Solid forms of diaminocarboxamide compounds, compositions thereof, and methods of use thereof

文档序号:1900105 发布日期:2021-11-30 浏览:20次 中文

阅读说明:本技术 二氨基甲酰胺化合物的固体形式、其组合物及其使用方法 (Solid forms of diaminocarboxamide compounds, compositions thereof, and methods of use thereof ) 是由 安东尼奥·克里斯蒂安·费雷蒂 汉华·曼 亚莱·穆斯尔希丁诺格卢 珍·徐 凯文·辛-英·咏 于 2015-01-29 设计创作,主要内容包括:本文涉及2-(叔丁基氨基)-4-((1R,3R,4R)-3-羟基-4-甲基环己基氨基)-嘧啶-5-甲酰胺的固体形式、其组合物及其使用方法。本文提供了涉及2-(叔丁基氨基)-4-((1R,3R,4R)-3-羟基-4-甲基环己基氨基)-嘧啶-5-甲酰胺(化合物I)的制剂、工艺、固体形式和使用方法。另一方面,本文提供了用于制备某些化合物的方法,包括本文所述的化合物I,以及用于这样的方法的中间体。在某些方面中,化合物I的固体形式用于抑制表达所述激酶的细胞中的激酶,例如JNK1或JNK2。另一方面,化合物I的固体形式用于治疗或预防一种或多种障碍,所述障碍选自间质性肺纤维化、系统性硬化、硬皮病、慢性同种移植肾病、抗体介导的排斥反应或狼疮。(This document relates to solid forms of 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide, compositions thereof, and methods of use thereof. Provided herein are formulations, processes, solid forms and methods of use involving 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide (compound I). In another aspect, provided herein are processes for preparing certain compounds, including compound I described herein, as well as intermediates useful in such processes. In certain aspects, the solid form of compound I is used to inhibit a kinase, e.g., JNK1 or JNK2, in a cell expressing said kinase. In another aspect, the solid form of compound I is for use in the treatment or prevention of one or more disorders selected from the group consisting of interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus.)

1. A crystalline form comprising compound 1 or a tautomer thereof:

it has an X-ray powder diffraction pattern comprising peaks at about 10.55, 13.61, and 19.84 ° 2 Θ.

2. Use of an effective amount of the solid form of claim 1 in the manufacture of a medicament for treating or preventing a disorder treatable or preventable by inhibition of a kinase pathway in a subject in need thereof.

3. Use of an effective amount of the solid form of claim 1 in the manufacture of a medicament for treating or preventing interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus in a subject in need thereof.

4. Use of an effective amount of the solid form of claim 1 in the manufacture of a medicament for treating or preventing a liver fibrosis disorder, diabetes, a metabolic syndrome leading to a liver fibrosis disorder, or a condition treatable or preventable by inhibition of a kinase pathway in a subject in need thereof.

Technical Field

Provided herein are processes for preparing 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide and solid forms of 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide, compositions thereof, methods of use thereof for treating diseases, disorders, or conditions, and solid forms for use in such methods.

Background

The determination and selection of solid forms for a pharmaceutical compound is complicated by the fact that variations in solid forms can affect a variety of physical and chemical properties that can provide benefits or disadvantages in processing, formulation, stability, bioavailability, storage, handling (e.g., transportation) of important pharmaceutical characteristics. Useful pharmaceutical solids include crystalline solids and amorphous solids depending on the product and its mode of administration. Amorphous solids are characterized by a lack of remote structural order, while crystalline solids are characterized by structural periodicity. The desired type of pharmaceutical solid depends on the particular application; amorphous solids are sometimes selected based on, for example, an enhanced dissolution profile, while properties of crystalline solids such as physical or chemical stability may be desirable (see, e.g., s.r. vipbaguta et al, adv. drug. deliv. rev. (2001) 48: 3-26; l.yu, adv. drug. deliv. rev. (2001) 48: 27-42).

Solid forms of pharmaceutical compounds, whether crystalline or amorphous, include single and multi-component solids. Single component solids consist essentially of a pharmaceutical compound or active ingredient without other compounds. The variety of single component crystalline materials can potentially result from polymorphism, where there are a variety of three-dimensional arrangements of specific pharmaceutical compounds (s.r. byrn et al, Solid State Chemistry of Drugs, (1999) SSCI, West Lafayette). RitonavirTMThe case emphasizes the importance of finding polymorphs, ritonavirTMIs an HIV protease inhibitor formulated as a soft gelatin capsule. After about two years of release of the product, the unexpected precipitation of new less soluble polymorphs in the formulation forced the product to be withdrawn from the market until more stable formulations were developed (see s.r. chemburkar et al, org. Process res.dev. (2000) 4: 413-.

Significantly, it is not possible to predict a priori whether crystalline forms of compounds exist, let alone how they are successfully prepared (see, e.g., Braga and Greponi, 2005, "Making crystals from crystals: a green route to crystals engineering and polymorphism," chem. Commun. 3635.: 3645 (in terms of crystal engineering, The result may be unpredictable if The instructions are not very accurate and/or if other external factors influence The process), "Jones et al, 2006, Pharmaceutical crystals: An empirical application to Physical Property engineering," MRS Bulletin 31: mangel 879 (it is currently not possible to computationally predict The number of observable polymorphs of even The simplest molecules), "PRICE, 2004," The prediction of The crystalline forms of compounds, "white route to crystal engineering 875"; polypeptide and polypeptide 319, "Crystal Structure Prediction and polymorphism," ACA Transactions 39: 14-23 (much need to be learned and done, let alone polymorphic forms) before the ability to predict crystal structure can be declared with any degree of confidence).

Compounds chemically designated 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide (alternatively designated 2- [ (1, 1-dimethylethyl) amino ] -4- [ [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl ] amino ] -5-pyrimidinecarboxamide) and tautomers thereof (collectively referred to herein as "compound 1") are disclosed in U.S. patent application publication No. 2013/0029987, published on 31.1.2013, and international publication No. WO2012/145569, each of which is incorporated herein by reference in its entirety.

The variety of possible solid forms creates a potential diversity in the physical and chemical properties of a given pharmaceutical compound. The discovery and selection of solid forms is important in the development of effective, stable and marketable pharmaceutical products.

The link between abnormal protein phosphorylation and the etiology or outcome of the disease is known for more than 20 years. Therefore, protein kinases have become a very important group of drug targets. (see Cohen, Nature, 1: 309-315 (2002), Gaestel et al, curr. Med. chem.14: 2214-223 (2007); Grimmiger et al, Nat. Rev. drug disc.9 (12): 956-970 (2010)). Various protein kinase inhibitors have been used clinically in the treatment of various diseases such as cancer and chronic inflammatory diseases including rheumatoid arthritis and psoriasis. (see Cohen, Eur. J. biochem., 268: 5001-5010 (2001); Protein Kinase Inhibitors for The Treatment of diseases: The reagents and The products, Handbook of Experimental Pharmacology, Springer Berlin Heidelberg, 167 (2005)).

JNK is a widely expressed serine/threonine kinase belonging to the mitogen-activated protein kinase (MAPK) family along with ERK (extracellular regulated kinase) and p 38. (Kyrinakis JM, Sci. STKE (48): pe1 (2000); Whitmarsh AJ et al. Sci. STKE (1): pe1 (1999); Schramek H, News Physiol. Sci.17: 62-7 (2002); Ichijo H, Oncogene 18 (45): 6087-93 (1999)). MAPK is an important mediator of cell surface to nucleus signaling using a phosphorylation cascade to generate a coordinated response by cells to external stimuli generated by phosphorylation of selected intracellular proteins, including transcription factors. In addition, JNK also phosphorylates non-nuclear proteins, such as IRS-1 and Bcl-2 family members. (Davis RJ, Trends biochem. Sci.9 (11): 470-473 (1994); Seger R et al, FASEB J.; 9 (9): 726-35 (1995); Fanger GR et al, curr. Opin. Genet. Dev.; 7 (1): 67-74 (1997)).

The elucidation of the complexity of protein kinase pathways and the complexity of the relationships and interactions among and between various protein kinases and kinase pathways underscores the development of drugs that can act as protein kinase modulators, modulators or inhibitors with beneficial activity on multiple kinases or multiple kinase pathways. Thus, there remains a need for new kinase modulators, such as JNK modulators, particularly solid forms of those kinase modulators.

Citation or identification of any reference in section 2 of this application shall not be construed as an admission that such reference is prior art to the present application.

Summary of The Invention

Provided herein are solid forms of compound 1:

its name is 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide, including its tautomers. Methods of making, isolating, and characterizing the solid forms are also provided.

In another aspect, provided herein are methods of making certain compounds, including compound 1 as described herein, as well as intermediates useful in these methods.

In certain aspects, the solid form of compound 1 is used to inhibit a kinase, e.g., JNK1 or JNK2, in a cell expressing the kinase. In other aspects, solid forms of compound 1 are used to treat or prevent disorders treatable or preventable by inhibition of the JNK pathway, as described herein. In another aspect, the solid form of compound 1 is for use in treating or preventing one or more disorders selected from interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy (chronic allograft nephropathy), antibody-mediated rejection, or lupus. In yet another aspect, the solid form of compound 1 is for use in treating or preventing a liver fibrosis disorder or diabetes and/or a metabolic syndrome leading to a liver fibrosis disorder, as described herein.

The present embodiments may be more completely understood by reference to the detailed description of the invention and the examples, which are intended to illustrate non-limiting embodiments.

Drawings

Figure 1 depicts a superposition of an X-ray powder diffraction (XRPD) pattern of form a (top) and a simulated XRPD pattern (bottom).

Figure 2 depicts the crystal packing diagram and hydrogen bonding diagram of form a.

Fig. 3 depicts a Scanning Electron Microscope (SEM) image of form a.

Figure 4 depicts a thermogravimetric analysis (TGA) thermogram of form a.

Figure 5 depicts the Differential Scanning Calorimetry (DSC) thermogram of form a.

Fig. 6 depicts a dynamic gas phase sorption (DVS) isotherm diagram of form a.

FIG. 7 depicts form A1H Nuclear Magnetic Resonance (NMR) spectrum.

Figure 8 depicts a superposition of XRPD patterns of form a before and after (above and below) DVS.

Figure 9 depicts the XRPD pattern of form a after 1 minute of compression at 2000-psi.

Figure 10 depicts an XRPD pattern for form B.

Figure 11 depicts a TGA thermogram of form B.

Figure 12 depicts the DSC thermogram for form B.

FIG. 13 depicts form B1H NMR spectrum.

Figure 14 depicts an XRPD pattern for form C.

Figure 15 depicts a TGA thermogram of form C.

Figure 16 depicts the DSC thermogram for form C.

FIG. 17 depicts form C 1H NMR spectrum.

Figure 18 depicts an XRPD pattern of form D.

Figure 19 depicts a TGA thermogram of form D.

Figure 20 depicts a DSC thermogram for form D.

FIG. 21 depicts form D1H NMR spectrum.

Figure 22 depicts an XRPD pattern of form E.

Figure 23 depicts a TGA thermogram of form E.

Figure 24 depicts a DSC thermogram for form E.

FIG. 25 depicts form E1H NMR spectrum.

Figure 26 depicts an XRPD pattern of form F.

Figure 27 depicts a TGA thermogram of form F.

Figure 28 depicts a DSC thermogram for form F.

FIG. 29 depicts form F1H NMR spectrum.

Figure 30 depicts an XRPD pattern of form G.

Figure 31 depicts a TGA thermogram of form G.

Figure 32 depicts a DSC thermogram for form G.

FIG. 33 depicts form G1H NMR spectrum.

Figure 34 depicts an XRPD pattern of form H.

Figure 35 depicts a TGA thermogram of form H.

Figure 36 depicts a DSC thermogram for form H.

Fig. 37 depicts a superposition of XRPD patterns of form a, form B, form C, form D, form E, form F, form G, and form H.

Figure 38 depicts an XRPD pattern of form I.

Figure 39 depicts the DSC thermogram for form I.

FIG. 40 depicts form I1H NMR spectrum.

Figure 41 depicts an XRPD pattern of an amorphous solid.

Figure 42 depicts a DSC thermogram for an amorphous solid.

FIG. 43 depicts an amorphous solid1H NMR spectrum.

Fig. 44 depicts liquid chromatography and mass spectrometry of an amorphous solid.

Figure 45 depicts a formal plot of the percentage of form a and form H of compound 1 in DMSO in water as a function of temperature.

Detailed Description

Definition of

As used herein, and in the specification and appended claims, the indefinite articles "a" and "an" and the definite article "the" include plural and singular references unless the context clearly dictates otherwise.

As used herein, and unless otherwise specified, the terms "about" and "approximately," when used with a dose, amount, or weight percentage of an ingredient of a composition or dosage form, refer to a dose, amount, or weight percentage recognized by one of ordinary skill in the art to provide a pharmacological effect equivalent to the pharmacological effect obtained from the specified dose, amount, or weight percentage. In certain embodiments, the term "about" or "approximately" when used in this context contemplates a dose, amount, or weight percent that is within 30%, within 20%, within 15%, within 10%, or within 5% of the specified dose, amount, or weight percent.

As used herein, and unless otherwise specified, the terms "about" and "approximately" when used in conjunction with a numerical value or range of values provided to characterize a particular solid form indicate that the value or range of values may deviate from what is reasonable to one of ordinary skill in the art while still describing a solid form, such as: specific temperatures or temperature ranges, for example, describing melting, dehydrating, desolvating, or glass transition temperatures; mass change, e.g., as a function of temperature or humidity; solvent or moisture content, measured for example in mass or percentage; or peak position, e.g., in, e.g., IR or raman spectroscopy or XRPD analysis. Techniques for characterizing crystalline forms and amorphous solids include, but are not limited to, thermogravimetric analysis (TGA), Differential Scanning Calorimetry (DSC), X-ray powder diffraction (XRPD), single crystal X-ray diffraction, vibrational spectroscopy (e.g., Infrared (IR) and raman spectroscopy, solid state and solution Nuclear Magnetic Resonance (NMR) spectroscopy, optical microscopy, hot stage optical microscopy, Scanning Electron Microscopy (SEM), electron crystallography and quantitative analysis, Particle Size Analysis (PSA), surface area analysis, solubility studies, and dissolution studies in some embodiments, the terms "about" and "approximately" when used in this context indicate that a value or range of values can vary within 30%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1.5%, 1%, 0.5%, or 0.25% of the recited value or range of values, for example, in some embodiments, the values of XRPD peak locations may vary up to ± 0.2 ° 2 θ (or ± 0.2 degrees 2 θ), while still describing a particular XRPD peak.

As used herein, and unless otherwise specified, "pure," i.e., crystalline substantially free of other crystalline or amorphous solids, contains less than about 10% by weight of one or more other crystalline or amorphous solids, less than about 5% by weight of one or more other crystalline or amorphous solids, less than about 3% by weight of one or more other crystalline or amorphous solids, or less than about 1% by weight of one or more other crystalline or amorphous solids.

As used herein, and unless otherwise specified, a "substantially physically pure" solid form is substantially free of other solid forms. In certain embodiments, the substantially physically pure crystalline form comprises less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.05%, or 0.01% of one or more other solid forms on a weight basis. Detection of other solid forms may be accomplished by any method apparent to one of ordinary skill in the art including, but not limited to, diffraction analysis, thermal analysis, elemental combustion analysis, and/or spectroscopic analysis.

As used herein, and unless otherwise specified, a "substantially chemically pure" solid form is substantially free of other chemical compounds (i.e., chemical impurities). In certain embodiments, a substantially chemically pure solid form comprises less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.05%, or 0.01% of one or more other chemical compounds on a weight basis. Detection of other chemical compounds may be accomplished by any method apparent to one of ordinary skill in the art including, but not limited to, chemical analysis methods such as mass spectrometry, spectroscopy, thermal analysis, elemental combustion analysis, and/or chromatography.

As used herein, and unless otherwise specified, "substantially free" of a chemical compound, solid form, or composition of another chemical compound, solid form, or composition means that the compound, solid form, or composition, in certain embodiments, contains less than about 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.05%, or 0.01% by weight of the other compound, solid form, or composition.

As used herein, unless otherwise specified, the terms "solvate" and "solvated" refer to a solid form of a material that comprises a solvent. The terms "hydrate" and "hydrated" refer to a solvate wherein the solvent is water. By "polymorphic form of a solvate" is meant the presence of more than one solid form of a particular solvate composition. Similarly, "polymorph of a hydrate" refers to the presence of more than one solid form of a particular hydrate composition. As used herein, the term "desolvated solvate" refers to a solid form of a substance that can be prepared by removing solvent from a solvate. As used herein, the terms "solvate" and "solvated" may also refer to a solvate of a salt, co-crystal, or molecular complex. As used herein, the terms "hydrate" and "hydrated" may also refer to a hydrate of a salt, co-crystal, or molecular complex.

An "alkyl" group is a saturated, partially saturated or unsaturated, straight or branched chain acyclic hydrocarbon having from 1 to 10 carbon atoms, typically from 1 to 8 carbons, or in some embodiments from 1 to 6, 1 to 4, or 2 to 6, or 2 to 4 carbon atoms. Representative alkyl groups include-methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, and-n-hexyl; and saturated branched alkyl includes-isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, -neopentyl, tert-amyl, -2-methylpentyl, -3-methylpentyl, -4-methylpentyl, 2, 3-dimethylbutyl, and the like. Examples of unsaturated alkyl groups include, but are not limited to, vinyl, allyl, -CH ═ CH (CH), among others3)、-CH=C(CH3)2、-C(CH3)=CH2、-C(CH3)=CH(CH3)、 -C(CH2CH3)=CH2、-C≡CH、-C≡C(CH3)、-C≡C(CH2CH3)、-CH2C≡CH、 -CH2C≡C(CH3) and-CH2C≡C(CH2CH3). Alkyl groups may be substituted or unsubstituted. When the alkyl group described herein is described as "substituted," it can be substituted with any substituent or substituents, such as those present in the exemplary compounds and embodiments disclosed herein, as well as halogen (chlorine, iodine, bromine, or fluorine), alkyl, hydroxyl, alkoxy, alkoxyalkyl, amino, alkylamino, carboxyl, nitro, cyano, mercapto, thioether, imine, imide, amidino, guanidino, enamine, aminocarbonyl, acylamino, phosphonate, phosphino, thiocarbonyl, sulfonyl, sulfone, sulfonamide, keto, aldehyde, ester, urea, urethane, oxime, hydroxylamine, alkoxyamine, aralkoxyamine, N-oxide, hydrazine, hydrazide, hydrazone, azide, isocyanate, isothiocyanate, cyanate ester, urea, urethane, Thiocyanate group, B (OH) 2Or O (alkyl) aminocarbonyl.

"cycloalkyl" is a saturated or partially saturated cyclic alkyl group of 3 to 10 carbon atoms having a single ring or multiple fused or bridged rings, which may be optionally substituted with 1 to 3 alkyl groups. In some embodiments, the cycloalkyl group has 3 to 8 ring members, while in other embodiments the number of ring carbon atoms is 3 to 5, 3 to 6, or 3 to 7. By way of example, such cycloalkyl groups include monocyclic structures such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, 1-methylcyclopropyl, 2-methylcyclopentyl, 2-methylcyclooctyl and the like, or polycyclic or bridged structures such as 1-bicyclo [1.1.1] pentyl, bicyclo [2.1.1] hexyl, bicyclo [2.2.1] heptyl, bicyclo [2.2.2] octyl, adamantyl and the like. Examples of unsaturated cycloalkyl radicals include, inter alia, cyclohexenyl, cyclopentenyl, cyclohexadienyl, butadienyl, pentadienyl, hexadienyl. Cycloalkyl groups may be substituted or unsubstituted. By way of example, such substituted cycloalkyl groups include cyclohexanol and the like.

An "aryl" group is an aromatic carbocyclic group of 6 to 14 carbon atoms having a single ring (e.g., phenyl) or multiple condensed rings (e.g., naphthyl or anthracenyl). In some embodiments, the aryl group contains 6-14 carbons in the ring portion of the group, and in other embodiments 6 to 12 or even 6 to 10 carbon atoms. Specific aryl groups include phenyl, biphenyl, naphthyl, and the like. The aryl group may be substituted or unsubstituted. The term "aryl" also includes groups containing fused rings, such as fused aromatic-aliphatic ring systems (e.g., indanyl, tetrahydronaphthyl, and the like).

"heteroaryl" is an aryl ring system having one to four heteroatoms as ring atoms in a heteroaromatic ring system, wherein the remaining atoms are carbon atoms. In some embodiments, heteroaryl groups contain 3 to 6 ring atoms in the ring portion of the group, and in other embodiments 6 to 9 or even 6 to 10 atoms. Suitable heteroatoms include oxygen, sulfur and nitrogen. In certain embodiments, the heteroaryl ring system is monocyclic or bicyclic. Non-limiting examples include, but are not limited to, groups such as pyrrolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, benzisoxazolyl (e.g., benzo [ d ] isoxazolyl), thiazolyl, pyrrolyl, pyridazinyl, pyrimidinyl, pyrazinyl, thienyl, benzothienyl, furyl, benzofuryl, indolyl (e.g., indol-2-oxo or isoindolin-1-oxo), azaindolyl (pyrrolopyridyl or 1H-pyrrolo [2, 3-b ] pyridyl), indazolyl, benzimidazolyl (e.g., 1H-benzo [ d ] imidazolyl), imidazopyridinyl (e.g., azabenzimidazolyl or 1H-imidazo [4, 5-b ] pyridyl), pyrazolopyridyl, triazolopyridinyl, Benzotriazolyl (e.g., 1H-benzo [ d ] [1, 2, 3] triazolyl), benzoxazolyl (e.g., benzo [ d ] oxazolyl), benzothiazolyl, benzothiadiazolyl, isoxazolopyridinyl, thionaphthyl, purinyl, xanthyl, adenylyl, guanyl, quinolinyl, isoquinolinyl (e.g., 3, 4-dihydroisoquinolin-1 (2H) -onyl), tetrahydroquinolinyl, quinoxalinyl, and quinazolinyl.

