol. 406, no. 1, 2011, pages 53 - 8
BLASINA ET AL., MOL. CANCER THER., vol. 7, no. 8, 2008, pages 2394 - 2404
BLOOD, vol. 105, no. 4, 15 February 2005 (2005-02-15), pages 1706 - 16
BLOOD, vol. 112, no. 6, 15 September 2008 (2008-09-15), pages 2439 - 2449
BLOOD, vol. 117, no. 6, 10 February 2011 (2011-02-10), pages 1947 - 57
CANCER RES., vol. 66, no. 16
CLIN CANCER RES, vol. 16, no. 2, 15 January 2010 (2010-01-15)
DUAN ET AL., FRONTIERS IN ONCOLOGY, vol. 4, 2014, pages 1 - 8
HANCOCK, J. PHARM. SCI., vol. 86, 1997, pages 1
MCNEELY, S. ET AL., PHARMACOLOGY & THERAPEUTICS, 2014
MINO R CAIRA ED - MONTCHAMP JEAN-LUC: "CRYSTALLINE POLYMORPHISM OF ORGANIC COMPOUNDS", TOPICS IN CURRENT CHEMISTRY; [TOPICS IN CURRENT CHEMISTRY], SPRINGER, BERLIN, DE, vol. 198, 1 January 1998 (1998-01-01), pages 163 - 208, XP001156954, ISSN: 0340-1022, [retrieved on 19990226], DOI: 10.1007/3-540-69178-2_5 *
NATURE, vol. 444, no. 7, 2006, pages 756 - 760
NYQVIST, H. E.: "Saturated Salt Solutions for Maintaining Specified Relative Humidities", INT. J. PHARMA. TECHNOL. PROD. MANUF., vol. 4, 1983, pages 47 - 48
P. DIMARTINO ET AL., J. THERMAL ANAL., vol. 48, 1997, pages 447 - 458
PHYS. REV., vol. A56, no. 6, 1997, pages 4554 - 4568
RON JENKINSROBERT L. SNYDER: "Introduction to X-ray Powder Diffraction", 1996, JOHN WILEY & SONS
TRENDS IN MOLECULAR MEDICINE, vol. 17, no. 2, February 2011 (2011-02-01)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023226658A1 (en) * 2022-05-25 2023-11-30 Sperogenix Therapeutics Limited Nitrogen-containing five-membered heterocyclic derivatives as checkpoint kinase 1 inhibitor and uses thereof

Similar Documents

Publication Publication Date Title
RU2562245C2 (en) (1S,2S,4R)-4-{4-[(1S)-2,3-DIHYDRO-1H-INDEN-1-YLAMINO]-7H-PYRROLO[2,3-d]PYRIMIDIN-7-YL}-2-HYDROXYCYCLOPENTYL)METHYL SULPHAMATE HYDROCHLORIDE
JP6809681B2 (en) Crystal polymorphism of the free base of 2-hydroxy-6-((2- (1-isopropyl-1H-pyrazole-5-yl) pyridin-3-yl) methoxy) benzaldehyde
CA2793413C (en) Crystal form of edoxaban tosylate monohydrate and method of producing same
US11591345B2 (en) Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo[f]imidazo[1,2-d][1,4]oxazepin-9-yl)amino)propanamide, and methods of production
TW201444834A (en) Solid forms of a selective CDK4/6 inhibitor
EP3347351B1 (en) Salts of a pim kinase inhibitor
JP2020514291A (en) Polymorphic forms of RAD1901-2HCL
CN114555562A (en) Solid state forms of (S) -2- (((S) -6, 8-difluoro-1, 2,3, 4-tetrahydronaphthalen-2-yl) amino) -N- (1- (2-methyl-1- (neopentylamino) propan-2-yl) -1H-imidazol-4-yl) pentanamide and uses thereof
CN111511727A (en) Epidermal growth factor receptor inhibitors
CA2393720C (en) Crystalline 2,5-dione-3-(1-methyl-1h-indol-3-yl)-4-[1-(pyridin-2-ylmethyl)piperidin-4-yl]-1h-indol-3-yl]-1h-pyrrole mono-hydrochloride
WO2021113661A1 (en) Amorphous and polymorphic form of a specific chk1 inhibitor
AU2021281365A1 (en) Solid forms of pralsetinib
US20220402936A1 (en) Crystalline form of a multi-tyrosine kinase inhibitor, method of preparation, and use thereof
CN113677683A (en) Crystalline forms of phosphoinositide 3 kinase (PI3K) inhibitors
WO2022242753A1 (en) Pharmaceutically acceptable salt of pyrazoloheteroaryl derivative and crystal form thereof
TW202313585A (en) Pharmaceutical salts
WO2023041061A1 (en) Fused bicyclic derivative, pharmaceutically acceptable salt, crystal form thereof and preparation method therefor
US10072029B2 (en) Crystalline form of a proteasome inhibitor
TW202342041A (en) Pharmaceutical composition and method for preparing active ingredient compound thereof
WO2022112951A1 (en) Solid state forms of hydrochloride salt of ((1s,2s,4r)-4-{4-[(1s)-2,3-dihydro-1h-inden-1-ylamino]-7h-pyrrolo[2,3-d]pyrimidin-7-yl}-2-hydroxycyclopentyl)methyl sulfamate
CN115515954A (en) Salts and polymorphic forms of 6-chloro-7- (4- (4-chlorobenzyl) piperazin-1-yl) -2- (1,3-dimethyl-1H-pyrazol-4-yl) -3H-imidazo [4,5-B ] pyridine
JP2024059626A (en) Solid Form
WO2023091974A2 (en) Salt and solid forms of (r)-1-(5-methoxy-1 h-indol-1-yl)-n,n-dimethylpropan-2-amine
CN112830930A (en) Crystal form of EOC317 and preparation method and application thereof
CA3041854A1 (en) Crystal of pyrido[3,4-d]pyrimidine derivative or solvate thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 20830428

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 20830428

Country of ref document: EP

Kind code of ref document: A1