"heterocyclyl" is an aromatic (also referred to as heteroaryl) or non-aromatic cycloalkyl group in which one to four ring carbon atoms are independently replaced by a heteroatom from the group consisting of O, S and N. In some embodiments, heterocyclyl includes 3 to 10 ring members, while other such groups have 3 to 5, 3 to 6, or 3 to 8 ring members. The heterocyclyl group may also be bonded to other groups at any ring atom (i.e., at any carbon atom or heteroatom of the heterocycle). Heterocycloalkyl groups may be substituted or unsubstituted. Heterocyclyl includes unsaturated, partially saturated and saturated ring systems, such as imidazolyl, imidazolinyl and imidazolidinyl (e.g., imidazolidin-4-one or imidazolidin-2, 4-dione). The term heterocyclyl includes fused ring classes including those containing fused aromatic and non-aromatic groups, for example, 1-and 2-aminotetralins, benzotriazolyl (e.g., 1H-benzo [ d ] [1, 2, 3] triazolyl), benzimidazolyl (e.g., 1H-benzo [ d ] imidazolyl), 2, 3-dihydrobenzo [1, 4] dioxinyl, and benzo [1, 3] dioxolyl. The term also includes bridged polycyclic ring systems containing heteroatoms, such as, but not limited to, quinuclidinyl. Representative examples of heterocyclyl groups include, but are not limited to, aziridinyl, azetidinyl, azepanyl, oxetanyl, pyrrolidinyl, imidazolidinyl (e.g., imidazolidin-4-keto or imidazolidin-2, 4-dione), pyrazolidinyl, thiazolidinyl, tetrahydrothienyl, tetrahydrofuranyl, dioxolyl, furyl, thienyl, pyrrolyl, pyrrolinyl, imidazolyl, imidazolinyl, pyrazolyl, pyrazolinyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, benzisoxazolyl (e.g., benzo [ d ] isoxazolyl), thiazolyl, thiazolinyl, isothiazolyl, thiadiazolyl, oxadiazolyl, piperidinyl, piperazinyl (e.g., piperazin-2-keto), morpholinyl, thiomorpholinyl, tetrahydropyranyl (e.g., tetrahydro-2H-pyranyl), Tetrahydrothiopyranyl, oxathiahexyl, dithianyl, pyranyl, pyridyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, dihydropyridinyl, dihydrodithiinyl, 1, 4-dioxaspiro [4, 5] decyl, homopiperazinyl, quinolyl, indolyl (e.g., indol-2-onyl or isoindolin-1-onyl), indolinyl, isoindolyl, isoindolinyl, azaindolyl (pyrrolopyridyl or 1H-pyrrolo [2, 3-b ] pyridyl), indazolyl, indolizinyl, benzotriazolyl (e.g., 1H-benzo [ d ] [1, 2, 3] triazolyl), benzimidazolyl (1H-benzo [ d ] imidazolyl or 1H-benzo [ d ] imidazol-2 (3H) -onyl), Benzofuranyl, benzothienyl, benzothiazolyl, benzooxadiazolyl, benzoxazinyl, benzodiazacyclohexenyl, benzoxazolylthiohexenyl, benzothiazinyl, benzoxazolyl (i.e., benzo [ d ] oxazolyl), benzothiazolyl, benzothiadiazolyl, benzo [1, 3] dioxolyl, pyrazolopyridyl (e.g., 1H-pyrazolo [3, 4-b ] pyridyl, 1H-pyrazolo [4, 3-b ] pyridyl), imidazopyridinyl (e.g., azabenzoimidazolyl or 1H-imidazo [4, 5-b ] pyridyl), triazolopyridinyl, isoxazolopyridinyl, purinyl, xanthine, adenine, guanine, quinolyl, isoquinolyl (e.g., 3, 4-dihydroisoquinolin-1 (2H) -keto), Quinolizinyl, quinoxalinyl, quinazolinyl, cinnolinyl, phthalazinyl, naphthyridinyl, pteridinyl, thionaphthyl, dihydrobenzothiazinyl, dihydrobenzofuranyl, dihydroindolyl, dihydrobenzodioxinyl, tetrahydroindolyl, tetrahydroindazolyl, tetrahydrobenzimidazolyl, tetrahydrobenzotriazolyl, tetrahydropyrrolopyridyl, tetrahydropyrazolopyridyl, tetrahydroimidazopyridinyl, tetrahydrotriazolopyridinyl, tetrahydropyrimidin-2 (1H) -one, and tetrahydroquinolinyl. Representative non-aromatic heterocyclic groups do not include fused ring species that include fused aromatic groups. Examples of non-aromatic heterocyclic groups include aziridinyl, azetidinyl, azepinyl, pyrrolidinyl, imidazolidinyl (e.g., imidazolidin-4-keto or imidazolidin-2, 4-dione), pyrazolidinyl, thiazolidinyl, tetrahydrothienyl, tetrahydrofuranyl, piperidinyl, piperazinyl (e.g., piperazin-2-keto), morpholinyl, thiomorpholinyl, tetrahydropyranyl (e.g., tetrahydro-2H-pyranyl), tetrahydrothiopyranyl, oxathioheterocyclohexyl, dithianyl, 1, 4-dioxaspiro [4, 5] decyl, homopiperazinyl, quinazo, or tetrahydropyrimidin-2 (1H) -one. Representative substituted heterocyclyl groups may be mono-or multiply-substituted, such as, but not limited to, pyridyl or morpholinyl, substituted or disubstituted with various substituents, such as those listed below.

"cycloalkylalkyl" is a group of the formula: -alkyl-cycloalkyl, wherein alkyl and cycloalkyl are as defined above. Substituted cycloalkylalkyl groups may be substituted at the alkyl, cycloalkyl, or both alkyl and cycloalkyl portions of the group. Representative cycloalkylalkyl groups include, but are not limited to, methylcyclopropyl, methylcyclobutyl, methylcyclopentyl, methylcyclohexyl, ethylcyclopropyl, ethylcyclobutyl, ethylcyclopentyl, ethylcyclohexyl, propylcyclopentyl, propylcyclohexyl, and the like.

"aralkyl" is a group of the formula: -alkyl-aryl, wherein alkyl and aryl are as defined above. Substituted aralkyl groups may be substituted at the alkyl, aryl, or both alkyl or aryl portions of the group. Representative aralkyl groups include, but are not limited to, benzyl and phenethyl and fused (cycloalkylaryl) alkyl groups, such as 4-ethyl-indanyl.

"Heterocyclylalkyl" is a group of the formula: -alkyl-heterocyclyl, wherein alkyl and heterocyclyl are as defined above. Substituted heterocyclylalkyl groups may be in the radical alkylThe radical, the heterocyclic group, or both the alkyl and heterocyclic groups are partially substituted. Representative heterocyclylalkyl groups include, but are not limited to, 4-ethyl-morpholinyl, 4-propylmorpholinyl, furan-2-ylmethyl, furan-3-ylmethyl, pyridin-3-ylmethyl, tetrahydrofuran-2-ylethyl, and indol-2-ylpropyl. When a group described herein other than alkyl is considered "substituted," it may be substituted with any suitable substituent or substituents. Illustrative examples of substituents are those in the exemplary compounds and embodiments disclosed herein, as well as halogen (chloro, iodo, bromo, or fluoro); an alkyl group; a hydroxyl group; an alkoxy group; an alkoxyalkyl group; an amino group; an alkylamino group; a carboxyl group; a nitro group; a cyano group; a mercapto group; a thioether group; imino; an imide group; an amidino group; guanidino; an alkenylamine group; an aminocarbonyl group; an acylamino group; a phosphonate group; a phosphine group; a thiocarbonyl group; a sulfonyl group; a sulfone group; a sulfonamide group; a ketone group; an aldehyde group; an ester group; a urea group; a urethane group; an oxime group; a hydroxyamino group; an alkoxyamino group; an aralkyloxyamino group; an N-oxide; a hydrazine group; a hydrazide group; a hydrazone group; an azide; an isocyanate group; an isothiocyanate group; a cyanate ester group; a thiocyanate group; oxy (═ O); b (OH) 2(ii) a O (alkyl) aminocarbonyl; cycloalkyl, which may be monocyclic or fused or non-fused polycyclic (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or heterocyclyl, which may be monocyclic or fused or non-fused polycyclic (e.g., pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiazinyl); monocyclic or fused or non-fused polycyclic aryl or heteroaryl (e.g., phenyl, naphthyl, pyrrolyl, indolyl, furyl, thienyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, pyrazolyl, pyridyl, quinolyl, isoquinolyl, acridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, benzimidazolyl, benzothienyl, or benzofuryl); an aryloxy group; an aralkyloxy group; a heterocyclyloxy group; and heterocyclylalkoxy groups.

"halogen" is chlorine, iodine, bromine or fluorine.

"hydroxyalkyl" is an alkyl group as described above substituted with one or more hydroxyl groups.

"alkoxy" is-O- (alkyl) wherein alkyl is as defined above.

"alkoxyalkyl" is- (alkyl) -O- (alkyl), wherein alkyl is as defined above.

The "amine" group being of the formula-NH2A group of (1).

The "hydroxylamine" group is of the formula-N (R) #) OH or-NHOH, wherein R#Is a substituted or unsubstituted alkyl, cycloalkyl, cycloalkylalkyl, aryl, aralkyl, heterocyclyl or heterocyclylalkyl group as defined herein.

The "alkoxyamine" group being of the formula-N (R)#) A group of O-alkyl or-NHO-alkyl, wherein R#As defined above.

The "arylalkoxyamine" group being of the formula-N (R)#) A radical of O-aryl or-NHO-aryl, in which R#As defined above.

"alkylamine" groups are of the formula-NH-alkyl or-N (alkyl)2Wherein each alkyl group is independently as defined above.

"aminocarbonyl" is of the formula-C (═ O) N (R)#)2、-C(=O)NH(R#) or-C (═ O) NH2Wherein each R is#As defined above.

"acylamino" is of the formula-NHC (═ O) (R)#) or-N (alkyl) C (═ O) (R)#) Wherein each alkyl and R#Independently as defined above.

"O (alkyl) aminocarbonyl" is of the formula-O (alkyl) C (═ O) N (R)#)2-O (alkyl) C (═ O) NH (R)#) or-O (alkyl) C (═ O) NH2Wherein each R is#Independently as defined above.

The "N-oxide" group being of the formula-N+-O-A group of (1).

"carboxy" is a group of formula-C (═ O) OH.

The "keto" group being of the formula-C (═ O) (R)#) Wherein R is#As defined above.

An "aldehyde" group is a group of the formula-CH (═ O).

The "ester" group is of the formula-C: (=O)O(R#) or-OC (═ O) (R)#) Wherein R is#As defined above.

The "urea" group being of the formula-N (alkyl) C (═ O) N (R)#)2N (alkyl) C (═ O) NH (R)#) -N (alkyl) C (═ O) NH2、-NHC(=O)N(R#)2、-NHC(=O)NH(R#) or-NHC (═ O) NH2 #Wherein each alkyl and R#Independently as defined above.

An "imino" group is of the formula-N ═ C (R)#)2or-C (R)#)=N(R#) Wherein each R is#Independently as defined above.

An "imide" group is of the formula-C (═ O) N (R #) C (═ O) (R ═ O)#) or-N ((C ═ O) (R)#))2Wherein each R is#Independently as defined above.

The "carbamate" group is of the formula-OC (═ O) N (R)#)2、-OC(=O)NH(R#)、 -N(R#)C(=O)O(R#) or-NHC (═ O) O (R)#) Wherein each R is#Independently as defined above.

An "amidine" group is of the formula-C (═ N (R)#))N(R#)2、-C(=N(R#))NH(R#)、-C(=N(R#))NH2、 -C(=NH)N(R#)2、-C(=NH)NH(R#)、-C(=NH)NH2、-N=C(R#)N(R#)2、-N=C( R#)NH(R#)、-N=C(R#)NH2、-N(R#)C(R#)=N(R#)、-NHC(R#)=N(R#)、-N(R#) C(R#) (R) NH or-NHC#) A group of NH wherein each R#Independently as defined above.

The "guanidine" group being of the formula-N (R)#)C(=N(R#))N(R#)2、-NHC(=N(R#))N(R#)2、 -N(R#)C(=NH)N(R#)2、-N(R#)C(=N(R#))NH(R#)、-N(R#)C(=N(R#))NH2、 -NHC(=NH)N(R#)2、-NHC(=N(R#))NH(R#)、-NHC(=N(R#))NH2、-NHC(=NH)NH(R#)、-NHC(=NH)NH2、-N=C(N(R#)2)2、-N=C(NH(R#))2or-N ═ C (NH)2)2Wherein each R is#Independently as defined above.

The "enamine" group being of the formula-N (R)#)C(R#)=C(R#)2、-NHC(R#)=C(R#)2、 -C(N(R#)2)=C(R#)2、-C(NH(R#))=C(R#)2、-C(NH2)=C(R#)2、 -C(R#)=C(R#)(N(R#)2)、-C(R#)=C(R#)(NH(R#) or-C (R)#)=C(R#)(NH2) Wherein each R is#Independently as defined above.

An "oxime" group is of the formula-C (═ NO (R)#))(R#)、-C(=NOH)(R#)、-CH(=NO(R#) or-CH (═ NOH), where each R is#Independently as defined above.

The "hydrazide" group is of the formula-C (═ O) N (R)#)N(R#)2、-C(=O)NHN(R#)2、 -C(=O)N(R#)NH(R#)、-C(=O)N(R#)NH2、-C(=O)NHNH(R#)2or-C (═ O) NHNH 2Wherein each R is#Independently as defined above.

The "hydrazine" group being of the formula-N (R)#)N(R#)2、-NHN(R#)2、-N(R#)NH(R#)、-N(R#)NH2、 -NHNH(R#)2or-NHNH2Wherein each R is#Independently as defined above.

The "hydrazone" group is of the formula-C (═ C)N-N(R#)2)(R#)2、-C(=N-NH(R#))(R#)2、-C(=N-NH2)(R#)2、 -N(R#)(N=C(R#)2) or-NH (N ═ C (R)#)2) Wherein each R is#Independently as defined above.

The "azido" group being of the formula-N3A group of (1).

An "isocyanate" group is a group of the formula-N ═ C ═ O.

An "isothiocyanate" group is a group of the formula-N ═ C ═ S.

The "cyanate ester" group is a group of formula-OCN.

The "thiocyanate" group is a group of the formula-SCN.

"thioether" group is of the formula-S (R)#) Wherein R is#As defined above.

"Thiocarbonyl" is of the formula-C (═ S) (R)#) Wherein R is#As defined above.

"sulfinyl" is of the formula-S (═ O) (R)#) Wherein R is#As defined above.

The "sulfone" group being of the formula-S (═ O)2(R#) Wherein R is#As defined above.

"Sulfonylamino" is of the formula-NHSO2(R#) or-N (alkyl) SO2(R#) Wherein each alkyl and R#As defined above.

The "sulfonamide" group being of the formula-S (═ O)2N(R#)2or-S (═ O)2NH(R#) or-S (═ O)2NH2Wherein each R is#Independently as defined above.

A "phosphonate" group is of the formula-P (═ O) (O (R)#))2、-P(=O)(OH)2、-OP(=O)(O(R#))(R#) or-OP (═ O) (OH) (R) #) Wherein each R is#Independently as defined above.

The "phosphine" group beingformula-P (R)#)2Wherein each R is#Independently as defined above.

"tautomer" refers to the isomeric forms of a compound that are in equilibrium with each other. The concentration of the isomeric forms will depend on the environment in which the compound is placed and may vary depending on, for example, whether the compound is a solid or an organic or aqueous solution. For example, in aqueous solution, pyrazoles may exhibit the following isomeric forms, which are referred to as tautomers of each other:

as one skilled in the art readily understands, various functional groups and other structures may exhibit tautomerism, and all tautomers of compound 1 are within the scope of the present invention.

The term "composition" as used herein, unless otherwise specified, is intended to encompass a product comprising the specified ingredient(s) (and if indicated, in the specified amount (s)), as well as any product which results, directly or indirectly, from combination of the specified ingredient(s). By "pharmaceutically acceptable" it is meant that the diluent, excipient or carrier in the formulation must be compatible with the other ingredient(s) of the formulation and not deleterious to the recipient thereof.

The term "solid form" refers to a physical form that is not primarily a liquid or gas. As used herein and unless otherwise specified, when the term "solid form" is used herein to refer to compound 1, it is meant to encompass physical forms of compound 1 that are not primarily liquid or gaseous. The solid form may be a crystalline form or a mixture thereof. In certain embodiments, the solid form is also a liquid crystal. In certain embodiments, the term "solid form comprising compound 1" includes a crystalline form comprising compound 1. In certain embodiments, the solid form of compound 1 is form a, form B, form C, form D, form E, form F, form G, form H, form I, an amorphous solid, or a mixture thereof.

As used herein and unless otherwise specified, the term "crystalline" when used in reference to a compound, substance, modification, material, component, or product means that the compound, substance, modification, material, component, or product is substantially crystalline, as determined by X-ray diffraction. See, e.g., Remington: the Science and Practice of Pharmacy, 21 st edition, Lippincott, Williams and Wilkins, Baltimore, MD (2005); the United States Pharmacopeia, 23 rd edition, 1843-1844 (1995).

The term "crystalline form" or "crystalline form" refers to a solid form that is crystalline. In certain embodiments, the crystalline form of the substance may be substantially free of amorphous solids and/or other crystalline forms. In certain embodiments, the crystalline form of a substance may comprise less than about 1%, less than about 2%, less than about 3%, less than about 4%, less than about 5%, less than about 6%, less than about 7%, less than about 8%, less than about 9%, less than about 10%, less than about 15%, less than about 20%, less than about 25%, less than about 30%, less than about 35%, less than about 40%, less than about 45%, or less than about 50% by weight of one or more amorphous solids and/or other crystalline forms. In certain embodiments, the crystalline form of the substance may be physically and/or chemically pure. In certain embodiments, the crystalline form of a substance may be about 99%, about 98%, about 97%, about 96%, about 95%, about 94%, about 93%, about 92%, about 91%, or about 90% physically and/or chemically pure.

Unless otherwise specified, the terms "amorphous" or "amorphous solid" mean that the material, component, or product in question is not substantially crystalline as determined by X-ray diffraction. In particular, the term "amorphous solid" describes a disordered solid form, i.e. a solid form lacking a remote crystalline order. In certain embodiments, the amorphous solid of matter may be substantially free of other amorphous solids and/or crystalline forms. In certain embodiments, an amorphous solid of a substance may comprise less than about 1%, less than about 2%, less than about 3%, less than about 4%, less than about 5%, less than about 10%, less than about 15%, less than about 20%, less than about 25%, less than about 30%, less than about 35%, less than about 40%, less than about 45%, or less than about 50% by weight of one or more other amorphous solids and/or crystalline forms on a weight basis. In certain embodiments, the amorphous solid of matter may be physically and/or chemically pure. In certain embodiments, the amorphous solid of a substance is about 99%, about 98%, about 97%, about 96%, about 95%, about 94%, about 93%, about 92%, about 91%, or about 90% physically and/or chemically pure.

"JNK" refers to a protein expressed by JNK1, JNK2, or JNK3 gene or isoforms thereof (Gupta, S., Barrett, T., Whitmarsh, A.J., Cavanagh, J., Sluss, H.K., Derijard, B. and Davis, R.J.the EMBO J.15: 2760-.

As used herein, "treating" or "treatment" refers to a complete or partial reduction of a disorder, disease, or condition provided herein or one or more symptoms associated with the disorder, disease, or condition, or a cessation of further progression or worsening of those symptoms, or a reduction or eradication of the cause of the disorder, disease, or condition itself. In one embodiment, the disorder is a condition that can be treated or prevented by inhibiting the JNK pathway, as described herein. In another embodiment, the disorder is selected from interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus. In yet another embodiment, the disorder is liver fibrosis disorder or diabetes and/or metabolic syndrome leading to liver fibrosis disorder, as described herein. In some embodiments, the disorder is a liver fibrosis disorder, such as non-alcoholic steatohepatitis, steatosis (i.e., fatty liver), cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular carcinoma or liver fibrosis with chronic or repeated alcohol intake (alcoholic hepatitis), with infection (e.g., viral infection such as HCV), with liver transplantation, or with drug-induced liver injury (e.g., acetaminophen toxicity). In some embodiments, "treating" or "treatment" refers to a complete or partial reduction in the disorder, disease or condition or symptoms associated with diabetes or a metabolic syndrome that results in liver fibrosis disorders, such as non-alcoholic steatohepatitis, steatosis (i.e., fatty liver), hepatitis, or cirrhosis, or a slowing or stopping of further progression or worsening of those symptoms. In one embodiment, the symptom is jaundice.

As used herein, "preventing" refers to delaying and/or preventing, in whole or in part, the onset, recurrence, or spread of a disorder, disease, or condition; preventing the subject from acquiring the disorder, disease, or condition; or reducing the risk of acquiring a disorder, disease or condition in a subject. In one embodiment, the disorder is a condition that can be treated or prevented by inhibiting the JNK pathway, as described herein. In another embodiment, the disorder is selected from interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus. In one embodiment, the disorder is a liver fibrosis disorder or diabetes or metabolic syndrome leading to a liver fibrosis disorder as described herein, or a symptom thereof.

The term "effective amount" in combination with a solid form of compound 1 refers to an amount that is capable of treating or preventing the disorders, diseases, or conditions disclosed herein, or symptoms thereof.

A "patient" or "subject" is defined herein to include animals, such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, monkeys, chickens, turkeys, quail or guinea pigs, and the like, in one embodiment mammals, and in another embodiment humans. In one embodiment, the subject is a human having or at risk of having: interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus. In another embodiment, the subject is a human having or at risk of having: a liver fibrosis disorder or diabetes or a metabolic syndrome leading to a liver fibrosis disorder, or a disorder that can be treated or prevented by inhibition of the JNK pathway, or a symptom thereof.

Compound 1

The solid forms, formulations, and methods of use provided herein relate to a solid form (e.g., polymorph) of compound 1:

said compound 1 has the alternative name 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide or 2- [ (1, 1-dimethylethyl) amino ] -4- [ [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl ] amino ] -5-pyrimidinecarboxamide, including tautomers thereof.

In another aspect, provided herein are methods of making specific compounds, including compound 1 as described herein, as well as intermediates useful in such methods.

Compound 1 can be prepared using reagents and methods known in the art, including the methods provided in U.S. patent application publication No. 2013/0029987 published on 31.1.2013 and international patent application publication No. WO2012/145569, each of which is incorporated herein by reference in its entirety.

It should be noted that if there is a discrepancy between a depicted structure and the name of that structure, the depicted structure is more pertinent. Further, if the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold or dashed lines, the structure or portion of the structure should be interpreted as encompassing all stereoisomers of it.

Process for preparing compound 1

By way of example, and not limitation, the diaminopyrimidine compounds of formula (iv) may be prepared as outlined in scheme 1 shown below and in the examples described herein.

Scheme 1

In certain embodiments of formula (iv), R2Is substituted or unsubstituted C1-8Alkyl, or substituted or unsubstituted saturated cycloalkyl. In certain embodiments of formula (iv), R1Is substituted or notSubstituted C1-8Alkyl, or substituted or unsubstituted cycloalkyl.

In some embodiments, R2Is (1R, 3R, 4R) -3-hydroxy-4-methyl-cyclohexyl, tert-butyl or 1-bicyclo [1.1.1]And (4) pentyl.

In some embodiments, R2Is composed of

In some embodiments, R1Is tert-butyl, trans-4-hydroxy-cyclohexyl or (1R, 3S) -3-hydroxy-cyclohexyl.

In some embodiments, R1Is composed of

In one embodiment, the compound of formula (iv) is compound 1.

Use of R in a solvent (e.g., Tetrahydrofuran (THF), N-methyl-2-pyrrolidone (NMP), or water) in the presence of a base (e.g., diisopropylethylamine, potassium carbonate, dipotassium hydrogen phosphate, tripotassium phosphate, or sodium bicarbonate) at about 0 ℃ to about 25 ℃2NH2(ii) Treatment of 2, 4-dichloropyrimidine-5-carboxamide (i) to provide R 2Introduction of a side chain to produce a compound of formula (iii). By subsequent reaction at elevated temperature (e.g., about 60 ℃ to about 85 ℃), optionally under nitrogen pressure, in the presence of a base (e.g., tert-butylamine or sodium carbonate) or a Lewis acid (e.g., ZnCl)2) Using R in an organic solvent (e.g., acetonitrile, EtOAc, THF, NMP, dimethyl sulfoxide (DMSO), or sulfolane) in the presence of (C)1NH2Treatment further derivatizes the desired regioisomer (regioisomer) compound to provide R1Introduction of a side chain to produce a compound of formula (iv). Recrystallization of the compound of formula (iv) in a solvent system (e.g., 2-propanol/water or ethanol/water) provides the compound of formula (iv) with improved purity.

In one aspect, provided herein is a method of preparing a compound of formula (iv):

said method comprising reacting a compound of formula (iii)

With R in the presence of a base or a Lewis acid in a solvent1NH2Contacting;

wherein R is1Is substituted or unsubstituted C1-8Alkyl, or substituted or unsubstituted saturated cycloalkyl; and

R2is substituted or unsubstituted C1-8Alkyl, or substituted or unsubstituted saturated cycloalkyl.

In some embodiments, the solvent is DMSO, sulfolane, acetonitrile, DMF, DMAc, NMP, EtOH, n-PrOH, IPA, n-BuOH, t-BuOH, EtOAc, IPAc, toluene, 2-MeTHF, THF, DCM, or a mixed solvent such as: THF/water, THF/NMP, sulfolane/water, DMSO/water, IPA/water, EtOH/water. In some embodiments, the solvent is acetonitrile, EtOAc, THF, NMP, DMSO, or sulfolane.

In some embodiments, the base is N, N-diisopropylethylamine, DBU, triethylamine, tert-butylamine, sodium carbonate, potassium carbonate, sodium bicarbonate, sodium acetate, or potassium phosphate. In some embodiments, the base is tert-butylamine or sodium carbonate.

In some embodiments, the lewis acid is ZnCl2、ZnBr2、AlCl3、Zn(OTf)2. In some embodiments, the lewis acid is ZnCl2

In some embodiments, the contacting is performed at an elevated temperature, e.g., about 60 ℃ to about 85 ℃.

In some embodiments, the contacting is performed under nitrogen pressure.

In some embodiments, the method further comprises preparing a compound of formula (iii)

The process comprises reacting 2, 4-dichloropyrimidine-5-carboxamide (i) with R in a solvent in the presence of a base2NH2(ii) And (4) contacting.

In some embodiments, the solvent is THF, NMP, water, or a mixed solvent, such as THF/water or NMP/water. In one embodiment, the solvent is THF, NMP, or THF/water. In some embodiments, the base is N, N-diisopropylethylamine, potassium carbonate, dipotassium hydrogen phosphate, tripotassium phosphate, or sodium bicarbonate. In some embodiments, the base is N, N-diisopropylethylamine, potassium carbonate, or sodium bicarbonate. In some embodiments, the contacting is performed at about 0 ℃ to about 25 ℃.

In one aspect, provided herein is a method of purifying a compound of formula (iv):

wherein R is1Is substituted or unsubstituted C1-8Alkyl, or substituted or unsubstituted cycloalkyl; and

R2is substituted or unsubstituted C1-8Alkyl, or substituted or unsubstituted cycloalkyl,

the process comprises 1) dissolving a compound of formula (iv) in a first solvent at a first temperature; 2) adding a second solvent to the resulting solution; 3) cooling the solution to a second temperature; and 4) collecting the solid.

In some embodiments, the method further comprises seeding with form a. In certain embodiments, the method further comprises seeding with form a after step 2) and before step 3). In certain embodiments, the method further comprises seeding with form a during step 3). In certain embodiments, the method further comprises seeding with form a after step 3) and before step 4). In some such embodiments, form a is micronized. In certain embodiments, the method further comprises seeding with micronized form a after step 2) and before step 3).

In some embodiments, the first solvent is: i) a mixture of 2-propanol and water (e.g., wherein the volume ratio of 2-propanol and water in the mixture is about 3: 1); ii) DMSO; or iii) ethanol.

In some embodiments, the second solvent is water.

In some embodiments, the first temperature is about 60 ℃ to about 70 ℃.

In some embodiments, the second temperature is from about 0 ℃ to about 25 ℃.

Provided herein are compounds having the following formula (iii):

and a tautomer thereof, and the tautomers thereof,

wherein R is2Is substituted or unsubstituted C1-8Alkyl, or substituted saturated cycloalkyl.

In certain embodiments of formula (iii), R2Is (1R, 3R, 4R) -3-hydroxy-4-methyl-cyclohexyl, tert-butyl or 1-bicyclo [1.1.1]And (4) pentyl.

In certain embodiments of formula (iii), R2Is composed of

In one embodiment, provided herein is a method of making compound 1 as illustrated in scheme 2 shown below and in the examples described herein.

Scheme 2

In one embodiment, treatment of 2, 4-dichloropyrimidine-5-carboxamide (i) with (1R, 2R, 5R) -5-amino-2-methylcyclohexanol hydrochloride (v) in the presence of potassium carbonate in THF at about 0 ℃ to about 25 ℃ provides for the introduction of the (1R, 2R, 5R) -5-amino-2-methylcyclohexanol side chain to produce compound (vi). Followed by the use of t-BuNH in DMSO at about 68 deg.C2Treatment or ZnCl in ACN2Using t-BuNH in the presence of2Treatment provides t-BuNH 2Introduction of a side chain to give compound 1. Recrystallization of compound 1 from a mixture of IPA and water at about 70 ℃ provides compound 1 with improved purity.

In one aspect, provided herein is a method of making a compound of formula (a):

the method comprises reacting a compound of formula (9 a):

contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is methanol, 2-propanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 a):

the process comprises separating a diastereomeric mixture of compounds of formula (9a and 9b) by chiral separation:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC). In one embodiment, the diastereomeric mixture is a 1: 1 mixture.

In some embodiments, the method further comprises preparing a diastereomeric mixture of compounds of formulae (9a and 9 b):

the method comprises reacting a compound of formula (8):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent in the presence of a base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane ((+) -diisopinocampheylborane). In one embodiment, the hydroboration agent is BH3THF. In one embodiment, the oxidizing agent is H2O2Or oxone. In another embodiment, the oxidizing agent is H2O2. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF. In yet another embodiment, the base is NaOH.

In some embodiments, the method further comprises preparing a compound of formula (8):

the method comprises reacting a compound of formula (7):

optionally with Boc in the presence of a base in an organic solvent2And (4) contacting with O. In one embodiment, the organic solvent is DCM or an ether. In one embodiment, the base is triethylamine.

In some embodiments, the method further comprises preparing a compound of formula (7):

the method comprises reacting a compound of formula (6):

contacting with an azide in an organic solvent, followed by reduction of the resulting azide derivative in an organic solvent.

In one embodiment, the azidation agent is NaN3. In another embodiment, the reducing agent is LiAlH 4. In some embodiments, the solvent is selected from DMF, toluene, ACN, DCM, THF, or an ether.

In some embodiments, the method further comprises preparing a compound of formula (6):

the method comprises reacting a compound of formula (5):

in the presence of a base, with p-toluenesulfonyl chloride in an organic solvent.

In some embodiments, the organic solvent is selected from DMF, toluene, ACN, DCM, THF, or an ether. In other embodiments, the base is triethylamine or pyridine.

In some embodiments, the method further comprises preparing a compound of formula (5):

the method comprises reacting a compound of formula (4):

in a solvent with a reducing agent.

In some embodiments, the reducing agent is LiAlH4. In other embodiments, the solvent is THF or an ether.

In some embodiments, the method further comprises preparing a compound of formula (4):

the method comprises reacting a compound of formula (3):

with Zn and NaI in the presence of acetic acid.

In some embodiments, the method further comprises preparing a compound of formula (3):

the method comprises reacting a compound of formula (2):

in a solvent with a peracid.

In some embodiments, the peracid is m-CPBA. In other embodiments, the solvent is DCM.

In some embodiments, the method further comprises preparing a compound of formula (2):

the method comprises ozonolysis of a compound of formula (Y) in the presence of ozone:

in some embodiments, the method further comprises preparing a compound of formula (Y):

the method comprises reacting (-) -limonene ((-) -limonene) having the formula:

in a solvent with a peracid.

In some embodiments, the peracid is m-CPBA. In other embodiments, the solvent is DCM.

In one aspect, provided herein is a process for preparing a compound of formula (10):

the method comprises reacting a compound of formula (9 b):

contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is 2-propanol, methanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 b):

the process comprises separating a diastereomeric mixture of compounds of formula (9a and 9b) by employing a chiral separation process:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC). In one embodiment, the diastereomeric mixture is a 1: 1 mixture.

In some embodiments, the method further comprises preparing a diastereomeric mixture of compounds of formulae (9a and 9 b):

the method comprises reacting a compound of formula (8):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent in the presence of a base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane. In one embodiment, the hydroboration agent is BH3THF. In one embodiment, the oxidizing agent is H2O2Or potassium hydrogen persulfate. In another embodiment, the oxidizing agent is H2O2. In yet another embodiment, the base is NaOH. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF.

In some embodiments, the method further comprises preparing a compound of formula (8):

the method comprises reacting a compound of formula (7):

optionally with Boc in the presence of a base in an organic solvent2And (4) contacting with O. In one embodiment, the organic solvent is DCM or an ether. In one embodiment, the base is triethylamine.

In some embodiments, the method further comprises preparing a compound of formula (7):

the method comprises reacting a compound of formula (6):

contacting with an azide in an organic solvent, followed by reduction of the resulting azide derivative in an organic solvent.

In one embodiment, the azidation agent is NaN3. In another embodiment, the reducing agent is LiAlH4. In some embodiments, the solvent is selected from DMF, toluene, ACN, DCM, THF, or an ether.

In some embodiments, the method further comprises preparing a compound of formula (6):

the method comprises reacting a compound of formula (5):

in the presence of a base, with p-toluenesulfonyl chloride in an organic solvent.

In some embodiments, the organic solvent is selected from DMF, toluene, ACN, DCM, THF, or an ether. In other embodiments, the base is triethylamine or pyridine.

In some embodiments, the method further comprises preparing a compound of formula (5):

the method comprises reacting a compound of formula (4):

in a solvent with a reducing agent.

In some embodiments, the reducing agent is LiAlH4. In other embodiments, the solvent is THF or an ether.

In some embodiments, the method further comprises preparing a compound of formula (4):

the method comprises reacting a compound of formula (3):

with Zn and NaI in the presence of acetic acid.

In some embodiments, the method further comprises preparing a compound of formula (3):

the method comprises reacting a compound of formula (2):

In a solvent with a peracid.

In some embodiments, the peracid is m-CPBA. In other embodiments, the solvent is DCM.

In some embodiments, the method further comprises preparing a compound of formula (2):

the method comprises ozonolysis of a compound of formula (Y) in the presence of ozone:

in some embodiments, the method further comprises preparing a compound of formula (Y):

the method comprises reacting (-) -limonene having the formula:

in a solvent with a peracid.

In some embodiments, the peracid is m-CPBA. In other embodiments, the solvent is DCM.

In one aspect, provided herein is a process for preparing a compound of formula (a):

the method comprises reacting a compound of formula (9 a):

contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is 2-propanol, methanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 a):

the process comprises separating a diastereomeric mixture of compounds of formula (9a and 9b) by employing a chiral separation process:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), chiral HPLC, chiral LC, recrystallization, or chiral resolution. In one embodiment, the chiral separation method is recrystallization from a solvent, and the solvent is MTBE.

In some embodiments, the method further comprises preparing a diastereomeric mixture of compounds of formulae (9a and 9 b):

the method comprises reacting a compound of formula (8):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent under an aqueous base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane. In one embodiment, the hydroboration agent is (+) -diisopinocampheylborane. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF. In some embodiments, the oxidizing agent is H2O2Or potassium hydrogen persulfate. In some embodiments, the oxidizing agent is H2O2. In other embodiments, the base is NaOH.

In some embodiments, the method further comprises preparing a compound of formula (8):

the method comprises reacting a compound of formula (18):

contacting diphenyl phosphorazidate in an organic solvent in the presence of a base, followed by the addition of t-butanol and CuCl.

In some embodiments, the organic solvent is toluene. In other embodiments, the base is triethylamine.

In some embodiments, the method further comprises preparing a compound of formula (18):

The method comprises reacting a compound of formula (17):

in a solvent with an aqueous base.

In one embodiment, the base is LiOH or NaOH. In another embodiment, the solvent is MeOH.

In some embodiments, the method further comprises preparing a compound of formula (17):

the method comprises reacting a compound of formula (11):

with a compound of formula (12):

in a solvent in the presence of a catalyst of formula e (15 and 16):

in one embodiment, the amount of catalyst (16) used for the reaction is lower than the amount of catalyst (15). In another embodiment, the loading of the catalyst (15) is 5 to 20 mol%. In some embodiments, the solvent is toluene. In other embodiments, the contacting is performed at a temperature of about-20 ℃ to about 0 ℃. In yet another embodiment, the contacting is conducted at a temperature of about-15 ℃.

In some embodiments, the method further comprises preparing a compound of formula (15):

the method comprises reacting a compound of formula (13):

with a compound of formula (14):

in a solvent.

In one embodiment, the solvent is toluene. In another embodiment, the contacting is conducted at reflux temperature.

In one aspect, provided herein is a process for preparing a compound of formula (a):

The method comprises reacting a compound of formula (9 a):

contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is 2-propanol, methanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 a):

the process comprises separating a diastereomeric mixture of compounds of formula (9a and 9b) by chiral separation:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is recrystallization in a solvent. In one embodiment, the recrystallization solvent is MTBE.

In some embodiments, the method further comprises preparing a diastereomeric mixture of compounds of formulae (9a and 9 b):

the method comprises reacting a compound of formula (8):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent in the presence of a base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane. In one embodimentThe boron hydride agent is B2H6、9-BBN、 BCl3/Me3SiH or (+) -diisopinocampheylborane. In another embodiment, the oxidizing agent is H 2O2Or potassium hydrogen persulfate. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF. In yet another embodiment, the base is NaOH.

In some embodiments, the method further comprises preparing a compound of formula (8):

the method comprises the utilization of CDI, NH2OH andtBuOH carries out the Curtius rearrangement of the compound of formula (18):

in some embodiments, the method further comprises preparing a compound of formula (18):

the process comprises resolving a compound of formula (20) with a chiral amine:

in one embodiment, the chiral amine is (S) -phenylethylamine or (R) -phenylethylamine.

In some embodiments, the method further comprises preparing a compound of formula (20):

the method comprises reacting a compound of formula (19):

with a compound of formula (12):

contact, followed by treatment with a base, followed by acid post-treatment. In one embodiment, the base is NaOH. In another embodiment, the acid post-treatment uses H2SO4The process is carried out. In some embodiments, the contacting is performed at a temperature of about 25 ℃.

In one aspect, provided herein is a process for preparing a compound of formula (a):

the method comprises reacting a compound of formula (9 a):

Contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is 2-propanol, methanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 a):

the process comprises separating a diastereomeric mixture of compounds of formula (9a and 9b) by employing a chiral separation process:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is recrystallization in a solvent. In one embodiment, the recrystallization solvent is MTBE.

In some embodiments, the method further comprises preparing a diastereomeric mixture of compounds of formulae (9a and 9 b):

the method comprises reacting a compound of formula (8):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent in the presence of a base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane. In one embodiment, the borohydride is B2H6、9-BBN、 BCl3/Me3SiH or (+) -diisopinocampheylborane. In another embodiment, the oxidizing agent is H2O2Or potassium hydrogen persulfate. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF. In yet another embodiment, the base is NaOH.

In some embodiments, the method further comprises preparing a compound of formula (8):

the method comprises reacting a compound of formula (18):

contacting diphenyl phosphorazidate in an organic solvent in the presence of a base, followed by the addition of t-butanol and CuCl.

In some embodiments, the organic solvent is toluene. In other embodiments, the base is triethylamine.

In some embodiments, the method further comprises preparing a compound of formula (18):

the process comprises hydrolysing a compound of formula (22) by treatment with a base and an oxidising agent:

in one embodiment, R isiPr or CH2Ph. In one embodiment, the base is LiOH. In another embodiment, the oxidizing agent is H2O2

In some embodiments, the method further comprises preparing a compound of formula (22):

the method comprises reacting a compound of formula (21):

with a compound of formula (12):

under conditions suitable for the D-A reaction.

In one aspect, provided herein is a process for preparing a compound of formula (a):

the process comprises separating the enantiomers of formula (26a and 26b) by employing a chiral separation method:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC) or chiral resolution.

In some embodiments, the method further comprises preparing a mixture of enantiomers of formulae (26a and 26 b):

the process comprises reacting a mixture of enantiomers of formulae (25a and 25 b):

is contacted with a reducing agent.

In one embodiment, the reducing agent is hydrogen in the presence of Pd/C. In another embodiment, the reducing agent is Zn in EtOH in the presence of acetic acid.

In some embodiments, the method further comprises preparing a mixture of enantiomers of formulae (25a and 25 b):

the process comprises racemizing a mixture of four diastereomers of formulae (25a, 25b, 25c, and 25d) by treatment with a base:

in some embodiments, the base is selected from NaOH, NaOEt, or tBuOK.

In some embodiments, the method further comprises preparing a mixture of four diastereomers of formulae (25a, 25b, 25c, and 25 d):

the method comprises reacting a compound of formula (24):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent in the presence of a base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane. In one embodiment, the hydroboration agent is BH3. In another embodiment, the oxidizing agent is H 2O2Or potassium hydrogen persulfate. In another embodiment, the oxidizing agent is H2O2. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF. In yet another embodiment, the base is NaOH.

In some embodiments, the method further comprises preparing a compound of formula (24):

the method comprises reacting a compound of formula (23):

with a compound of formula (12):

under conditions suitable for the D-A reaction.

In one aspect, provided herein is a process for preparing a compound of formula (a):

the method comprises reacting a compound of formula (9 a):

contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is 2-propanol, methanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 a):

the process comprises separating a diastereomeric mixture of compounds of formulae (9a, 9b, 9c and 9d) by chiral separation:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC).

In some embodiments, the method further comprises preparing a diastereomeric mixture of compounds of formulae (9a, 9b, 9c, and 9 d):

the method comprises reacting a compound of formula (32):

with a hydroborating agent followed by treatment with an oxidizing agent in a solvent in the presence of a base.

In one embodiment, the hydroboration agent is BH3/THF、B2H6、9-BBN、BCl3/Me3SiH or (+) -diisopinocampheylborane. In one embodiment, the hydroboration agent is BH3. In another embodiment, the solvent is THF or EtOH. In another embodiment, the solvent is THF. In another embodiment, the oxidizing agent is H2O2Or potassium hydrogen persulfate. In some embodiments, the oxidizing agent is H2O2. In another embodiment, the base is NaOH. In yet another embodiment, the solvent is EtOH.

In some embodiments, the method further comprises preparing a compound of formula (32):

the method comprises reacting a compound of formula (31):

optionally with Boc in the presence of a base in a solvent2And (4) contacting with O.

In one embodiment, the solvent is DCM. In another embodiment, the base is triethylamine.

In some embodiments, the method further comprises preparing a compound of formula (31):

the method comprises reacting a compound of formula (30):

Is contacted with hydrazine.

In some embodiments, the method further comprises preparing a compound of formula (30):

the method comprises reacting a compound of formula (29):

contacting with a dehydrating agent.

In one embodiment, the dehydrating agent is KHSO4Or H2SO4

In some embodiments, the method further comprises preparing a compound of formula (29):

the method comprises reacting a compound of formula (27):

with a compound of formula (28):

in the presence of a base.

In one embodiment, the base is K2CO3

In one aspect, provided herein is a process for preparing a compound of formula (a):

the method comprises reacting a compound of formula (36):

contacting with diphenylphosphoryl azide in the presence of water.

In some embodiments, the method further comprises preparing a compound of formula (36):

the method comprises reacting a compound of formula (35):

the mixture is contacted with alkali to form a mixture,

wherein R ═ Me or iPr.

In some embodiments, the base is NaOH.

In some embodiments, the method further comprises preparing a compound of formula (35):

the method comprises reacting a compound of formula (34):

and Ti (OiPr)4Mg and TMS-Cl are contacted,

wherein R ═ Me or iPr.

In some embodiments, the method further comprises preparing a compound of formula (34):

The method comprises reacting a compound of formula (33):

is contacted with an alkoxide.

In one embodiment, the alkoxide is NaOMe or NaOiPr.

In some embodiments, the method further comprises preparing a compound of formula (33):

the method comprises reacting a compound of formula (18):

in NaHCO3In the presence of KI3And (4) contacting.

In one aspect, provided herein is a process for preparing a compound of formula (a):

the method comprises reacting a compound of formula (9 a):

contact with hydrochloric acid in a solvent.

In some embodiments, the solvent is 2-propanol, methanol, an ether, or dioxane.

In some embodiments, the method further comprises preparing a compound of formula (9 a):

the process comprises separating a diastereomeric mixture of a compound of formula (40) by employing a chiral separation process:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC).

In some embodiments, the method further comprises preparing a compound of formula (40):

the method comprises reacting a compound of formula (39):

Optionally with Boc in the presence of a base in a solvent2And (4) contacting with O.

In one embodiment, the solvent is DCM. In another embodiment, the base is triethylamine.

In some embodiments, the method further comprises preparing a compound of formula (39):

the method comprises reacting a compound of formula (38):

is contacted with a reducing agent.

In one embodiment, the reducing agent is NaBH4

In some embodiments, the method further comprises preparing a compound of formula (38):

the method comprises reacting a compound of formula (37):

with hydrogen in the presence of a catalyst.

In some embodiments, the catalyst is Pd/C or Pd (OH)2/C。

In one aspect, provided herein is a process for preparing a compound of formula (a):

the process comprises purifying a compound of formula (39) by employing a chiral separation process:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC).

In some embodiments, the method further comprises preparing a compound of formula (39):

the method comprises reacting a compound of formula (38):

Is contacted with a reducing agent.

In one embodiment, the reducing agent is NaBH4

In some embodiments, the method further comprises preparing a compound of formula (38):

the method comprises reacting a compound of formula (37):

with hydrogen in the presence of a catalyst.

In some embodiments, the catalyst is Pd/C or Pd (OH)2/C。

In one aspect, provided herein is a process for preparing a compound of formula (a):

the method comprises reacting a compound of formula (45 a):

with hydrogen in the presence of a catalyst.

In some embodiments, the catalyst is Pd/C.

In some embodiments, the method further comprises preparing a compound of formula (45 a):

the process comprises separating the diastereomers of the compound of formula (45) by employing a chiral separation method:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC).

In some embodiments, the method further comprises preparing a compound of formula (45):

the method comprises reacting a compound of formula (44 a):

is contacted with a reducing agent.

In one embodiment, the reducing agent is NaBH4

In some embodiments, the method further comprises preparing a compound of formula (44 a):

the process comprises isolating a diastereomeric mixture of a compound of formula (44):

which is isolated by resolution using a compound of the formula

In some embodiments, the method further comprises preparing a compound of formula (44):

the method comprises reacting a compound of formula (43):

with a chiral amine.

In one embodiment, the chiral amine is (S) -phenylethylamine or (R) -phenylethylamine.

In some embodiments, the method further comprises preparing a compound of formula (43):

the method comprises reacting a compound of formula (42):

with a compound of formula (41):

in the presence of a base.

In one embodiment, the base is KOtBu.

In one aspect, provided herein is a process for preparing a compound of formula (a):

the process comprises the de-tosylation of a compound of formula (52) by treatment with a base and thiophenol:

in one embodiment, the base is K2CO3Or DBU.

In some embodiments, the method further comprises preparing a compound of formula (52):

the method comprises using Pd (CF) in a solvent under hydrogen atmosphere 3CO2)2And S-SegPhos treatment of the asymmetrically reduced compound of formula (51):

in one embodiment, the solvent is TFE.

In some embodiments, the method further comprises preparing a compound of formula (51):

the method comprises reacting a compound of formula (50):

in the presence of a base, in a solvent.

In some embodiments, the solvent is CCl4. In other embodiments, the base is triethylamine. In some embodiments, the process is carried out at a temperature of about-23 ℃.

In some embodiments, the method further comprises preparing a compound of formula (50):

the method comprises reacting a compound of formula (49):

in a solvent withNH4Cl and Amberlyst A21.

In some embodiments, the solvent is ethanol.

In some embodiments, the method further comprises preparing a compound of formula (49):

the method comprises reacting a compound of formula (48 a):

with FeCl in a solvent3And (4) contacting.

In some embodiments, the solvent is DCM.

In some embodiments, the method further comprises purifying the compound of formula (48 a):

the method comprises separating a mixture of compounds of formula (48a and 48b) by employing a chiral separation method:

in one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC), recrystallization, chiral HPLC, chiral LC, or chiral resolution. In one embodiment, the chiral separation method is chiral Supercritical Fluid Chromatography (SFC) or chiral resolution.

In some embodiments, the method further comprises preparing a mixture of compounds of formula (48a and 48 b):

the method comprises reacting a compound of formula (47):

in a solvent in AlMe3With MeLi in the presence of (b).

In one embodiment, the solvent is heptane.

In some embodiments, the method further comprises preparing a compound of formula (47):

the method comprises reacting a compound of formula (46):

with a catalytic amount of p-TsOH in a solvent, followed by treatment with an oxidizing agent.

In one embodiment, the oxidizing agent is m-CPBA. In some embodiments, the solvent is DCM.

Solid forms of Compound 1

In certain embodiments, provided herein are solid forms of compound 1. In certain embodiments, the solid form is crystalline. In certain embodiments, the solid form is a one-component solid form. In certain embodiments, the solid form is a solvate.

While not wishing to be bound by any particular theory, certain solid forms are characterized by physical properties suitable for pharmaceutical and therapeutic dosage forms, such as stability, solubility, and dissolution. Furthermore, while not wishing to be bound by any particular theory, certain solid forms are characterized by physical properties (e.g., density, compressibility, hardness, morphology, lysis, viscosity, solubility, water absorption, electrical properties, thermal behavior, solid state reactivity, physical stability, and chemical stability) that affect the particular process (e.g., yield, filtration, washing, drying, milling, mixing, tableting, flowability, dissolution, formulation, and lyophilization) by which certain solid forms suitable for manufacturing solid dosage forms are prepared. Such properties may be determined using specific analytical chemistry techniques, including solid state analysis techniques as known in the art (e.g., X-ray diffraction, microscopy, spectroscopy, and thermal analysis).

The solid forms provided herein (e.g., form a, form B, form C, form D, form E, form F, form G, form H, form I, and amorphous solids of compound 1) can be characterized using a number of methods known to those skilled in the art including, but not limited to, single crystal X-ray diffraction, X-ray powder diffraction (XRPD), microscopy (e.g., Scanning Electron Microscope (SEM)), thermal analysis (e.g., Differential Scanning Calorimetry (DSC), dynamic gas adsorption (DVS), thermogravimetric analysis (TGA), and hot stage microscopy), spectroscopy (e.g., infrared, raman, and solid state nuclear magnetic resonance), Ultra High Performance Liquid Chromatography (UHPLC), and proton nuclear magnetic resonance (UHPLC) ((r))1H NMR) spectrum. The particle size and particle size distribution of the solid forms provided herein can be determined by conventional methods, such as laser light scattering techniques.

The purity of the solid forms provided herein can be determined by standard analytical methods, such as Thin Layer Chromatography (TLC), gel electrophoresis, gas chromatography, Ultra High Performance Liquid Chromatography (UHPLC), and Mass Spectrometry (MS).

It will be appreciated that the values of the peaks of the X-ray powder diffraction pattern may vary slightly from one machine to another or from one sample to another, and therefore the values quoted should not be interpreted as absolute values, but rather with acceptable variability, such as ± 0.2 ° 2 θ (see us pharmacopoeia, page 2228 (2003)).

In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) obtaining a slurry of form a in a solvent; 2) stirring the slurry at a specific temperature (e.g., about 25 ℃ or about 50 ℃) for a period of time (e.g., about 24 hours); and 3) collecting the solid from the slurry by filtration and optionally drying. In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) obtaining a slurry of form a in a solvent; 2) stirring the slurry at about 25 ℃ or about 50 ℃ for about 24 hours; and 3) collect the solids from the slurry through a 0.45 μm PTFE syringe filter and optionally air dry. In certain embodiments, the method for preparing a solid form of compound 1 is an equilibrium experiment, such as a slurry experiment.

In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) dissolving form a in a solvent to produce a solution; 2) filtering the solution if form a is not completely dissolved; and 3) evaporating the solution at a specific pressure (e.g., about 1atm) and a specific temperature (e.g., about 25 ℃ or about 50 ℃) to produce a solid. In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) dissolving form a in a solvent to produce a solution; 2) if form a is not completely dissolved, the solution is filtered through a 0.45 μm PTFE syringe filter; and 3) evaporating the solution under nitrogen at about 25 ℃ or about 50 ℃ under about 1atm atmospheric pressure to produce a solid. In certain embodiments, the method for preparing the solid form of compound 1 is an evaporation experiment.

In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) obtaining a saturated solution of form a in a solvent at a first temperature (e.g., about 60 ℃); 2) Stirring the solution at a first temperature for a period of time (e.g., 10 minutes); 3) filtering the solution; 4) slowly cooling the solution to a second temperature (e.g., about-5 ℃ to about 15 ℃); and 5) separating the solid from the solution and optionally drying. In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) obtaining a saturated solution of form a in a solvent at about 60 ℃; 2) the solution was stirred at about 60 ℃ for 10 minutes; 3) filtering the solution through a 0.45 μm PTFE syringe filter; 4) slowly cooling the solution to about 5 ℃; and 5) separating the solid from the solution and optionally air drying. In certain embodiments, the method for preparing the solid form of compound 1 is a cooling recrystallization experiment.

In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) obtaining a saturated solution of form a in a solvent at a first temperature (e.g., about 60 ℃); 2) Adding an anti-solvent to the saturated solution at a first temperature; 3) cooling to a second temperature (e.g., about-5 ℃ to about 15 ℃); and 4) collecting the solid if there is precipitation and evaporating the solvent to collect the solid if there is no precipitation; and 5) optionally drying. In certain embodiments, provided herein are methods for preparing a solid form of compound 1, comprising 1) obtaining a saturated solution of form a in a solvent at about 60 ℃; 2) adding an anti-solvent at about 60 ℃ to the saturated solution; 3) cooling to about 5 ℃; and 4) collecting the solid if there is precipitation and evaporating the solvent to collect the solid if there is no precipitation; and 5) optionally air drying. In certain embodiments, the volume ratio of solvent to anti-solvent is about 1: 9. In certain embodiments, the method for preparing a solid form of compound 1 is an anti-solvent recrystallization experiment.

In certain embodiments, the solvent is acetone, DCM, EtOAc, EtOH/H2O (about 1: 1), H2O, heptane, IPA, ACN/H2O (about 1: 1), MEK, MeOH, MTBE, n-BuOH, THF/H2O (about 1: 1), toluene, or sulfolane.

In certain embodiments, the anti-solvent is ACN, heptane, MTBE, or water.

Form A

In certain embodiments, provided herein is form a.

In one embodiment, form a is a solid form of compound 1. In one embodiment, form a is a non-stoichiometric channel hydrate solid form of compound 1. In another embodiment, form a is crystalline.

In certain embodiments, form a provided herein is obtained by an equilibration experiment, an evaporation experiment, and an anti-solvent recrystallization experiment (see table 1, table 2, and table 3). In certain embodiments, form a is obtained from a solvent system comprising MTBE, heptane, water, EtOH/H2O (about 1: 1), MeOH and asWater as anti-solvent, EtOH with MTBE and IPA as anti-solvent with heptane as anti-solvent.

In one embodiment, the method of preparing form a comprises the steps of: 1) mixing form H with a solvent (e.g., DMSO) mixture comprising water (e.g., at least about 70% water by volume); 2) stirring at a temperature (e.g., about 20 ℃ to about 25 ℃, such as about 22 ℃) for a period of time (e.g., 1 hour to about 6 hours, such as about 3 hours); and 3) collecting the solid and optionally drying.

In one embodiment, the method of preparing form a comprises the steps of: 1) mixing form H with a solvent (e.g., DMSO) mixture comprising water (e.g., at least about 50% water by volume); 2) heating to a temperature (e.g., about 60 ℃ to about 100 ℃, such as about 60 ℃ or about 70 ℃) for a period of time (e.g., about 1 hour to about 6 hours, such as about 3 hours); 3) cooling to a second temperature (e.g., about 10 ℃ to about 40 ℃, such as about 25 ℃); and 4) collecting the solid and optionally drying.

In one embodiment, the method of preparing form a comprises the steps of: 1) mixing form H with a solvent (e.g., DMSO) mixture comprising water (e.g., at least about 70% water by volume); 2) heating the resulting mixture to a first temperature (e.g., about 60 ℃ to about 100 ℃, such as about 60 ℃ or about 70 ℃) for a period of time (e.g., about 1 hour to about 6 hours, such as 3 hours); 3) Cooling the mixture to a second temperature (e.g., about 10 ℃ to about 40 ℃, such as about 25 ℃); and 4) collecting the solid and optionally drying.

In another embodiment, the method of making form a comprises the steps of: 1) mixing form H with a solvent (e.g., DMSO) mixture comprising at least about 70% water by volume; 2) heating the resulting mixture to a temperature (e.g., about 60 ℃ to about 100 ℃, such as about 60 ℃ or about 70 ℃) for about 1 hour to about 6 hours, such as about 3 hours; 3) cooling the mixture to a temperature (e.g., about 10 ℃ to about 40 ℃, such as about 25 ℃); and 4) collecting the solid and optionally drying.

In certain embodiments, a solid form provided herein, e.g., form a, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form a has an X-ray powder diffraction pattern substantially as shown in figure 1. In one embodiment, form a has one or more characteristic X-ray powder diffraction peaks at about 9.74, 10.55, 11.86, 12.98, 13.61, 15.90, 16.41, 17.20, 17.85, 18.04, 18.54, 19.29, 19.56, 19.84, 20.19, 21.37, 21.83, 22.90, 23.46, 23.84, 24.36, 24.88, 25.29, 26.14, 26.92, 27.83, 28.30, 28.69, 29.21, 30.50, 31.63, 32.11, 32.63, 33.17, 34.32, 34.74, 36.00, 36.56, 36.95, 37.26, 37.61, 38.40, 39.07, 39.34, or 39.64 ° 2 θ as depicted in fig. 1. In a specific embodiment, form a has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 10.55, 13.61, 17.20, 17.85, 18.04, 19.84, 22.90, or 24.36 ° 2 Θ. In another embodiment, form a has one, two, three, or four characteristic X-ray powder diffraction peaks at about 10.55, 13.61, 17.20, or 19.84 ° 2 Θ. In another embodiment, form a has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty-one, thirty-two, thirty-three, thirty-four, thirty-seven, thirty-six, thirty-eight, thirty-nine, forty-one, forty-two, forty-three, forty-four, or forty-five characteristic X-ray powder diffraction peaks as shown in table 8.

Table 7 shows a summary of crystallographic data from single crystal structure measurements. In one embodiment, form a has a crystal packing diagram substantially as shown in fig. 2. In one embodiment, form a is a solid form that crystallizes in space group P2(1)2 (1). In one embodiment, form a is a non-stoichiometric channel hydrate.

In one embodiment, form a has an SEM image substantially as shown in fig. 3.

In one embodiment, provided herein is form a having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 4. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 20 ℃ to about 300 ℃ includes a total mass loss of about 0.45% of the total mass of the sample at about 30 ℃ to about 150 ℃. Thus, in certain embodiments, the crystalline form loses about 0.1% to about 5%, e.g., about 0.45% or about 3.3% of its total mass when heated from ambient temperature to about 300 ℃.

In one embodiment, provided herein is form a having a DSC thermogram substantially as depicted in figure 5 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 223 ℃.

In one embodiment, provided herein is a form a having a DVS isotherm plot substantially as depicted in fig. 6.

In one embodiment, provided herein is a method having a structure substantially as depicted in fig. 71Form a of H NMR spectrum.

In yet another embodiment, form a is substantially pure. In certain embodiments, substantially pure form a is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form a is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form B

In certain embodiments, form B is provided herein.

In one embodiment, form B is a solid form of compound 1. In another embodiment, form B is crystalline. In one embodiment, form B is a solvated form of compound 1. In one embodiment, form B is an acetone solvated form of compound 1. In one embodiment, form B is an acetone semi-solvated form of compound 1.

In certain embodiments, form B provided herein is obtained by an equilibration experiment, an evaporation experiment, and an anti-solvent recrystallization experiment (see table 1, table 2, and table 3). In certain embodiments, form B is obtained from certain solvent systems comprising acetone, MEK, DCM, THF/H 2O (about 1: 1) and IPA with heptane as the anti-solvent.

In certain embodiments, the solid form provided herein, e.g., form B, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form B has an X-ray powder diffraction pattern substantially as shown in figure 10. In one embodiment, form B has one or more characteristic X-ray powder diffraction peaks at about 9.80, 10.30, 12.23, 14.62, 16.70, 17.29, 18.23, 18.59, 19.61, 20.19, 20.66, 20.94, 21.74, 23.03, 23.84, 24.32, 24.58, 25.88, 26.27, 26.86, 27.52, 28.35, 28.62, 29.63, 30.55, 30.87, 31.44, 32.12, 33.71, 33.95, 34.96, 35.94, 36.14, 36.56, 37.22, or 38.76 ° 2 θ as depicted in fig. 10. In a specific embodiment, form B has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 9.80, 10.30, 14.62, 17.29, 18.23, 20.66, 21.74, or 30.55 ° 2 Θ. In another embodiment, form B has one, two, three, or four characteristic X-ray powder diffraction peaks at about 9.80, 17.29, 18.23, or 21.74 ° 2 Θ. In another embodiment, form B has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, or thirty-six characteristic X-ray powder diffraction peaks as shown in table 9.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 11. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 8.5% of the total mass of the sample at about 75 ℃ to about 175 ℃. Thus, in certain embodiments, the crystalline form loses about 8.5% of its total mass when heated from ambient temperature to about 300 ℃. In certain embodiments, the crystalline form comprises 0.5 molar equivalents of solvent in the crystal lattice, corresponding to about 0.5 moles of acetone per mole of compound 1. The theoretical acetone content of the acetone hemisolvate of compound 1 was 8.3 wt% matching the observed TGA weight loss. In certain embodiments, the crystalline form is an acetone hemisolvate of compound 1.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 12 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 147 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 12 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 223 ℃.

In one embodiment, provided herein is form B having a 1H NMR spectrum substantially as depicted in fig. 13.

In yet another embodiment, form B is substantially pure. In certain embodiments, substantially pure form B is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form B is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form C

In certain embodiments, form C is provided herein.

In one embodiment, form C is a solid form of compound 1. In another embodiment, form C is crystalline. In one embodiment, form C is a solvated form of compound 1. In one embodiment, form C is an ethanol solvated form of compound 1. In one embodiment, form C is the ethanol semi-solvated form of compound 1.

In certain embodiments, form C provided herein is obtained by an equilibration experiment, an evaporation experiment, a cooling recrystallization experiment, and an anti-solvent recrystallization experiment (see table 1, table 2, and table 3). In certain embodiments, form C is obtained from certain solvent systems including ACN, ACN/H 2O (about 1: 1), EtOH/H2O (about 1: 1), IPA, MEK, EtOH with MTBE as anti-solvent, EtOH with heptane as anti-solvent, EtOH with ACN and IPA as anti-solvent with heptane as anti-solvent.

In certain embodiments, the solid form provided herein, e.g., form C, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form C has an X-ray powder diffraction pattern substantially as shown in figure 14. In one embodiment, form C has one or more characteristic X-ray powder diffraction peaks at about 9.83, 10.21, 12.16, 14.66, 15.52, 16.50, 17.26, 17.61, 17.91, 18.18, 18.65, 19.67, 19.99, 20.46, 21.86, 23.32, 23.78, 24.44, 25.65, 25.81, 26.28, 26.72, 27.46, 28.04, 28.30, 28.60, 29.56, 30.47, 30.70, 31.29, 31.77, 32.16, 32.94, 33.55, 34.00, 34.85, 35.14, 35.57, 35.90, 36.62, 37.76, or 38.93 ° 2 θ as depicted in fig. 14. In a specific embodiment, form C has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 9.83, 10.21, 12.16, 17.26, 17.61, 18.18, 20.46, or 21.86 ° 2 Θ. In another embodiment, form C has one, two, three, or four characteristic X-ray powder diffraction peaks at about 9.83, 10.21, 17.26, or 21.86 ° 2 Θ. In another embodiment, form C has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty-ten, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty-one, or forty-two characteristic X-ray powder diffraction peaks as shown in table 10.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 15. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 7.3% of the total mass of the sample at about 75 ℃ to about 175 ℃. Thus, in certain embodiments, the crystalline form loses about 7.3% of its total mass when heated from ambient temperature to about 300 ℃. In certain embodiments, the crystalline form comprises 0.5 molar equivalents of solvent in the crystal lattice, corresponding to about 0.5 moles of ethanol per mole of compound 1. The theoretical ethanol content of the ethanol hemisolvate of compound 1 was 6.7 wt% matching the observed TGA weight loss. In certain embodiments, the crystalline form is an ethanol hemisolvate of compound 1.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 16 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 143 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 16 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 224 ℃.

In one embodiment, provided herein is a method having a structure substantially as depicted in fig. 171Form C of H NMR spectrum.

In yet another embodiment, form C is substantially pure. In certain embodiments, substantially pure form C is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form C has a purity of no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 98.5%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%.

Form D

In certain embodiments, form D is provided herein.

In one embodiment, form D is a solid form of compound 1. In another embodiment, form D is crystalline. In one embodiment, form D is a solvated form of compound 1. In one embodiment, form D is the methanol solvated form of compound 1. In one embodiment, form D is a methanol semi-solvated form of compound 1.

In certain embodiments, form D provided herein is obtained by an equilibration experiment, an evaporation experiment, a cooling recrystallization experiment, and an anti-solvent recrystallization experiment (see table 1, table 2, and table 3). In certain embodiments, form D is obtained from a solvent system comprising MeOH and MeOH with MTBE as the anti-solvent.

In certain embodiments, a solid form provided herein, e.g., form D, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form D has an X-ray powder diffraction pattern substantially as shown in figure 18. In one embodiment, form D has one or more characteristic X-ray powder diffraction peaks at about 10.37, 12.85, 13.41, 15.68, 16.25, 17.02, 17.54, 17.73, 18.34, 19.52, 19.93, 20.78, 21.09, 21.54, 22.47, 23.11, 23.55, 23.92, 24.51, 24.99, 25.81, 26.47, 26.88, 27.33, 27.83, 28.19, 28.64, 30.08, 30.82, 31.20, 31.60, 32.02, 32.50, 33.58, 34.25, 35.39, 35.87, 36.55, 36.81, 37.06, 37.77, or 38.60 ° 2 θ as depicted in fig. 18. In a specific embodiment, form D has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 10.37, 13.41, 17.54, 17.73, 19.52, 21.54, 22.47, or 23.92 ° 2 Θ. In another embodiment, form D has one, two, three, or four characteristic X-ray powder diffraction peaks at about 10.37, 13.41, 19.52, or 22.47 ° 2 Θ. In another embodiment, form D has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty-ten, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, forty-one, forty-two characteristic X-ray powder diffraction peaks as shown in table 11.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 19. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 4% of the total mass of the sample at about 100 ℃ to about 160 ℃. Thus, in certain embodiments, the crystalline form loses about 4% of its total mass when heated from ambient temperature to about 300 ℃. In certain embodiments, the crystalline form comprises 0.5 molar equivalents of solvent in the crystal lattice, corresponding to about 0.5 moles of methanol per mole of compound 1. The theoretical methanol content of the methanol hemisolvate of compound 1 was 4.7 wt% matching the observed TGA weight loss. In certain embodiments, the crystalline form is a methanol hemisolvate of compound 1.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 20 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 170 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 20 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 223 ℃.

In one embodiment, provided herein is a method having a structure substantially as depicted in fig. 211Form D of H NMR spectrum.

In yet another embodiment, form D is substantially pure. In certain embodiments, substantially pure form D is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form D is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form E

In certain embodiments, form E is provided herein.

In one embodiment, form E is a solid form of compound 1. In another embodiment, form E is crystalline. In one embodiment, form E is a solvated form of compound 1. In one embodiment, form E is the n-butanol solvated form of compound 1. In one embodiment, form E is the n-butanol semi-solvated form of compound 1.

In certain embodiments, form E provided herein is obtained by an equilibration experiment and an evaporation experiment (see tables 1 and 2). In certain embodiments, form E is obtained from a solvent system comprising n-butanol.

In certain embodiments, a solid form provided herein, e.g., form E, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form E has an X-ray powder diffraction pattern substantially as shown in figure 22. In one embodiment, form E has one or more characteristic X-ray powder diffraction peaks at about 8.70, 9.92, 10.36, 11.97, 14.50, 15.51, 16.39, 17.29, 18.37, 19.55, 20.10, 21.81, 23.21, 23.45, 24.17, 24.61, 25.44, 25.83, 26.23, 26.45, 26.61, 27.64, 28.48, 29.19, 29.97, 30.39, 30.81, 31.36, 31.66, 32.62, 33.67, 34.75, 35.24, 35.96, 36.48, 37.20, 37.62, 38.93, or 39.20 ° 2 θ as depicted in fig. 22. In a specific embodiment, form E has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 9.92, 10.36, 11.97, 14.50, 17.29, 18.37, 20.10, or 21.81 ° 2 Θ. In another embodiment, form E has one, two, three, or four characteristic X-ray powder diffraction peaks at about 9.92, 17.29, 18.37, or 21.81 ° 2 Θ. In another embodiment, form E has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-eight, twenty-nine, thirty-ten, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, thirty-eight, or thirty-nine characteristic X-ray powder diffraction peaks as shown in table 12.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 23. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 10.3% of the total mass of the sample at about 75 ℃ to about 175 ℃. Thus, in certain embodiments, the crystalline form loses about 10.3% of its total mass when heated from ambient temperature to about 300 ℃. In certain embodiments, the crystalline form comprises 0.5 molar equivalents of solvent in the crystal lattice, corresponding to about 0.5 moles of n-butanol per mole of compound 1. The theoretical n-butanol content of the n-butanol hemisolvate of compound 1 was 10.3 wt% matching the observed TGA weight loss. In certain embodiments, the crystalline form is an n-butanol hemisolvate of compound 1.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 24 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 124 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 24 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 224 ℃.

In one embodiment, provided herein is a method having a structure substantially as depicted in fig. 251Form E of H NMR spectrum.

In yet another embodiment, form E is substantially pure. In certain embodiments, substantially pure form E is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form E is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form F

In certain embodiments, provided herein is form F.

In one embodiment, form F is a solid form of compound 1. In another embodiment, form F is crystalline. In one embodiment, form F is a solvated form of compound 1. In one embodiment, form F is a toluene solvated form of compound 1. In one embodiment, form F is a 0.3 molar toluene solvated form of compound 1.

In certain embodiments, form F provided herein is obtained by an equilibration experiment (see table 1). In certain embodiments, form F is obtained from a solvent system comprising toluene.

In certain embodiments, a solid form provided herein, e.g., form F, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form F has an X-ray powder diffraction pattern substantially as shown in figure 26. In one embodiment, form F has one or more characteristic X-ray powder diffraction peaks, as shown in figure 26, at about 8.07, 9.21, 10.58, 10.88, 12.06, 14.56, 14.87, 16.28, 17.45, 17.79, 18.53, 19.65, 20.03, 20.85, 21.10, 23.72, 24.41, 25.11, 25.98, 26.61, 27.94, 29.25, 30.40, 32.00, 34.06, 35.72, 36.58, or 37.59 ° 2 θ. In a specific embodiment, form F has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 8.07, 9.21, 12.06, 17.45, 17.79, 18.53, 20.85, or 21.10 ° 2 Θ. In another embodiment, form F has one, two, three, or four characteristic X-ray powder diffraction peaks at about 17.45, 18.53, 20.85, or 21.10 ° 2 Θ. In another embodiment, form F has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, or twenty-eight characteristic X-ray powder diffraction peaks as shown in table 13.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 27. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 6.9% of the total mass of the sample at about 75 ℃ to about 175 ℃. Thus, in certain embodiments, the crystalline form loses about 6.9% of its total mass when heated from ambient temperature to about 300 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 28 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 113 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 28 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 223 ℃.

In one embodiment, provided herein is a method having a structure substantially as depicted in fig. 291Form F of H NMR spectrum. In one embodiment, of form F 1The H NMR spectrum showed that form F contained about 0.3 molar equivalents of toluene. In certain embodiments, form F is 0.3 molar equivalents of compound 1 as a toluene solvate.

In yet another embodiment, form F is substantially pure. In certain embodiments, substantially pure form F is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form F is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form G

In certain embodiments, form G is provided herein.

In one embodiment, form G is a solid form of compound 1. In another embodiment, form G is crystalline. In one embodiment, form G is a solvated form of compound 1. In one embodiment, form G is the EtOAc solvated form of compound 1. In one embodiment, form G is the EtOAc hemisolvated form of compound 1.

In certain embodiments, form G provided herein is obtained by an equilibration experiment, an evaporation experiment, and an anti-solvent recrystallization experiment (see table 1, table 2, and table 3). In certain embodiments, form G is obtained from a solvent system comprising EtOAc.

In certain embodiments, a solid form provided herein, e.g., form G, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form G has an X-ray powder diffraction pattern substantially as shown in figure 30. In one embodiment, form G has one or more characteristic X-ray powder diffraction peaks at about 8.63, 9.51, 10.34, 12.14, 14.43, 16.44, 16.94, 17.33, 17.90, 18.58, 19.10, 20.09, 20.41, 20.80, 21.28, 22.66, 23.62, 24.33, 25.55, 25.65, 26.42, 26.89, 27.00, 27.78, 28.83, 29.86, 31.22, 31.77, 32.67, 33.90, 34.28, 35.04, 35.44, 36.24, 36.57, 37.59, 38.00, or 38.76 ° 2 θ as depicted in fig. 30. In a specific embodiment, form G has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 9.51, 10.34, 16.94, 17.33, 17.90, 21.28, 28.83, or 31.22 ° 2 Θ. In another embodiment, form G has one, two, three, or four characteristic X-ray powder diffraction peaks at about 9.51, 10.34, 17.90, or 21.28 ° 2 Θ. In another embodiment, form G has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-eight, twenty-nine, thirty-ten, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, or thirty-eight characteristic X-ray powder diffraction peaks as shown in table 14.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 31. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 11.9% of the total mass of the sample at about 75 ℃ to about 175 ℃. Thus, in certain embodiments, the crystalline form loses about 11.9% of its total mass when heated from about ambient temperature to about 300 ℃. In certain embodiments, the crystalline form comprises 0.5 molar equivalents of solvent in the crystal lattice, corresponding to about 0.5 moles of EtOAc per mole of compound 1. The theoretical EtOAc content of the EtOAc hemisolvate of compound 1 was 12.1 wt% matching the observed TGA weight loss. In certain embodiments, the crystalline form is the EtOAc hemisolvate of compound 1.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 32 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 116 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 32 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 223 ℃.

In one embodiment, provided herein is a method having a structure substantially as depicted in fig. 331Form G of H NMR spectrum. In one embodiment, of form G1The H NMR spectrum showed form G to contain about 0.5 molar equivalents of EtOAc. In certain embodiments, form G is the EtOAc hemisolvate of compound 1.

In yet another embodiment, form G is substantially pure. In certain embodiments, substantially pure form G is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form G is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form H

In certain embodiments, provided herein is form H.

In one embodiment, form H is a solid form of compound 1. In another embodiment, form H is crystalline. In one embodiment, form H is a solvated form of compound 1. In one embodiment, form H is a DMSO solvated form of compound 1. In one embodiment, form H is a DMSO semi-solvated form of compound 1.

In certain embodiments, form H provided herein is obtained by an equilibration experiment, an evaporation experiment, a cooling recrystallization experiment, and an anti-solvent recrystallization experiment. In certain embodiments, form H is obtained from certain solvent systems, which include DMSO.

In certain embodiments, provided herein is a method of making form H, comprising the steps of: 1) mixing 2-chloro-4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) pyrimidine-5-carboxamide with tert-butylamine and DMSO; 2) heating to a temperature (e.g., about 55 to about 80 ℃, such as about 68 ℃) for a period of time (e.g., about 40 hours to about 80 hours, such as about 60 hours); 3) cooling to ambient temperature; 4) adding water; and 5) collecting the solid and optionally drying. In one embodiment, the temperature is from about 55 to about 80 ℃, such as about 68 ℃. In one embodiment, the period of time is from about 40 hours to about 80 hours, such as about 60 hours. In another embodiment, water is added over a period of about 1 hour to about 4 hours, such as about 2 hours.

In certain embodiments, provided herein are solid forms. E.g., form H, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form H has an X-ray powder diffraction pattern substantially as shown in figure 34. In one embodiment, form H has one or more characteristic X-ray powder diffraction peaks at about 8.69, 9.74, 10.23, 12.17, 14.64, 15.38, 16.33, 17.22, 18.04, 18.55, 20.10, 20.62, 21.76, 23.10, 24.18, 25.65, 26.18, 26.78, 27.27, 27.83, 28.43, 29.50, 30.00, 30.54, 31.03, 32.07, 32.65, 33.41, 33.74, 34.86, 35.25, 35.77, 36.22, 36.62, 37.08, 37.59, or 38.78 ° 2 θ as depicted in fig. 34. In a specific embodiment, form H has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 9.74, 10.23, 14.64, 17.22, 18.04, 18.55, 21.76, or 24.18 ° 2 Θ. In another embodiment, form H has one, two, three, or four characteristic X-ray powder diffraction peaks at about 9.74, 17.22, 18.04, or 21.76 ° 2 Θ. In another embodiment, form H has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-seven, twenty-eight, twenty-nine, thirty-ten, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, or thirty-seven characteristic X-ray powder diffraction peaks as shown in table 15.

In one embodiment, provided herein is a crystalline form of compound 1 having a TGA thermogram substantially corresponding to the representative TGA thermogram as depicted in figure 35. In certain embodiments, the TGA thermogram exhibited by the crystalline form when heated from about 25 ℃ to about 300 ℃ includes a total mass loss of about 11.2% of the total mass of the sample at about 75 ℃ to about 175 ℃. Thus, in certain embodiments, the crystalline form loses about 11.2% of its total mass when heated from ambient temperature to about 300 ℃. In certain embodiments, the crystalline form comprises 0.5 molar equivalents of solvent in the crystal lattice, corresponding to about 0.5 moles of DMSO per mole of compound 1. The theoretical DMSO content of the DMSO hemisolvate of compound 1 was 10.8 wt%, matching the observed TGA weight loss. In certain embodiments, the crystalline form is a DMSO hemisolvate of compound 1.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 36 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with a maximum at about 160 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 36 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 222 ℃.

In yet another embodiment, form H is substantially pure. In certain embodiments, substantially pure form H is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form H is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Form I

In certain embodiments, provided herein is form I.

In one embodiment, form I is a solid form of compound 1. In another embodiment, form I is crystalline. In one embodiment, form I is a solvated form of compound 1. In one embodiment, form I is a sulfolane solvated form of compound 1. In one embodiment, form I is a 0.75 molar sulfolane solvated form of compound 1.

In certain embodiments, form I provided herein is obtained by a cooling recrystallization experiment and an anti-solvent recrystallization experiment. In certain embodiments, form I is obtained from a solvent system comprising sulfolane and water. In certain embodiments, form I is obtained from a solvent mixture of sulfolane and water (e.g., about 1: 1).

In certain embodiments, a solid form provided herein, e.g., form I, is substantially crystalline as shown by, e.g., X-ray powder diffraction measurements. In one embodiment, form I has an X-ray powder diffraction pattern substantially as shown in figure 38. In one embodiment, form I has one or more characteristic X-ray powder diffraction peaks at about 7.94, 10.50, 10.80, 11.86, 13.54, 13.92, 14.79, 16.00, 17.26, 18.27, 18.82, 19.48, 19.78, 20.65, 21.31, 21.78, 22.83, 23.53, 24.12, 24.75, 25.66, 26.29, 27.71, 28.18, 28.73, 29.17, 30.01, 30.52, 31.18, 31.60, 31.85, 32.36, 32.93, 33.59, 34.20, 34.76, 35.42, 36.56, or 37.67 ° 2 θ as depicted in fig. 38. In a specific embodiment, form I has one, two, three, four, five, six, seven, or eight characteristic X-ray powder diffraction peaks at about 7.94, 10.50, 11.86, 16.00, 17.26, 18.27, 20.65, or 24.12 ° 2 Θ. In another embodiment, form I has one, two, three, or four characteristic X-ray powder diffraction peaks at about 7.94, 16.00, 18.27, or 20.65 ° 2 Θ. In another embodiment, form I has one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, fifteen, sixteen, seventeen, eighteen, nineteen, twenty-one, twenty-two, twenty-three, twenty-four, twenty-five, twenty-six, twenty-eight, twenty-nine, thirty-ten, thirty-one, thirty-two, thirty-three, thirty-four, thirty-five, thirty-six, thirty-seven, thirty-eight, thirty-nine, thirty-eight, or thirty-nine characteristic X-ray powder diffraction peaks as shown in table 16.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 39 comprising an endothermic event with a maximum at about 118 ℃ when heated from about 25 ℃ to about 300 ℃.

In one embodiment, provided herein is a crystalline form of compound 1, having a DSC thermogram as depicted in figure 39 when heated from about 25 ℃ to about 300 ℃, the DSC thermogram comprising an endothermic event with an onset temperature of about 213 ℃.

In yet another embodiment, form I is substantially pure. In certain embodiments, substantially pure form I is substantially free of other solid forms, such as amorphous solids. In certain embodiments, substantially pure form I is not less than about 95% pure, not less than about 96% pure, not less than about 97% pure, not less than about 98% pure, not less than about 98.5% pure, not less than about 99% pure, not less than about 99.5% pure, or not less than about 99.8% pure.

Amorphous solid

In certain embodiments, provided herein are amorphous solids of compound 1.

In certain embodiments, the amorphous solid provided herein is obtained by heat treating form a. In certain embodiments, the heat treatment process comprises: (1) the temperature of equilibrium form a at a particular temperature (e.g., about 25 ℃); (2) heating to a first temperature (e.g., about 235 ℃) at a first rate (e.g., about 10 ℃ per minute); (3) isothermally holding for a first period of time (e.g., about 2 minutes); (4) cooling at a second rate (e.g., about 30 ℃ per minute) to a second temperature (e.g., about-10 ℃); (5) adjusting the temperature at a third rate (e.g., about 0.64 ℃ per 40 seconds); (6) isothermally holding for a second period of time (e.g., about 5 minutes); (7) heating to a third temperature (e.g., about 213 ℃) at a fourth rate (e.g., about 3 ℃ per minute); and (8) collecting the resulting solid.

In one embodiment, the amorphous solid has an X-ray powder diffraction pattern substantially as shown in figure 41.

In one embodiment, provided herein is an amorphous solid of compound 1 having a DSC thermogram as depicted in figure 42, the DSC thermogram comprising a glass transition temperature of 106.6 ℃ when heated from about 25 ℃ to about 300 ℃.

In yet another embodiment, the amorphous solid of compound 1 is substantially pure. In certain embodiments, the substantially pure amorphous solid of compound 1 is substantially free of other solid forms, such as form a, form B, form C, form D, form E, form F, form G, form H, and form I. In certain embodiments, the substantially pure amorphous solid has a purity of no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 98.5%, no less than about 99%, no less than about 99.5%, or no less than about 99.8%.

Application method

The solid form of compound 1 is useful as a medicament for treating, preventing or ameliorating a condition in an animal or a human. Furthermore, solid forms of compound 1 have activity against protein kinases, in particular JNK1 and/or JNK 2. Thus, provided herein are a number of uses for solid forms of compound 1, including the treatment or prevention of those diseases described below. The methods provided herein comprise administering to a subject in need thereof an effective amount of one or more solid forms of compound 1.

In one aspect, provided herein is a method of inhibiting a kinase in a cell expressing said kinase, comprising contacting said cell with an effective amount of a solid form of compound 1. In one embodiment, the kinase is JNK1, JNK2, or a mutant or isoform thereof, or a combination thereof. For example, the solid form of compound a is form a, form B, form C, form D, form E, form F, form G, form H, form I, an amorphous solid, or a mixture thereof. In further aspects, provided herein are solid forms of compound 1 for use in such methods of inhibiting a kinase in a cell expressing said kinase.

In another aspect, provided herein are methods for treating or preventing one or more disorders selected from the group consisting of: interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus, comprising administering to a subject in need thereof an effective amount of a solid form of compound 1. In some such embodiments, the lupus is lupus erythematosus (e.g., discoid lupus erythematosus or cutaneous lupus erythematosus) or systemic lupus. In further aspects, provided herein are solid forms of compound 1 for use in such methods of treating or preventing one or more disorders selected from the group consisting of: interstitial pulmonary fibrosis, systemic sclerosis, scleroderma, chronic allograft nephropathy, antibody-mediated rejection or lupus.

In another aspect, provided herein is a method for treating or preventing a liver fibrosis disorder, such as non-alcoholic steatohepatitis, steatosis (i.e., fatty liver), cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, hepatitis, hepatocellular carcinoma, and liver fibrosis with chronic or repeated alcohol intake (alcoholic hepatitis), infection (e.g., viral infection such as HCV), liver transplantation, or drug-induced liver injury (e.g., acetaminophen toxicity), comprising administering to a subject in need thereof an effective amount of a solid form of compound 1. In some such aspects, provided herein are methods for treating or preventing diabetes or metabolic syndrome leading to liver fibrosis disorders such as non-alcoholic steatohepatitis, steatosis (i.e., fatty liver), cirrhosis, primary sclerosing cholangitis, primary biliary cirrhosis, and hepatitis, comprising administering to a subject in need thereof an effective amount of a solid form of compound 1. In further aspects, provided herein are solid forms of compound 1 for use in such methods.

In another aspect, provided herein is a method for treating or preventing a disorder treatable or preventable by inhibiting JNK1 and/or JNK2, the method comprising administering to a subject in need thereof an effective amount of a solid form of compound 1. Examples of such conditions include rheumatoid arthritis; rheumatoid spondylitis; osteoarthritis; asthma, bronchitis; allergic rhinitis; chronic obstructive pulmonary disease; cystic fibrosis; inflammatory bowel disease; irritable bowel syndrome; mucous colitis; ulcerative colitis; crohn's disease; huntington's disease; hepatitis; pancreatitis; nephritis; multiple sclerosis; lupus erythematosus; type II diabetes; obesity; atherosclerosis; restenosis after angioplasty; left ventricular hypertrophy; myocardial infarction; stroke; ischemic injury to the heart, lungs, intestines, kidneys, liver, pancreas, spleen and brain; acute or chronic organ transplant rejection; preservation of organs for transplantation; organ failure or limb impairment (e.g., including but not limited to, that caused by ischemia reperfusion loss, trauma, severe physical injury, car accident, crush loss, or graft failure); graft versus host disease; (ii) endotoxic shock; multiple organ failure; psoriasis; burns caused by exposure to fire, chemicals or radiation; eczema; dermatitis; skin grafting; ischemia; ischemic conditions associated with surgery or traumatic injury (e.g., traffic accidents, gunshot injuries, or limb compression); epilepsy; alzheimer's disease; parkinson's disease; immune response to bacterial or viral infection; cachexia; angiogenesis and proliferative diseases; a solid tumor; and cancers of various tissues, such as colon, rectum, prostate, liver, lung, bronchi, pancreas, brain, head, neck, stomach, skin, kidney, cervix, blood, larynx, esophagus, mouth, pharynx, bladder, ovary, or uterus. In other aspects, provided herein are solid forms of compound 1 for use in such methods. In general, all compounds of the present invention are intended for use in methods of treating all diseases disclosed.

Pharmaceutical compositions and routes of administration

The solid form of compound 1 can be administered orally, topically or parenterally to patients in the form of conventional preparations such as capsules, microcapsules, tablets, granules, powders, lozenges, pills, suppositories, injections, suspensions, syrups, patches, creams, lotions, ointments, gels, sprays, solutions and emulsions. Suitable formulations may be prepared by a commonly employed method using conventional organic or inorganic additives such as excipients (e.g., sucrose, starch, mannitol, sorbitol, lactose, glucose, cellulose, talc, calcium phosphate, or calcium carbonate), binders (e.g., cellulose, methylcellulose, hydroxymethylcellulose, polypropylpyrrolidone, polyvinylpyrrolidone, gelatin, acacia, polyethylene glycol, sucrose, or starch), disintegrants (e.g., starch, carboxymethylcellulose, hydroxypropyl starch, low-substituted hydroxypropylcellulose, sodium bicarbonate, calcium phosphate, or calcium citrate), lubricants (e.g., magnesium stearate, light anhydrous silicic acid, talc, or sodium lauryl sulfate), flavoring agents (e.g., citric acid, menthol, glycine, or orange powder), preservatives (e.g., sodium benzoate, sodium bisulfite, methyl or propyl paraben), stabilizers (e.g., citric acid, sodium citrate or acetic acid), suspending agents (e.g., methylcellulose, polyvinylpyrrolidone or aluminum stearate), dispersing agents (e.g., hydroxypropylmethylcellulose), diluents (e.g., water), and waxes (e.g., cocoa butter, white petrolatum or polyethylene glycol). An effective amount of the solid form of compound 1 in the pharmaceutical composition can be at a level that will exhibit the desired effect; for example, from about 0.005 mg/kg subject body weight to about 10mg/kg donor body weight in a unit dose for oral and parenteral administration.

The dosage of the solid form of compound 1 to be administered to a subject is quite widely variable and can be at the discretion of the health care practitioner. In general, the solid form of compound 1 can be administered to a subject at a dose of about 0.005mg/kg body weight of the subject to about 10mg/kg body weight of the subject one to four times a day, but the above dose can be appropriately changed depending on the age, body weight and medical condition of the subject and the administration type. In one embodiment, the dose is from about 0.01mg/kg to about 5mg/kg, from about 0.05mg/kg to about 1mg/kg, from about 0.1mg/kg to about 0.75mg/kg, or from about 0.25mg/kg to about 0.5 mg/kg of subject body weight. In one embodiment, one dose is administered per day. The amount of solid form of compound 1 administered in any given case will depend on factors such as the solubility of the active ingredient, the formulation used, and the route of administration. In one embodiment, the application of the local concentration provides an intracellular exposure or concentration of about 0.01-10 μ M.

In another embodiment, provided herein is a method for treating or preventing a disease or disorder, comprising administering to a subject in need thereof from about 0.375 mg/day to about 750 mg/day, from about 0.75 mg/day to about 375 mg/day, from about 3.75 mg/day to about 75 mg/day, from about 7.5 mg/day to about 55 mg/day, or from about 18 mg/day to about 37 mg/day of compound 1 in a solid form.

In another embodiment, provided herein is a method for treating or preventing a disease or disorder, comprising administering to a subject in need thereof a solid form of compound 1 at about 1 mg/day to about 1200 mg/day, about 10 mg/day to about 1200 mg/day, about 100 mg/day to about 1200 mg/day, about 400 mg/day to about 1200 mg/day, about 600 mg/day to about 1200 mg/day, about 400 mg/day to about 800 mg/day, about 60 mg/day to about 720 mg/day, about 240 mg/day to about 720 mg/day, or about 600 mg/day to about 800 mg/day. In specific embodiments, the methods disclosed herein comprise administering to a subject in need thereof 400 mg/day, 600 mg/day, or 800 mg/day of a solid form of compound 1.

In another embodiment, provided herein is a method for treating or preventing a disease or disorder, comprising administering to a subject in need thereof about 10 mg/day to about 720 mg/day, about 10 mg/day to about 480 mg/day, about 60 mg/day to about 720 mg/day, or about 240 mg/day to about 720 mg/day of compound 1 in a solid form.

In another embodiment, provided herein is a unit dose formulation comprising solid forms of compound 1 of about 10mg and 100mg, about 1mg and 200mg, about 35mg to about 1400mg, about 125mg to about 1000mg, about 250mg to about 1000mg, or about 500mg to about 1000 mg.

In specific embodiments, provided herein are unit dose formulations comprising about 100mg or 400mg of a solid form of compound 1.

In another embodiment, provided herein is a unit dose formulation comprising about 1mg, 5mg, 10mg, 15mg, 20mg, 30mg, 35mg, 50mg, 60mg, 70mg, 100mg, 120mg, 125mg, 140mg, 175mg, 200mg, 240mg, 250mg, 280mg, 350mg, 480mg, 500mg, 560mg, 700mg, 720mg, 750mg, 1000mg, or 1400mg of solid form of compound 1.

In another embodiment, provided herein is a unit dose formulation comprising about 10mg, 30mg, or 100mg of the solid form of compound 1.

The solid form of compound 1 can be administered once, twice, three times, four times or more daily. In a specific embodiment, a dose of 600mg or less is administered as a once daily dose, and a dose of more than 600mg is administered twice daily in an amount equal to half the total daily dose. In one embodiment, the solid form of compound 1 can be administered once daily for 14 days.

The solid form of compound 1 may be administered orally for convenience. In one embodiment, when administered orally, a solid form of compound 1 is administered with the meal and water. In another embodiment, the solid form of compound 1 is dispersed in water or fruit juice (e.g., apple juice or orange juice) and administered orally as a suspension.

The solid form of compound 1 can also be administered intradermally, intramuscularly, intraperitoneally, transdermally, intravenously, subcutaneously, intranasally, epidurally, sublingually, intracerebrally, intravaginally, transdermally, rectally, transmucosally, by inhalation, or topically to the ear, nose, eye, or skin. The mode of administration is at the discretion of the health care practitioner and may depend in part on the site of the medical condition.

In one embodiment, provided herein are capsules containing a solid form of compound 1 without additional carriers, excipients, or vehicles.

In another embodiment, provided herein are compositions comprising an effective amount of a solid form of compound 1 and a pharmaceutically acceptable carrier or vehicle, wherein the pharmaceutically acceptable carrier or vehicle may comprise an excipient, diluent, or mixture thereof. In one embodiment, the composition is a pharmaceutical composition.

The compositions may be in the form of tablets, chewable tablets, capsules, solutions, parenteral solutions, lozenges, suppositories, suspensions and the like. The compositions may be formulated to contain the daily dose or a suitable fraction of the daily dose in a dosage unit which may be a single tablet or capsule or a suitable volume of liquid. In one embodiment, the solution is prepared from a water soluble salt such as a hydrochloride salt. In general, all compositions are prepared according to known methods in pharmaceutical chemistry. Capsules can be prepared by mixing the solid form of compound 1 with a suitable carrier or diluent and filling the capsules with the appropriate amount of the mixture. Commonly used carriers and diluents include, but are not limited to, inert powdered materials, such as various starches; powdered cellulose, especially crystalline and microcrystalline cellulose; sugars such as fructose, mannitol and sucrose; cereal flour; and similar edible powders.

Tablets may be prepared by direct compression, wet granulation or dry granulation. The preparation is usually added with a diluent, a binder, a lubricant and a disintegrating agent and the compound. Typical diluents include, for example, various types of starch, lactose, mannitol, kaolin, calcium phosphate or sulfate, inorganic salts (e.g., sodium chloride), and powdered sugar. Powdered cellulose derivatives are also useful. Typical tablet binders are substances such as starch, gelatin and sugars (e.g. lactose, fructose, glucose, etc.). Natural and synthetic gums are also suitable, including acacia, alginate, methylcellulose, polyvinylpyrrolidone, and the like. Polyethylene glycol, ethyl cellulose, and waxes may also serve as binders.

Lubricants in tablet formulations may require lubricants to prevent the tablets and punches from sticking in the die. Lubricants may be selected from such slippery solids as talc, magnesium and calcium stearate, stearic acid and hydrogenated vegetable oils. Tablet disintegrants are substances that swell when wet to break up the tablet and release the compound. Including starches, clays, celluloses, algins, and gums. More specifically, for example, corn and potato starch, methylcellulose, agar, bentonite, wood cellulose, powdered natural sponge, anion exchange resin, alginic acid, guar gum, citrus pulp and carboxymethyl cellulose, and sodium lauryl sulfate may be used. The tablets may be coated with sugar as a flavoring and sealing agent, or with a film-forming protective agent to modify the dissolution properties of the tablet. The compositions may also be formulated as chewable tablets, for example by using a substance such as mannitol in the formulation.

When it is desired to administer compound 1 in solid form as a suppository, a typical base may be used. Cocoa butter is a traditional suppository base that may be modified by adding waxes to slightly raise its melting point. Water-miscible suppository bases, in particular comprising polyethylene glycols of various molecular weights, are widely used.

In certain embodiments, the pharmaceutical compositions provided herein comprise form a, including substantially pure form a.

In certain embodiments, the pharmaceutical compositions provided herein comprise form B, including substantially pure form B.

In certain embodiments, the pharmaceutical compositions provided herein comprise form C, including substantially pure form C.

In certain embodiments, the pharmaceutical compositions provided herein comprise form D, including substantially pure form D.

In certain embodiments, the pharmaceutical compositions provided herein comprise form E, including substantially pure form E.

In certain embodiments, the pharmaceutical compositions provided herein comprise form F, including substantially pure form F.

In certain embodiments, the pharmaceutical compositions provided herein comprise form G, including substantially pure form G.

In certain embodiments, the pharmaceutical compositions provided herein comprise form H, including substantially pure form H.

In certain embodiments, the pharmaceutical compositions provided herein comprise form I, including substantially pure form I.

In certain embodiments, the pharmaceutical compositions provided herein comprise an amorphous solid, including a substantially pure amorphous solid.

In certain embodiments, the pharmaceutical compositions provided herein comprise a mixture of one or more solid forms of compound 1, including form a, form B, form C, form D, form E, form F, form G, form H, form I, and amorphous solids, wherein each possible combination of solid forms of compound 1 is possible.

Examples

The following examples are provided by way of illustration and not limitation. The following abbreviations are used in the description and examples:

ACN: acetonitrile

Am, and (2): amorphous form

Amphos: p-dimethylaminophenyl di-tert-butylphosphine

APT: active pharmaceutical ingredient

Boc: tert-butyloxycarbonyl radical

n-BuOH: n-butanol

dba: diphenylmethylene acetone

DBU: 1, 8-diazabicyclo [5.4.0] undec-7-ene

DCM: methylene dichloride

DIPEA: n, N-diisopropylethylamine

DMAc: n, N-dimethyl acetamide

DMF: n, N-dimethylformamide

DMSO, DMSO: dimethyl sulfoxide

DSC: differential scanning calorimetry

DVS: dynamic gas phase adsorption

EDTA: ethylenediaminetetraacetic acid

ESI: electrospray ionization

EtOAc: ethyl acetate

EtOH: ethanol

FTTR: fourier transform infrared spectroscopy

HPLC: high performance liquid chromatography

IPA: 2-propanol

IPAc: acetic acid isopropyl ester

LCMS: liquid chromatography-mass spectrometry

MEK: methyl ethyl ketone

MeOH: methanol

2-MeTHF: 2-methyltetrahydrofuran

mp: melting Point

MS: mass spectrometry

MTBE: methyl tert-butyl ether

NBS: n-bromosuccinimide

NMP: n-methyl-2-pyrrolidone

NMR: nuclear magnetic resonance

RH: relative humidity

RT: at room temperature

Rx recrystallization

S: solvent(s)

SDTA: single differential thermal analysis

SM: starting material

(S) - (-) -5, 5-bis (diphenylphosphino) -4, 4-di-1, 3-benzodioxole

S-SegPhos alkenes

TA: thermal analysis

Tf: trifluoromethanesulfonic acid ester or trifluoromethanesulfonyl group

TFA: trifluoroacetic acid

TFE: 2, 2, 2-trifluoroethanol

TGA: thermogravimetric analysis

TGA-MS/TG-MS: coupling of thermogravimetric analysis with mass spectrometry

THF: tetrahydrofuran (THF)

TLC: thin layer chromatography

XRPD: powder X-ray diffraction

Synthetic examples

The following non-limiting synthetic examples show methods for preparing compound 1. NAMEs of chemical structures were generated using ACD/NAME (Advanced Chemistry Development, inc., Ontario, Canada) and chemical structures were plotted using Chemdraw (cambridge soft, Perkin Elmer, Waltham, MA).

Example 1: 2- (tert-butylamino) -4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl ] amino } pyrimidine-5-carboxamide

2-chloro-4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl]Amino } pyrimidine-5-carboxamide: (1R, 2R, 5R) -5-amino-2-methylcyclohexanol hydrochloride (16.0kg), 2, 4-dichloropyrimidine-5-carboxamide (19.0kg), K were added to a reactor at 25 ℃2CO3(14.9kg) and THF (160L). The batch was cooled to 0 ℃ and water (160L) was added. The batch was stirred at 0 ℃ for a further 1 hour, warmed to 25 ℃ and held for 16 hours. Water (288L) was added to the batch while maintaining the batch at 25 ℃, the batch was cooled to 15 ℃ and stirring was continued for 4 hours. The batch was filtered, rinsed twice with water (2X 80L) and dried in a vacuum oven at 40 deg.C for 24 hours under a nitrogen purge to give 2-chloro-4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl ] ]Amino } pyrimidine-5-carboxamide as a white powder (23.3kg, 86% yield).1H NMR(DMSO-d6)δ0.93(d,J=5.7Hz,3H),0.97-1.29(m,4H),1.63-1.68(m,1H),1.75-1.88(m,1H),2.09-2.13(m,1H), 3.00-3.08(m,1H),3.80-3.95(m,1H),4.65(d,J=5.1Hz,1H),7.69(br.s.,1H), 8.20(br.s.,1H),8.53(s,1H),9.22(d,J=7.5Hz,1H)。

2- (tert-butylamino) -4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl]Amino } pyrimidine-5-carboxamide (compound 1): the reactor was charged with 2-chloro-4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl]Amino } pyrimidine-5-carboxamide (41kg), tert-butylamine (105.3kg) and DMSO (205L). The batch was heated to 68 ℃ under a nitrogen pressure of 10psig, held for 80 hours, and cooled to 25 ℃. The batch was filtered through a 0.45 μm in-line filter into the second reactor. The batch was heated to 60 ℃ and loaded with water (205L) through a 0.45 μm in-line filter. The batch was seeded with micronized compound 1(820 g), stirred at 60 ℃ for more than 1 hour, and water (615L) was loaded to the batch through a 0.45 μm in-line filter over 3 hours at 60 ℃. The batch was stirred at 60 ℃ for 1 hour, cooled to 25 ℃ over 6 hours, filtered and washed with water (410mL) and filtered through a 0.45 μm in-line filter. The solid was dried in a vacuum oven at 40 ℃ for over 72 hours under a nitrogen purge to give 2- (tert-butylamino) -4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl)]Amino } pyrimidine-5-carboxamide, as form a and white solid (43.5kg, 94% yield). 1H NMR(DMSO-d6)δ0.95 (d,J=6.2Hz,3H),0.97-1.28(m,4H),1.37(s,9H),1.60-1.75(m,1H),1.83-2.00 (m,IH),2.06-2.26(m,1H),2.86-3.07(m,1H),3.74-4.01(m,1H),4.59(d,J= 5.7Hz,1H),6.65(br.s.,1H),7.03(br.s.,1H),7.57(br.s.,1H),8.36(s,1H), 8.93(br.s.,1H)。

2- (tert-butylamino) -4- { [ (IR, 3R, 4R) -3-hydroxy-4-methylcyclohexyl]Recrystallization of amino } pyrimidine-5-carboxamide (compound 1): the reactor was charged with 2- (tert-butylamino) -4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl]Amino } pyrimidine-5-carboxamide (30g), 2-propanol (203mL), and water (67.5 mL). The batch was heated to 35 ℃ and filtered at 35 ℃ through a 0.45 μm in-line filter into the second reactor. The first reactor and transfer line were flushed with a mixture of 2-propanol (33.75mL) and water (11.25 mL) filtered through a 0.45 μm filter. The batch was heated to 70 ℃ and water (36) was passed through a 0.45 μm in-line filter0mL) was loaded into the batch while maintaining the batch temperature at 70 ℃. The batch was cooled to 60 ℃ and the reaction mixture was cooled at 60 ℃ using compound 1(0.9g) in filtered 2-propanol: slurry in water mixture (9 mL; 1: 9v/v) was seeded. The batch was stirred at 60 ℃ for 30 minutes, cooled to 0 ℃, stirred at 0 ℃ for 14 hours, filtered, and filtered through a 0.45 μm in-line filter using 2-propanol: the water mixture (60 mL; 1: 9v/v 60mL) was washed. The batch was dried in a vacuum oven at 40 ℃ for 72 hours under a nitrogen purge to give 2- (tert-butylamino) -4- { [ (1R, 3R, 4R) -3-hydroxy-4-methylcyclohexyl) ]Amino } pyrimidine-5-carboxamide, as form a and white solid (26g, 85% yield).1H NMR(DMSO-d6)δ0.95(d,J=6.2Hz,3H),0.97-1.28(m,4H),1.37 (s,9H),1.60-1.75(m,1H),1.83-2.00(m,1H),2.06-2.26(m,1H),2.86-3.07(m,1H),3.74-4.01(m,1H),4.59(d,J=5.7Hz,1H),6.65(br.s.,1H),7.03(br.s., 1H),7.57(br.s.,1H),8.36(s,1H),8.93(br.s.,1H)。

Example 2: 4- (tert-butylamino) -2- ((trans-4-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide

4- (tert-butylamino) -2-chloropyrimidine-5-carboxamide: a mixture of 2, 4-dichloro-pyrimidine-5-carboxamide (10.0g), DIPEA (11mL) in NMP (30mL) was stirred at 5 ℃. Tert-butylamine (6.6mL) was loaded to the mixture and the mixture was stirred at 25 ℃ for 16 h. Water (100mL) was added to the mixture at 25 ℃. The mixture was stirred for 1 hour. The suspension was filtered, washed with water (50mL) and dried in a vacuum oven at 40 ℃ for 24 hours under a nitrogen purge to give 4- (tert-butylamino) -2-chloropyrimidine-5-carboxamide as a white solid (8.7g, 84%).1H NMR(DMSO-d6) δ9.41(s,1H),8.55(s,1H),8.19(s,1H),7.67(s,1H),1.42(s,9H)。

4- (tert-butylamino) -2- ((trans-4-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide: 4- (tert-butylamino) -2-chloropyrimidine-5-carboxamide (0.5g), trans-4-aminocyclohexanol hydrochloride (b)0.40g)、 Na2CO3A mixture (0.28g) in NMP (3.5mL) was heated and held at 85 deg.C for 6 hours. The mixture was cooled to 35 ℃ and water (10mL) was added. After 30 minutes, the batch was cooled to 25 ℃ and held for 1 hour. The suspension was filtered, washed with water (2.5mL) and dried in a vacuum oven at 40 ℃ for 24 hours under a nitrogen purge to give 4- (tert-butylamino) -2- ((trans-4-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide as a white solid (0.6g, 89%). 1H NMR(DMSO-d6) δ 9.17 (width, s, 1H), 8.32(s, 1H), 7.01 (width, s, 1H), 4.52(d, J ═ 4.5Hz, 1H), 3.70-3.25(m, 2H), 1.84(m, 4H), 1.41(s, 9H), 1.33-1.16(m, 4H).

Recrystallization of 4- (tert-butylamino) -2- ((trans-4-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide: a mixture of 4- (tert-butylamino) -2- ((trans-4-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide (0.2g) in ethanol (1.0mL) was heated to 60 ℃ and held for 30 minutes. Water (4mL) was loaded over 1 hour. The mixture was cooled to 25 ℃ over 1 hour and held for 1 hour. The suspension was filtered, washed with water (4mL) and dried in a vacuum oven at 40 ℃ for 24 hours under a nitrogen purge to give 4- (tert-butylamino) -2- ((trans-4-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide (0.18g, 90% yield).1H NMR(DMSO-d6) δ 9.17 (width, s, 1H), 8.32(s, 1H), 7.01 (width, s, 1H), 4.52(d, J ═ 4.5Hz, 1H), 3.70-3.25(m, 2H), 1.84(m, 4H), 1.41(s, 9H), 1.33-1.16(m, 4H).

Example 3: 4- (bicyclo [1.1.1] pent-1-ylamino) -2- (((1R, 3S) -3-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide

4- (bicyclo [ 1.1.1)]Pent-1-ylamino) -2-chloropyrimidine-5-carboxamide: 2, 4-dichloro-pyrimidine-5-carboxamide (2g), bicyclo [1.1.1] was stirred at 25 deg.C ]A mixture of pentan-1-amine hydrochloride (1.18g), sodium bicarbonate (1.75g) and NMP (10mL) for 24 hours. Water (10mL) was charged and the reaction temperature was maintained below 30 ℃ and the mixture was stirred at 25 ℃ for 2 hours.The suspension was filtered and washed with NMP: water (1: 110 mL) then water (2X10mL) and dried in a vacuum oven at 40 ℃ under a nitrogen purge to give 4- (bicyclo [ 1.1.1)]Pent-1-ylamino) -2-chloropyrimidine-5-carboxamide as a white solid (1.97g, 83% yield).1H NMR(DMSO-d6)δ2.14(s,6H),2.51-2.53 (m,1H),7.76(br.s.,1H),8.23(br.s.,1H),8.60(s,1H),9.57(s,1H)。

4- (bicyclo [ 1.1.1)]Pent-1-ylamino) -2- (((1R, 3S) -3-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide: 4- (bicyclo [ 1.1.1)]A mixture of pent-1-ylamino) -2-chloropyrimidine-5-carboxamide (44g), (1S, 3R) -3-aminocyclohexanol (27.6g), potassium carbonate (38.2g) and DMSO (300mL) was heated at 85 ℃ for 12 hours. After cooling to room temperature, water (2L) and a mixture of THF and EtOAc (1: 1, 2L) were added. The aqueous phase was separated and the organic layer was washed with saturated brine (2L). The organic layer was concentrated under reduced pressure to give a crude product as a purple foam which was triturated with hot acetonitrile (1L). After cooling to room temperature, the solid was filtered and washed with acetonitrile (200 mL). The solid was dried in a vacuum oven at 50 ℃ to give 4- (bicyclo [ 1.1.1) ]Pent-1-ylamino) -2- (((1R, 3S) -3-hydroxycyclohexyl) amino) pyrimidine-5-carboxamide as an off-white solid (4g, 79% yield).1H NMR(DMSO-d6)δ0.91-1.31(m,4H),1.60-1.89(m,3H),2.01-2.20(m, 7H),3.34(s,1H),3.37-3.52(m,1H),3.58-3.85(m,1H),4.65(d,J=4.3Hz, 1H),6.90(br.s.,1H),7.20(d,J=7.9Hz,1H),7.61(br.s.,1H),8.37(s,1H), [9.23(s,0.14H)],9.41(s,0.86H)。

Example 4: 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide

A mixture of 2-chloro-4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) pyrimidine-5-carboxamide (4g), tert-butylamine (14mL) and DMSO (20mL) was heated to 68 ℃ and held for 60 hours. After cooling to room temperature, water (20X volume, 80mL) was added over 2 hours. The slurry was stirred for 2 hours and the crude product was collected by suction filtration as the DMSO hemisolvate of 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) -pyrimidine-5-carboxamide (form H).

Example 5: synthetic route 1 for (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt

Scheme 1 was used to prepare (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt starting from limonene. Epoxidation of (-) -limonene using m-CPBA yields compound (Y). Using O3Cleavage of the double bond in compound (Y) followed by Baeyer-Villiger oxidation provides compound (3). The epoxide of compound (3) is converted back to olefin (4). Reductive hydrolysis of the acetyl group in compound (4) affords alcohol (5). The chiral center of compound (5) is converted by the sequence of tosylation, azide addition and reduction to give compound (7). Using Boc 2O protects compound (7) to yield compound (8). Introduction of the trans hydroxyl group by hydroboration/oxidation of compound (8) affords a 1: 1 mixture of diastereomers of compounds (9a) and (9 b). The diastereoisomers were separated by chiral SFC to give compound (9 a). Deprotection of compound (9a) using an acid such as HCl affords (1R, 2R, 5R) -5-amino-2-methylcyclohexanol HCl salt (A).

Example 6: synthetic route 2 for (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt are prepared starting from isoprene using scheme 2. Scheme 2 shares intermediate compound (8) with scheme 1. The asymmetric D-A reaction of isoprene (12) and ester (11) in the presence of catalysts (15) and (16) provided compound (17) in > 98% ee. The catalyst (15) is formed by the reaction of the compound (13) and the compound (14). Hydrolysis of compound (17) with a base (e.g., LiOH or NaOH) provides acid (18). Curtis rearrangement of (18) using Diphenylphosphorylazide (DPPA) followed by tert-butanol addition gave stereochemically retained compound (8). Introduction of a trans hydroxyl group by hydroboration/oxidation of the compound (8) gives a mixture of diastereomers of the compounds (9a) and (9 b). When (+) -diisopinocampheylborane prepared from (-) - α -pinene and borane-methyl sulfide is used as the borohydride, a ratio of 5-8: 1 of compounds 9a and 9b is obtained. The diastereomers were separated by recrystallization using MTBE to afford compound 9 a. Deprotection of compound 9a with acid affords (1R, 2R, 5R) -5-amino-2-methylcyclohexanol HCl salt (a). Enantiomeric purity can be further improved by recrystallization from 2-propanol.

During the formation of compound (17), several reaction conditions affect the enantioselectivity:

triflimide (16) loading: the loading of trifluoromethanesulfonimide (16) must be lower than that of catalyst (15). As shown in the following Table, in the case where the catalyst (15) was in excess (e.g., 0.3 eq: 0.2eq, 0.24 eq: 0.20eq, and 0.24 eq: 0.15eq, respectively) relative to the trifluoromethanesulfonimide (16), the enantioselectivity and the conversion rate were high. However, loading only 0.05 eq of trifluoromethanesulfonimide in the reaction completed above gave compound (17) at different% ee. When the total amount of trifluoromethanesulfonimide (16) as in columns 1 and 2 was lower than catalyst (15), no decrease in ee was observed. When the total amount of trifluoromethanesulfonimide (16) as in columns 3 and 4 is higher than catalyst (15), the ee of compound (17) decreases to 50% in 1 hour, then to 0% after 2.5 h. When the amount of catalyst (15) (0.18eq) was lower than trifluoromethanesulfonimide (16) (0.20eq) at the beginning of the reaction, compound (17) had 50% at the 1h time point and was completely racemic after 16h (column 5).

Loading a catalyst: the loading of the catalyst (15) is 5-20 mol%. When the reaction was carried out at-20 ℃, compound (17) had 99% ee regardless of catalyst loading.

Reaction temperature: higher reaction temperatures result in lower enantioselectivities. The reaction is preferably carried out at-20-0 ℃ to obtain > 98% ee.

Example 7: synthesis of (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt route 3

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared starting from isoprene using scheme 3. D-A reaction of isoprene with acryloyl chloride (19) affords racemic compound (20). Resolution of compound (20) using a chiral amine such as (S) -or (R) -phenethylamine gives enantiomerically enriched acid (18). Following the procedure of scheme 2, the Curtis rearrangement of (18) gives compound (8) retaining stereochemistry. Reagents other than azidodiphenylphosphate, e.g. CDI/NH2OH/tBuOH, can be used in Curtis rearrangement reactions. Introduction of a trans hydroxyl group by hydroboration/oxidation of the compound (8) gives a mixture of diastereomers of the compounds (9a) and (9 b). When diisopinocampheylborane is used as the borohydride, a yield of-5-8: 1 of compounds (9a) and (9 b). The diastereoisomers were separated by recrystallization using MTBE to give compound (9 a). Deprotection of compound (9a) using an acid such as HCl affords (1R, 2R, 5R) -5-amino-2-methylcyclohexanol HCl salt (A).

Example 8: synthesis route 4 of (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared starting from isoprene using scheme 4. Chiral compound (21) (R ═ iPr, CH2Ph) and isoprene to give compound (22). Hydrolysis of compound (22) affords intermediate compound (18). Like scheme 2, the Curtis rearrangement of (18) gives (8). Introduction of a trans hydroxyl group by hydroboration/oxidation of the compound (8) gives a mixture of diastereomers of the compounds (9a) and (9 b). When (+) -diisopinocampheylborane is used as the borohydride, a ratio of compounds (9a) and (9b) of-5-8: 1 is obtained. The diastereoisomers were separated by recrystallization using MTBE to give compound (9 a). Deprotection of compound 5 with acid provides (1R, 2R, 5R) -5-amino-2-methylcyclohexanol HCl salt (a).

Example 9: synthetic route 5 for (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared starting from nitroethylene (23) and isoprene using scheme 5. D-A reaction of nitroethylene (23) and isoprene gives racemic compound (24). Hydroboration/oxidation of compound (24) introduces the trans hydroxyl group to give a mixture of the four diastereomers (25 a-d). Diastereomer (25) is treated with a base such as NaOH, NaOEt or KOtBu to give a mixture of two enantiomers s (25a) and (25 b). (25a) Reduction of the nitro group of (25b) to give amines (26a) and (26 b). The compounds (26a) and (26b) are separated by resolution or chiral SFC to give (1R, 2R, 5R) -5-amino-2-methylcyclohexanol or its HCl salt (a).

Example 10: synthesis of (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt route 6

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared starting from amine (27) using scheme 6. The use of compound (28) to protect amine (27) as described in international patent application publication WO2012/145569 gives phthalimide (29). Using acids such as H2SO4/KHSO4Dehydration affords olefin (30). Deprotection of (30) affords amine (31). The amine was protected to give racemic (32). The trans-hydroxyl group is introduced by hydroboration/oxidation of compound (32) to give a mixture of the four diastereomers (9 a-d). Compound 9a was purified by chiral SFC as described in scheme 1. Deprotection of compound 9a using an acid such as HCl provides (1R, 2R, 5R) -5-amino-2-methylcyclohexanol HCl salt (a).

Example 11: synthesis of (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt route 7

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared using scheme 7 starting from (R) -acid (18), which can be prepared as described in scheme 2. (18) Lactonization of the resulting product to give the lactone (33). (33) Reaction with an alkoxide (such as NaOMe or NaOiPr) affords an epoxide (34). By Ti (OiPr) 4the/Mg/TMSCl opened the epoxide to give (35). (35) is hydrolyzed, followed by Curtis rearrangement of (36) to give (1R, 2R, 5R) -5-amino-2-methylcyclohexanol or its HCl salt.

Example 12: synthesis of (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt route 8

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared starting from aniline (37) using scheme 8. Reduction of (37) by catalytic hydrogenation gives compound (38). Using reducing agents (e.g. NaBH)4) Reduction of (38) affords compound (39). Using chiral SFC pureThe compound (39) is reacted to provide (1R, 2R, 5R) -5-amino-2-methylcyclohexanol or its HCl salt (a) alternatively, the amine of the compound (39) is protected using a Boc group to provide a mixture of diastereomers (40) the compound (40) is purified by chiral SFC to provide compound (9a) deprotection of compound (9a) using an acid such as HCl provides (1R, 2R, 5R) -5-amino-2-methylcyclohexanol HCl salt (a).

Example 13: synthesis of (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt route 9

(1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt can be prepared starting from methyl ethyl ketone using scheme 9. (41) The reaction of (A) and (42) gives the diketone (43). A chiral amine such as (S) -phenethylamine or (R) -phenethylamine is added to the ketone to give (44). Resolution of (44) gives enantiomerically enriched (44 a). Reduction of compound (44a) yields a mixture of diastereomers (45). Compound (45a) is purified by chiral SFC or resolution. The hydrogenation deprotection of compound (45a) provides (1R, 2R, 5R) -5-amino-2-methylcyclohexanol or its HCl salt (a).

Example 14: synthetic route 10 for (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt

Scheme 10 can be used to prepare (1R, 2R, 5R) -5-amino-2-methylcyclohexanol and its HCl salt starting from compound (46). Ketal formation followed by epoxidation of (46) affords (47). By using AlMe3The MeLi opens the epoxide to introduce the trans alcohol, giving (48a) and (48 b). Compound (48a) is purified by chiral SFC or resolution. Deprotection of (48a) affords ketone (49). (49) Reaction with hydroxylamine produces a hydroxyimine (50). (50) Tosylation of (a) gives tosylimide (51). Using Pd (CF)3CO2)2/S-SegPhos/H2TFE or other chiralityAsymmetric reduction of (51) by the catalyst affords tosylamide (52). Deprotection of compound (52) provides (1R, 2R, 5R) -5-amino-2-methylcyclohexanol or a salt thereof (a).

Solid forms

Analytical method

Polymorphic screens of compound 1 were performed to investigate whether different solid forms could be produced under different conditions (e.g. different solvents, temperature and humidity changes).

Solvents used for polymorph screening were HPLC or reagent grade including n-BuOH, acetone, ACN/water, DCM, DMSO, EtOAc, EtOH/water, heptane, IPA, MEK, MeOH, MTBE, THF/water, toluene and water.

All solid samples generated in polymorph screening were analyzed by XRPD. Use of Cu Ka radiation on a PANalytic Empyrean or Thermo ARL X' TRA X-ray powder diffractometerXRPD analysis was performed.

The PANalytical Empyrean instrument is equipped with a fine focus X-ray tube. The voltage and the amount of current of the X-ray generator were set at 45kV and 40mA, respectively. The diverging slots are set at 1/16 ° and 1/8 °, and the receiving slots are set at 1/16 °. Diffracted radiation was measured using a Pixel 2D detector. The continuous theta-2 theta scan from 3 deg. to 40 deg. 2 theta is set to step size 0.013 or 0.026 with the sample rotation speed set to 4. Peak positions were checked using sintered alumina standards.

Thermo ARL X' TRA instruments are equipped with a fine focus X-ray tube. The voltage and current amounts of the X-ray generator were set to 45kV and 40mA, respectively. The divergence slits were set at 4mm and 2mm, and the measurement slits were set at 0.5mm and 0.2 mm. Diffracted radiation was measured using a Peltier-cooled Si (Li) solid state detector. Continuous scans of theta-2 theta at 2.40 deg./min (0.5sec/0.02 deg. steps) from 1.5 deg. to 40 deg. 2 theta were used. Sintered alumina standards were used to check peak positions.

DSC analysis was performed on a TA Discovery differential scanning calorimeter. Indium was used as a calibration standard. About 2-5mg of the sample was placed in a DSC pan. The sample was heated at a rate of 10 ℃/min under nitrogen until the final temperature was 300 ℃. Melting points are reported as extrapolated onset temperatures.

TGA analysis was performed on a TA Discovery thermogravimetric analyzer. Calcium oxalate was used for performance check. An accurately weighed sample of about 2-10mg was placed on a tray and loaded into a TGA oven. The sample was heated at a rate of 10 ℃/min under nitrogen until the final temperature was 300 ℃.

Morphological analysis of the samples was performed on an Even Mini SEM. A small amount of sample was spread on a sample holder, then coated with gold and viewed using 500x magnification.

Hygroscopicity was measured on Surface Measurement Systems DVS. Typically, 5-20mg sample sizes are loaded into DVS instrument sample trays and the samples are analyzed on a DVS automated sorption analyzer at room temperature. The relative humidity was increased from 0% to 90% RH and then to 95% RH in 10% RH steps. The relative humidity is then reduced in a similar manner to achieve a complete adsorption/desorption cycle.

1H NMR spectra were obtained on a Bruker 300MHz NMR spectrometer. Dissolving the sample in DMSO-d6And analyzed using 32 scans.

Equilibration/slurry and Evaporation experiments

Equilibration (also known as slurry experiments) and evaporation experiments were performed by adding an excess of compound 1 up to 2 mL of the test solvent. The resulting mixture was stirred at room temperature and 50 ℃ for at least 24 h, respectively. Upon reaching equilibrium, the saturated supernatant was removed, filtered using a 0.45 μm PTFE filter and allowed to evaporate in an open vial under nitrogen at room temperature and 50 ℃ respectively. The solid from the equilibration was isolated and air dried before analysis.

Equilibration experiments were performed at room temperature and 50 ℃ using form a as starting material. The results are summarized in table 1. The solid separated from the MTBE, heptane and water as form a was confirmed by XRPD pattern. All other solvents gave new forms. The solid isolated from acetone, DCM, THF and THF/water was designated form B. Solids isolated from EtOH/water, EtOH, ACN/water and IPA were designated form C. The solid isolated from MeOH is designated form D. The solid isolated from n-BuOH is designated form E. The solid isolated from toluene is designated form F. The solid isolated from EtOAc was designated form G. The solid isolated from DMSO was designated form H. All forms except form a were found to be solvates during further characterization.

TABLE 1 equilibration of form A at room temperature and 50 deg.C

-: is not carried out

The evaporation experiments were carried out at room temperature and 50 ℃. The results are summarized in table 2. Solvents showing sufficient solubility for form a gave similar solvate forms as observed during the equilibration experiments.

TABLE 2 Evaporation experiments for form A at room temperature and 50 deg.C

-: cannot be analyzed

Antisolvent recrystallization and cooling recrystallization experiments

For cooling recrystallization, each selected solvent (MeOH, EtOH/water) was saturated with compound 1 at 60 ℃. The solution was stirred at 60 ℃ for 10 minutes, filtered using a 0.45 μm PTFE syringe filter, then allowed to cool to room temperature naturally and then placed in a refrigerator. The solid obtained from recrystallization was isolated and air dried before analysis.

For anti-solvent recrystallization, the selected solvents (MeOH, EtOH, IPA, and EtOAc) were saturated with compound 1 at 60 ℃. Once the solid was completely dissolved, a portion of the solution was filtered into a pre-heated vial and the selected anti-solvent (water, MTBE or heptane) was added at 60 ℃. The mixture was naturally cooled to room temperature, and then placed in a refrigerator. The solid resulting from recrystallization was isolated and air dried before analysis.

MeOH, EtOH/water, IPA and EtOAc were used as the single or primary solvent. Water, MTBE and heptane were used as anti-solvents. The results are summarized in table 3. Crystallization using only water as the anti-solvent yields form a. All other solvents or solvent combinations gave solvate forms similar to those observed during the equilibrium experiments.

TABLE 3 summary of recrystallization experiments

n/a: not applicable.

Additional experiments were performed using DMSO as the main solvent. The isolated solid was found to be a new form and was designated form H.

Transformation experiments

Additional format conversion experiments were performed to determine the interconversion between solid forms. The results are summarized in table 4. The solvated form was kept isothermally at 150 ℃ for 5min to give a solid identical to form a. All aqueous slurries also yielded form a.

TABLE 4 transformation experiments for Compound 1

Starting solid form Solvent/conditions Temperature/condition XRPD results
Form B Heating of In 1Keeping the temperature at 50 ℃ for 5 minutes Form A
Form C Heating of Maintaining at 150 deg.C for 5 min Form A
Form D Heating of Maintaining at 150 deg.C for 5 min Form A
Form E Heating of Maintaining at 150 deg.C for 5 min Form A
Form F Heating of Maintaining at 150 deg.C for 5 min Form A
Form G Heating of Maintaining at 150 deg.C for 5 min Form A
Form H Heating of Maintaining at 150 deg.C for 5 min Form A
Form B Slurry in water RT, 5 days Form A
Form C Slurry in water RT, 5 days Form A
Form D Slurry in water RT, 5 days Form A
Form E Slurry in water RT, 5 days Form A
Form F Slurry in water RT, 5 days Form A
Form G Slurry in water RT, 5 days Form A
Form H Slurry in water RT, 5 days Form A

Summary of polymorphic forms

A total of eight crystalline forms of compound 1 were found in this polymorph screening study. A stacked view of these forms of XRPD patterns is shown in fig. 37, and the physical characteristics are summarized in table 5.

TABLE 5 summary of physical characterization of the crystalline form of Compound 1

n/a: is not available.

Form A

Form a is a non-stoichiometric channel hydrate crystalline solid form of compound 1. This form is obtained mainly from recrystallization or slurry experiments in aqueous or "water-rich" solvent systems.

Form a can also be obtained by conversion from form H. A mixture of crude form H (4g) and water (40 mL) was heated to 70 ℃ for 3 hours. After cooling to room temperature, the product was collected by suction filtration. The wet cake was dried in a vacuum oven at 40 ℃ for 16 hours under a nitrogen purge to give 2- (tert-butylamino) -4- ((1R, 3R, 4R) -3-hydroxy-4-methylcyclohexylamino) pyrimidine-5-carboxamide as form a and a white solid (3.54g, 80%).

The effect of temperature (22 ℃ -70 ℃) and water composition in DMSO (50% -88%) on the stability of form a and form H of compound 1 is reflected in table 6 and fig. 45. This information indicates that form a is the thermodynamically stable form in a water-rich water/DMSO mixture (> 70%).

TABLE 6 Stable form of form H after slurry experiments at 50 to 88% water/DMSO ratio and 22 ℃ to 70 ℃

Form A was favored by 1: 1 (50% water) to 1: 4 DMSO: water (80% water) at 60 ℃ and remained as form A in 70-88% water in DMSO at 22 ℃. 70% of the water in DMSO is at the edge of the form transition between form a and form H. Thus, the final solvent composition was chosen to be 80% water in DMSO. These results show that for the synthesis of compound 1, using 5X volumes of DMSO, after completion of the reaction, addition of 20X volumes of 60 ℃ water to the reaction mixture will provide compound 1 as form a.

Form a has the crystalline XRPD pattern shown in figure 1. The crystal habit is cubic or rod-like, as shown in FIG. 2. The TGA and DSC thermograms of form a are shown in fig. 4 and 5, respectively. The DSC thermogram shows only one major event, with an onset temperature of 223 ℃, corresponding to melting/decomposition. A TGA weight loss of 0.45% was observed up to 150 ℃. Of form A1The H NMR spectrum was consistent with the structure of compound 1 (see fig. 7).

Moisture sorption/desorption behavior of form a was determined by DVS. The results are summarized in fig. 6. A 2.3% total mass change was observed between 0 and 95% RH with a 1.3% sharp change between 0 and 10% RH. The XRPD diffractogram of the sample showed no change after undergoing adsorption/desorption cycles (see fig. 8). A sharp change between 0 and 10% RH was observed for several samples, but the amount of water uptake varied between samples. For all form a samples analyzed, the total water uptake between 0 and 95% RH was about 0.5% to 2%.

Form a was further characterized using single crystal X-ray diffraction. The structure was analyzed in space group P2(1), 2 (1). The crystal data and structure refinement are summarized in table 7. The powder x-ray pattern was calculated and matched to the observed experimental XRPD pattern of form a, as shown in figure 1. Fractional occupancy of water molecules is found in the crystal lattice. Inclusion of about 20% occupancy reduced the R factor from 5.2% to 3.6%. The unit cell packing diagram along the b-axis as shown in fig. 2 shows channeled water molecules in the lattice. These observations suggest that form a is a channel hydrate. The theoretical water content is 1.1 wt% for 0.2 molar equivalents of water and 2.7 wt% for 0.5 molar equivalents of water.

TABLE 7 form A Crystal data and Structure refinement

Form a stability was further characterized by compression testing and form conversion experiments. At about 1 minute of applied 2000-psi pressure, the material was still in form a with a slightly broader diffraction peak (see fig. 9). The results from the form conversion experiments in table 4 show that all solvated forms were converted to form a after desolvation by heating or slurrying in water. These results indicate that form a is the most stable or developable form of compound 1.

Figure 1 provides an XRPD pattern for form a. A list of X-ray diffraction peaks for form a is provided in table 8 below.

TABLE 8X-ray diffraction peaks of form A

Fig. 3 is an SEM image of form a.

Form A has an inherent solubility of 0.038mg/mL after 24 hours at 25 ℃ and 0.289mg/mL at pH 4.5. Although form a is a channel hydrate, it has relatively slow water absorption at room temperature. However, form a can potentially absorb up to 3% water after 7 months of storage at 40 ℃/75% RH. Water absorption may strongly depend on the humidity of the storage conditions, so it is recommended to protect compound 1 from moisture during storage.

Form B

Form B is prepared from form A in acetone, CH 2Cl2Or recrystallization in THF or slurry experiments. Form B has a crystalline XRPD pattern as shown in figure 10. TGA and DSC thermograms of form B obtained from acetone are shown in fig. 11 and 12, respectively. The TGA weight loss of 8.5 wt% corresponds to a small broad DSC peak at about 147 ℃ and can be attributed to solvent loss in form B. Initial temperatureThe major DSC peak at 223 ℃ corresponds to the melting/degradation of form a. Obtaining samples of form B1H-NMR spectrum and showed about 0.5 molar equivalent of acetone (see FIG. 13). The theoretical acetone content of the hemisolvate of compound 1 was 8.3 wt% matching the observed TGA weight loss. These observations indicate that form B is the acetone hemisolvate of compound 1. Form conversion experiments showed that heating form B above the desolvation temperature yielded form a. A slurry of form B in water also produces form a.

A list of X-ray diffraction peaks for form B is provided in table 9 below.

TABLE 9X-ray diffraction peaks of form B

FIG. 13 provides form B1H NMR(DMSO-d6) Wherein δ 0.94(d, J ═ 6.4Hz, 3H), 0.96-1.04(m, 1H), 1.04-1.28(m, 3H), 1.36(s, 9H), 1.60-1.74(m, 1H), 1.83-1.98 (m, 1H), 2.09(s, 3H, acetone), 2.10-2.19(m, 1H), 2.89-3.04(m, 1H), 3.76-3.99 (m, 1H), 4.57(d, J ═ 5.5Hz, 1H), 6.64(br.s., 1H), 6.94(br.s., 1H), 7.51 (br.s., 1H), 8.34(s, 1H), 8.93(br.s., 1H).

Form C

Form C was obtained from recrystallization or slurry experiments of form a in EtOH/water, EtOH, ACN or IPA. Form C has the crystalline XRPD pattern shown in figure 14. TGA and DSC thermograms of form C obtained from EtOH/water are shown in fig. 15 and 16, respectively. The TGA weight loss of 7.3 wt% corresponds to a small broad DSC peak at about 143 ℃ and can be attributed to solvent loss in form C. The main DSC peak with an onset temperature of 224 ℃ corresponds to the melting/degradation of form a. Obtaining samples of form C1H-NMR spectrum and showed about 0.5 molar equivalent of EtOH (see FIG. 17). The theoretical EtOH content of the hemisolvate of Compound 1 is6.7 wt%, matching the observed TGA weight loss. These observations indicate that form C is an ethanol hemisolvate of compound 1. Form conversion experiments showed that heating form C above the desolvation temperature yielded form a. A slurry of form C in water also produced form a.

A list of X-ray diffraction peaks for form C is provided in table 10 below.

TABLE 10X-ray diffraction peaks of form C

FIG. 17 provides form C1H NMR(DMSO-d6) Wherein δ 0.94(d, J ═ 6.4Hz, 3H), 1.00-1.27(m, 5.6H) { includes 1.02(t, J ═ 7.0Hz, 1.6H, ethanol) }, 1.36(s, 9H), 1.67(dd, J ═ 3.3, 13.1Hz, 1H), 1.81-2.00(m, 1H), 2.10-2.24(m, 1H), 2.87-3.05 (m, 1H), 3.32(s, 4H), 3.44(qd, J ═ 5.1, 7.0Hz, 1H, ethanol), 3.74-3.99(m, 1H), 4.35(t, J ═ 5.1Hz, 1H), 4.57(d, J ═ 5.7, 1H), 6.92 (m, 1H), 6.92, 1H, 1 br, 1H), 7.7.7.7H, 6H, 6.92, 6H, 1 br, 1H, and br 1H.

Form D

Form D was obtained from recrystallization or slurry experiments of form a in MeOH. Form D has the crystalline XRPD pattern shown in figure 18. The TGA and DSC thermograms of form D are shown in fig. 19 and 20, respectively. A TGA weight loss of about 4 wt% corresponds to a small DSC peak at about 170 ℃ and can be attributed to solvent loss in form D. The main DSC peak with an onset temperature of 223 ℃ corresponds to the melting/degradation of form a. Obtaining samples of form D1H-NMR spectrum and showed about 0.5 molar equivalent of MeOH (see FIG. 21). The theoretical MeOH content of the hemisolvate of compound 1 was 4.7 wt%, similar to the observed TGA weight loss. These observations indicate that form D is a methanol hemisolvate of compound 1. Form conversion experiment showsHeating form D above the desolvation temperature affords form a. A slurry of form D in water also produced form a.

A list of X-ray diffraction peaks for form D is provided in table 11 below.

TABLE 11X-ray diffraction peaks of form D

FIG. 21 provides form D1H NMR(DMSO-d6) Wherein δ 0.94(d, J ═ 6.4Hz, 3H), 0.96-1.04(m, 1H), 1.05-1.28(m, 3H), 1.36(s, 9H), 1.67(dd, J ═ 3.1, 13.1Hz, 1H), 1.84-1.97(m, 1H), 2.08-2.20(m, 1H), 2.86-3.04(m, 1H), 3.17(d, J ═ 5.3Hz, 1.6H, methanol), 3.76-3.99(m, 1H), 4.09(q, J ═ 5.3Hz, 1H), 4.57(d, J ═ 5.5Hz, 1H), 6.65(br.s., 1H), 6.95(br.s., 1H), 7.47 (br.47, 8.47, 1H), 1.93 (br.8, 8H., 1H).

Form E

Form E was obtained from recrystallization or slurry experiments of form a in n-BuOH. Form E has the crystalline XRPD pattern shown in figure 22. TGA and DSC thermograms of form E are shown in fig. 23 and 24, respectively. The TGA weight loss of 10.3 wt% corresponds to a small broad DSC peak at about 124 ℃ and can be attributed to solvent loss in form E. The main DSC peak with an onset temperature of 224 ℃ corresponds to the melting/degradation of form a. Obtaining samples of form E1H-NMR spectrum and showed about 0.5 molar equivalent of n-BuOH (see FIG. 25). The theoretical n-BuOH content of the hemisolvate of Compound 1 was 10.3 wt% matching the TGA weight loss observed. These observations indicate that form E is an n-BuOH hemisolvate of compound 1. Form conversion experiments showed that heating form E above the desolvation temperature gave form a. A slurry of form E in water also produces form a.

A list of X-ray diffraction peaks for form E is provided in table 12 below.

TABLE 12X-ray diffraction peaks of form E

FIG. 25 provides form E1H NMR(DMSO-d6) Wherein δ 0.85(t, J ═ 7.2Hz, 1.5H, n-butanol), 0.94(d, J ═ 6.4Hz, 3H), 0.96-1.04(m, 1H), 1.04-1.25(m, 3H), 1.25-1.46 (m, 11H) { { includes 1.36(s, 9H), 1.3-1.46(m, 2H, n-butanol) }, 1.67(dd, J ═ 3.2, 13.0Hz, 1H), 1.81-2.00(m, 1H), 2.10-2.24(m, 1H), 2.86-3.05(m, 1H), 3.35-3.44(m, 1H, n-butanol), 3.75-3.99(m, 1H), 4.31(t, J ═ 5.2H, 0.57H), 7.7.7H, 7H, 7 br, 7.7H, 7, 7.7 br, 6H, 1H, 3 br, 7.7H, 7 br, 7H, 1H, and br.

Form F

Form F was obtained from recrystallization or slurry experiments of form a in toluene. Form F has the crystalline XRPD pattern shown in figure 26. The diffuse character of the diffractogram indicates low crystallinity of the sample. TGA and DSC thermograms of form F are shown in fig. 27 and 28, respectively. The TGA weight loss of 6.9 wt% corresponds to a small broad DSC peak at about 113 ℃ and can be attributed to solvent loss in form F. The main DSC peak with an onset temperature of 223 ℃ corresponds to the melting/degradation of form a. Obtaining samples of form F1H-NMR spectrum and showed about 0.3 molar equivalents of toluene (see fig. 29), matching the observed TGA weight loss. These observations indicate that form F is a 0.3 mole toluene solvate of compound 1. Form conversion experiments showed that heating form F above the desolvation temperature gave form a. A slurry of form F in water also produced form a.

A list of X-ray diffraction peaks for form F is provided in table 13 below.

TABLE 13X-ray diffraction peaks of form F

FIG. 29 provides form F1H NMR(DMSO-d6) Wherein δ 0.94(d, J ═ 6.4Hz, 3H), 0.96-1.04(m, 1H), 1.04-1.29(m, 3H), 1.35(s, 9H), 1.67(dd, J ═ 3.3, 13.1Hz, 1H), 1.90(d, J ═ 9.3Hz, 1H), 2.06-2.23(m, 1H), 2.30(s, 0.9H, toluene), 2.89-3.04 (m, 1H), 3.71-4.00(m, 1H), 4.57(d, J ═ 5.7Hz, 1H), 6.64(br.s., 1H), 6.94(br.s., 1H), 7.08-7.30(m, 1.4H, toluene), 7.50 (br.8, 1H., 8.93, 1H., 1H), 1.93.

Form G

Form G was obtained from recrystallization or slurry experiments of form a in EtOAc. Form G has the crystalline XRPD pattern shown in figure 30. The TGA and DSC thermograms for form G are shown in figure 31 and figure 32, respectively. The TGA weight loss of 11.9 wt% corresponds to a small broad DSC peak at about 116 ℃ and can be attributed to solvent loss in form G. The main DSC peak with an onset temperature of 223 ℃ corresponds to the melting/degradation of form a. Obtaining samples of form G1H-NMR spectrum and showed about 0.5 molar equivalent of EtOAc (see FIG. 33). The theoretical EtOAc content of the hemisolvate of compound 1 was 12.1 wt% matching the observed TGA weight loss. These observations indicate that form G is the EtOAc hemisolvate of compound 1. Form conversion experiments showed that heating form G above the desolvation temperature yielded form a. A slurry of form G in water also produces form a.

A list of X-ray diffraction peaks for form G is provided in table 14 below.

TABLE 14X-ray diffraction peaks of form G

FIG. 33 provides form G1H NMR(DMSO-d6) Wherein δ 0.94(d, J ═ 6.4Hz, 3H), 0.96-1.04(m, 1H), 1.04-1.29(m, 5H) { includes 1.17(t, J ═ 9.0Hz, EtOAc) }, 1.29-1.46(m, 9H), 1.60-1.76(m, 1H), 1.86-1.96(m, 1H), 1.99(s, 1.4H, EtOAc), 2.04-2.16(m, 1H), 2.88-3.06(m, 1H), 3.75-3.97(m, 1H), 4.03(q, J ═ 7.1Hz, 1H, EtOAc), 4.57(d, J ═ 5.7Hz, 1H), 6.65(br.s., 1H), 6.94 (br.7, 1H), 1H, 52.52, br.8, br.3H, 1H, 3.3.6.6.6.6.

Form H

Form H was obtained from recrystallization or slurry experiments of form a in DMSO. Form H has the crystalline XRPD pattern shown in figure 34. TGA and DSC thermograms of form H are shown in fig. 35 and fig. 36, respectively. The TGA thermogram shows a step weight loss of 11.2 wt%, corresponding to a small broad DSC peak at about 160 ℃, and can be attributed to solvent loss in form H. The main DSC peak with an onset temperature of 222 ℃ corresponds to the melting/degradation of form a. The theoretical DMSO content of the hemisolvate of compound 1 was 10.8 wt%, matching the observed TGA weight loss. These observations indicate that form H is a DMSO hemisolvate of compound 1. Form conversion experiments showed that heating form H above the desolvation temperature gave form a. A slurry of form H in water also produced form a.

A list of X-ray diffraction peaks for form H is provided in table 15 below.

TABLE 15X-ray diffraction peaks of form H

Of form H1H NMR (MeOD) provides δ 1.03(d, J ═ 6.2Hz, 3H), 1.05-1.19(m, 1H), 1.19-1.38(m, 3H), 1.45(s, 9H), 1.78(dq, J ═ 3.3, 13.2Hz, 1H), 1.90-2.16 (m, 1H), 2.16-2.40(m, 1H)),2.65(s,3H,DMSO),2.95-3.24(m,1H), 3.85-4.21(m,1H),8.25(s,1H)。

Form I

Form I was obtained from recrystallization of form a in sulfolane and water (1: 1). Form I has the crystalline XRPD pattern shown in figure 38. The DSC thermogram for form I is shown in figure 39. The DSC peak at about 118 ℃ can be attributed to solvent loss in form I. The main DSC peak with a maximum temperature of 213 ℃ corresponds to the melting/degradation of form a. Of form I 1The H-NMR spectrum showed about 0.75 molar equivalent of sulfolane (see FIG. 40). These observations indicate that form I is a 0.75 mole sulfolane solvate of compound 1.

A list of X-ray diffraction peaks for form I is provided in table 16 below.

TABLE 16X-ray diffraction peaks of form I

FIG. 40 provides form I1H NMR(DMSO-d6) Wherein δ 0.94(d, J ═ 6.2Hz, 3H), 0.96-1.04(m, 1H), 1.11(s, 3H), 1.36(s, 9H), 1.59-1.74(m, 1H), 1.83-1.98 (m, 1H), 2.00-2.20(m, 4H), 2.80-3.18(m, 4H), 3.74-4.02(m, 1H), 4.57(d, J ═ 5.5Hz, 1H), 6.64(br.s., 1H), 7.02(br.s., 1H), 7.60(br.s., 1H), 8.34(s, 1H), 8.82-9.06(m, 1H).

Amorphous solid

The amorphous solid of compound 1 was obtained from the heat treatment of form a. The heat treatment process comprises the following steps: (1) equilibrating the temperature of form a at 25 ℃; (2) heating to 235 ℃ at a speed of 10 ℃/min; (3) keeping the temperature for 2 minutes; (4) cooling to-10 deg.C at 30 deg.C/min; (5) adjusting the temperature to 0.64 ℃ every 40 seconds; (6) keeping the temperature for 5 minutes; (7) heating to 213 deg.C at a rate of 3 deg.C per minute; and (8) collecting the resulting solid.

The amorphous solid had an XRPD spectrum as shown in figure 41. The DSC thermogram of the amorphous solid sample is shown in figure 42. The amorphous solid has a glass transition temperature of about 106.6 ℃.

FIG. 43 and FIG. 44 provide amorphous solids1H-NMR spectrum and LCMS.

Biological examples

Biochemical analysis

A. Time resolved fluorescence analysis

JNK1 analysis.384-well time resolved fluorescence assays can be used to monitor JNK1 activity. The JNK1 assay can be performed in the following assay buffer: 50mM HEPES, 10mM MgCl21mM EGTA, 2mM DTT and 0.01% Tween 20. To initiate the reaction, a ULight of 100nM may be usedTMLabeled 4EBP1 peptide (Perkin-Elmer) and 5. mu.M ATP were mixed with 500pM JNK1 (Carna Biosciences) to give a total assay volume of 20. mu.L in each well. The assay can be incubated at room temperature for 1 hour and stopped by adding 20 μ L of stop solution to each well using a mixture of 30mM EDTA and 4nM Eu-anti-4 EBP 1. The plates can be read on a Perkin-Elmer Envision reader.

JNK2 analysis384-well time resolved fluorescence assay can be used to monitor JNK2 activity. The JNK2 assay can be run in the following assay buffer: 50mM HEPES, 10mM MgCl21mM EGTA, 2mM DTT and 0.01% Tween 20. To initiate the reaction, ULight at 100nMTMThe labeled 4EBP1 peptide (Perkin-Elmer) and 5. mu.M ATP can be mixed with 500pM JNK2(Carna Biosciences) to give a total assay volume of 20. mu.L in each well. The assay can be incubated at room temperature for 1 hour and stopped by adding 20 μ L of stop solution to each well using a mixture of 30mM EDTA and 4nM Eu-anti-4 EBP 1. The plates can be read on a Perkin-Elmer Envision reader.

B.Cascade analysis

JNK1 analysis.JNK1The cascade kinase assay can be run in the following buffers: 50mM HEPES pH 7.5, 0.01% BRIJ-35, 10mM MgCl21mM EGTA and 1mM DTT. 10 μ L of kinase reaction mix containing 1.81-7.25ng JNK1, 25ng inactive MAPKAPK2, 100 μ M ATP and 2 μ M Ser/Thr 04 peptide can be prepared. The assay can be incubated at room temperature for 1 hour. Next, 5. mu.L of 1: 512 diluted chromogenic reagent A (Invitrogen, PV3295) can be added to the reaction mixture and incubation at room temperature continued for 1 h. The data can then be read on a fluorescent plate reader and analyzed.

JNK2 analysis.JNK2The cascade kinase assay can be run in the following buffers: 50mM HEPES pH 7.5, 0.01% BRIJ-35, 10mM MgCl21mM EGTA, 2mM DTT. 10 μ L of kinase reaction mix containing 0.38-1.5ng JNK2, 100ng inactive MAPKAPK2, 100 μ M ATP and 2 μ M Ser/Thr 04 peptide can be prepared. The assay can be incubated at room temperature for 1 h. Next, 5. mu.L of 1: 512 diluted chromogenic reagent A (Invitrogen, PV3295) can be added to the reaction mixture and incubation at room temperature continued for 1 h. The data can then be read on a fluorescent plate reader and analyzed.

C. Radioactivity analysis

JNK1 analysis.The radioactive JNK kinase assay can be performed in a 96-well plate format in a final volume of 100. mu.L. The final assay concentration can be 6.6. mu.M ATP (3 times ATP Km), 2.64 to 5. mu.g/mL JNK1, and 100. mu.g/mL cJUN. JNK1 can be diluted in the following dilution buffer (20mM HEPES pH 7.6, 0.1mM EDTA, 2.5mM MgCl)20.004% (w/v) Triton X100, 2. mu.g/ml leupeptin, 20mM glycerol B-phosphate, 0.1mM Na3VO4Dithiothreitol) and then diluted in substrate solution buffer (20mM HEPES pH 7.6, 50mM NaCl, 0.1mM EDTA, 2.5mM MgCl20.05% (w/v) Triton X100). JNK1/cJun cocktail (85. mu.l) can be added to inhibitor (5. mu.l) diluted in 100% DMSO to give a final DMSO assay concentration of 5% (v/v). Can allow forThe enzyme, substrate and inhibitor mixture was equilibrated at room temperature for 15 minutes. Can be prepared by adding 10. mu.L of kinase buffer (130mM MgCl)26mM dithiothreitol, 150mM p-nitrophenyl phosphate, 100. mu. Ci/ml. gamma-, [ solution of sodium dithiothreitol ], [ solution of sodium phosphate ] and sodium phosphate33P]-ATP) 10X ATP in the reaction was initiated. The reaction may be allowed to proceed for 60 minutes, and then the protein precipitated by trichloroacetic acid (final 7.2% TCA). After 30 minutes incubation with TCA, the reaction product can be collected onto a glass microfilter 96-well plate (Millipore MAHF CIH60) using a Packard Filtermate. The precipitate can be washed using phosphate buffered saline and the amount of phosphate incorporated into cJun can be quantified by scintillation counting using Packard Topcount-NXT. All assays can be performed under conditions where phosphate incorporation can be linear with respect to time and enzyme concentration. IC (integrated circuit) 50Values can be calculated as the concentration of inhibitor that can reduce c-Jun phosphorylation to 50% of control values.

JNK2 analysisThe assay can be performed in a 96-well plate format with a final volume of 100 μ L. The final assay concentration can be 6.6. mu.M ATP (3 times ATP Km), 0.2 to 0.53. mu.g/mL JNK2, and 100. mu.g/mL cJUN. JNK2 can be diluted in the following dilution buffer (20mM HEPES pH 7.6, 0.1mM EDTA, 2.5mM MgCl)20.004% (w/v) Triton X100, 2. mu.g/ml leupeptin, 20mM glycerol B-phosphate, 0.1mM Na3VO4Dithiothreitol) and then diluted in substrate solution buffer (20mM HEPES pH 7.6, 50mM NaCl, 0.1mM EDTA, 2.5mM MgCl20.05% (w/v) cJun premix in Triton X100). JNK2/cJun cocktail (85. mu.l) can be added to inhibitor (5. mu.l) diluted in 100% DMSO to give a final DMSO assay concentration of 5% (v/v). The enzyme, substrate and inhibitor mixture may be allowed to equilibrate for 15 minutes at room temperature. Can be prepared by adding 10. mu.L of kinase buffer (130mM MgCl)26mM dithiothreitol, 150mM p-nitrophenyl phosphate, 100. mu. Ci/ml. gamma-, [ solution of sodium dithiothreitol ], [ solution of sodium phosphate ] and sodium phosphate33P]-ATP) 10X ATP in the reaction was initiated. The reaction may be allowed to proceed for 60 minutes, and then the protein precipitated by trichloroacetic acid (final 7.2% TCA). After 30 minutes incubation with TCA, the reaction product can be collected onto a glass microfilter 96-well plate (Millipore MAHF CIH60) using a Packard Filtermate. Can be used for The precipitate is washed with phosphate buffered saline and the amount of phosphate incorporated into cJun can be quantified by scintillation counting using a Packard Topcount-NXT. All assays can be performed under conditions where phosphate incorporation can be linear with respect to time and enzyme concentration. IC (integrated circuit)50Values can be calculated as the concentration of inhibitor that can reduce c-Jun phosphorylation to 50% of control values.

Cell analysis

RAW264.7 phospho-cJun whole cell assay.RAW264.7 cells can be purchased from American Tissue Culture Collection (American Tissue Culture Collection) and maintained in growth Medium consisting of 90% high glucose du Modified Eagle Medium (Invitrogen), 10% fetal bovine serum (Hyclone), and 2mM L-glutamine (Invitrogen). All cells can be incubated at 37 ℃ in 95% air and 5% CO2Culturing in medium. Cells can be 1.0x105Individual cells/well density plates were plated in 120. mu.L growth medium in 96-well plates. A stock solution of diaminopyrimidine compounds (30mM) may be serially diluted in DMSO, further diluted in growth medium, and may be added to each well as a 10x concentrated solution in a volume of 15 μ Ι _ in, mixed, and allowed to incubate with the cells. The compound vehicle (DMSO) can be maintained at a final concentration of 0.2% in all wells. After 30 minutes, cells can be activated using lipopolysaccharide (ALEXIS Biochemicals) at a final concentration of 25 ng/mL. Lipopolysaccharide can be added as a 10x concentrated solution to the culture medium, and in 15 u L/hole volume. The cell plate can be cultured for 1h, after which the cell culture medium is removed. The level of c-Jun protein that can be phosphorylated at serine 63 can be measured according to the manufacturer's instructions for the whole cell lysate kit-Phospho-c-Jun (Ser 63) assay (Meso Scale Discovery), except that the NaCl concentration in the lysis buffer can be raised to a final concentration of 350 mM. IC (integrated circuit) 50Values can be calculated as the concentration of diaminopyrimidine compound that reduces the level of phospho-Jun protein to 50% of the signal window. Certain of the compounds in tables 1, 2 and 3 had an IC in this assay of 0.01-30. mu.M50The value is obtained.

Jurkat T-cell IL-2 production assay. Jurkat T cells (clone E6-1) were purchased from the American tissue culture Collection and maintained in growth medium consisting of RPMI 1640 medium (Mediatech) containing 2mM L-glutamine, with 10% fetal bovine serum (Hyclone) and penicillin/streptomycin. All cells can be incubated at 37 ℃ in 95% air and 5% CO2Culturing in medium. Cells can be 1.0x105Individual cells/well density plates were plated in 120. mu.L of medium in 96-well plates. A stock solution of diaminopyrimidine compound (20mM) may be diluted in growth medium and added to each well as a 10x concentrated solution in a volume of 15 μ Ι _, mixed, and allowed to pre-incubate with the cells for 30 minutes. The compound vehicle (dimethylsulfoxide) can be maintained at a final concentration of 0.2% in all samples. After 30 minutes, cells can be activated with PMA (phorbol myristate acetate; final concentration 50 ng/mL) and PHA (phytohemagglutinin; final concentration 1. mu.g/mL). PMA and PHA can be added as 10 Xconcentrated solutions prepared in growth medium and added at a volume of 15. mu.L/well. The cell plate can be cultured for 6 hours. Cells can be pelleted by centrifugation and the medium removed and stored at-20 ℃. Media aliquots can be analyzed according to the manufacturer's instructions for the human IL-2 tissue culture kit (Meso Scale Discovery). IC (integrated circuit) 50Values can be calculated as the concentration of diaminopyrimidine compound that reduces IL-2 production to 50% of the signal window. Certain of the compounds in tables 1, 2 and 3 had an IC in this assay of 0.01-10. mu.M50The value is obtained.

Clinical protocol

A phase 1 randomized two part study evaluating the safety, tolerability and pharmacokinetics of single and multiple escalated doses of compound 1 in healthy subjects.

The primary objective was to evaluate the safety and tolerability of single and multiple oral doses of compound 1 in healthy subjects.

A secondary objective was to assess the Pharmacokinetics (PK) of compound 1 after single and multiple oral doses.

Design of research

This is a two-part study conducted in at most two study centers.

Part 1 is a randomized, double-blind, placebo-controlled study to evaluate safety, tolerability, and PK of compound 1 in healthy subjects after a single oral dose. Researchers and study participants will be blinded to treatment throughout the study, while the sponsor will remain blind. The study design of choice was an ascending dose in sequential groups.

In part 1, approximately 56 subjects will be randomized and included in seven scheduled cohorts. Each cohort will consist of eight subjects; six subjects will receive compound 1 and two will receive placebo.

During the course of part 1, each subject will participate in the screening phase, baseline phase, treatment phase and follow-up. The subject will be screened for eligibility. Subjects meeting all inclusion criteria and not the exclusion criteria at screening will return to clinical base for baseline assessment on day-1. And will live in clinical premises from day-1 to day 4. Subjects will receive a single oral dose of the study product (IP; compound 1 or placebo) on day 1 in the fasted state according to the randomized protocol. Blood and urine samples will be collected at pre-specified times for PK and/or clinical laboratory assessments and/or exploratory analysis. Safety will be monitored throughout the study. Subjects will leave the clinical base on day 4 after completion of the required study procedure and return to the clinical base for follow-up on day 7 (± 1 day window). In the event that the subject discontinued the study, an early End (ET) follow-up will be performed.

After each contemporaneous group, the security data will be checked and the PK data will be checked as needed. Parameters were examined before each dose increment and a particular dose increment.

Part 2 is a randomized, double-blind, placebo-controlled study to evaluate safety, tolerability, and PK of compound 1 in healthy subjects after multiple oral doses (up to 14 days of administration). Researchers and study participants will be blinded to treatment throughout the study, while the sponsor will remain blind. The study design of choice was an ascending dose in sequential groups.

Part 2 did not begin until a total daily dose of up to and including 240mg had been evaluated in part 1. Only the safe and well tolerated dose in part 1 will be administered in part 2.

In part 2, approximately 48 subjects will be randomized and included in the six scheduled cohorts. Each cohort will consist of eight subjects; six subjects will receive compound 1 and two will receive placebo.

During the part 2 procedure, each subject will participate in the screening phase, baseline phase, treatment phase and follow-up. The subject will be screened for eligibility. Subjects meeting all inclusion criteria and not meeting exclusion criteria at screening will return to clinical base for baseline assessment on day 1 and will live at clinical base on days 1 to 17. The first dose of IP (compound 1 or placebo) will be administered on day 1 under a randomized regimen in the fasted state. The same total daily dose will be given in the fasted state on days 2 to 14. Blood samples will be collected at pre-specified times for PK, clinical laboratory assessments, and/or exploratory biomarkers. Urine samples will be collected at pre-specified times for clinical laboratory evaluation. Safety will be monitored throughout the study. Subjects will leave the clinical base on day 17 after completing the required study procedure and will return to the clinical base for follow-up on day 21 (± 1 day window). In the event that the subject discontinued the study, an ET follow-up will be performed.

After each contemporaneous group, the security data will be checked and the PK data will be checked as needed. Parameters were examined before each dose increment and a particular dose increment.

Study population: approximately 104 healthy adult subjects from any race (male or infertile female) aged 18 to 50 and including 18 and 50 will be included in the study, with approximately 56 subjects participating in part 1 and up to approximately 48 subjects participating in part 2.

The study duration is as follows: the estimated duration of the study from the first follow-up of the first subject to the last follow-up of the last subject, including parts 1 and 2, was about 8 months.

The estimated duration of the clinical period of part 1 from the first follow-up to the last follow-up of the first subject was about 4 months. From screening to follow-up, the estimated duration of participation in part 1 was about 4 weeks per subject.

Part 2 did not begin until a total daily dose of up to and including 240mg had been evaluated in part 1. Only the safe and well tolerated dose in part 1 will be administered in part 2. The estimated duration of the clinical period of part 1 from the first follow-up to the last follow-up of the first subject was about 6 months. From screening to follow-up, the estimated duration of participation in part 2 was about 6 weeks per subject.

The test endpoint is defined as the date the last subject completed the last follow-up of the study, or as the date the last required data point for primary, secondary and/or exploratory analysis is received from the last subject, as prescribed in the protocol and/or statistical analysis plan, at a later date.

Study treatment

Part 1:approximately 56 subjects will be randomized and included in seven planned cohorts with eight subjects per cohort. In each cohort, six subjects will receive compound 1 and two will receive placebo.

The dose in part 1 will be administered once daily (QD) as the Active Pharmaceutical Ingredient (API) (or matching placebo) in the capsule.

The following compound 1 dose levels in table 17 are intended for part 1.

TABLE 17 Compound 1 dosage levels in part 1

Contemporaneous group Compound 1 dose level (total daily dose)
1A 10mg
1B 30mg
1C 60mg
1D 120mg
1E 240mg
1F 480mg
1G 720mg

If a Gastrointestinal (GI) related event occurs, such as intolerable nausea or vomiting, the total daily dose may be reduced or BID may be given or three times daily (TID).

The study product will only be administered at one dose level at a time and the next dose will not begin until the safety and tolerability of the previous dose level has been evaluated and deemed acceptable by the investigator and sponsor's medical supervisor.

Section 2: part 2 did not begin until a total daily dose of up to and including 240mg had been evaluated in part 1. Only the safe and well tolerated dose in part 1 will be administered in part 2.

Approximately 48 subjects will be randomized and included in six scheduled cohorts with eight subjects per cohort. In each cohort, six subjects will receive compound 1 and two will receive placebo.

The planned dosing regimen in part 2 was compound 1 (or matched placebo) QD administration in capsules for 14 days. The following compound 1 dose levels in table 18 are intended for part 2.

TABLE 18 dosage levels of Compound 1 in part 2

Contemporaneous group Compound 1 dose level (total daily dose) Duration of time
2A 10mg Day by day 14 days
2B 30mg Day by day 14 days
2C 60mg Day by day 14 days
2D 120mg Day by day 14 days
2E 240mg Day by day 14 days
2F 480mg Day by day 14 days

The suggested dose level in part 2 may be modified and/or eliminated based on the data obtained by part 1. If the proposed dose escalation step needs to be changed, the maximum dose escalation step in part 2 will be ≦ 3-fold the previous dose level. In addition, the maximum dose administered in part 2 will not exceed the Maximum Tolerated Dose (MTD) in part 1 and will not exceed 480mg per day for 14 days.

If a GI-related event occurs, such as intolerable nausea or vomiting, the total daily dose may be reduced or may be administered BID or TID.

The study product will only be administered at one dose level at a time and the next dose will not begin until the safety and tolerability of the previous dose level has been evaluated and deemed acceptable by the investigator and sponsor's medical supervisor. Additionally, if a certain dose level is not tolerated in part 1, that dose level or any higher dose level will not be administered in part 2, except in cases where GI intolerance (e.g., nausea, vomiting) is mitigated by an alternative dosage regimen (i.e., BID or TID).

Safety will be monitored throughout the study. Safety assessments will include AE reports, PE, vital signs, 12-lead ECG, clinical laboratory safety tests (including liver function tests [ LFT ], total cholesterol, triglycerides, high density lipoprotein [ HDL ] and low density lipoprotein [ LDL ], in addition to standard clinical chemistry, hematology and urinalysis tests), examinations for concomitant medication/procedures, FOB testing and stool testing, and pregnancy testing of female subjects. All AEs will be monitored and recorded throughout the study from the time the Informed Consent Form (ICF) was signed until the study was completed, and when the investigator learned within 28 days after the last dose of IP (and any time after the investigator learned those SAEs after suspected to be relevant to IP). All concomitant medications and procedures will be reviewed and recorded from the time the subject signs the ICF until the study is complete. Follow-up will be scheduled for all subjects. If the subject discontinues the study for any reason, ET follow-up will be performed.

In both parts of the study, blood samples will be collected at pre-specified times to determine the amount of compound 1 in plasma. For cohorts 1C to 1G (planned dose levels of 60mg to 720mg) for part 1, urine samples will be collected at pre-specified times for exploratory metabolite analysis. Significant metabolites in plasma and urine will be determined and compound 1 in urine will be quantified as part of the exploratory assay.

The following PK parameters for compound 1 will be estimated as appropriate: maximum observed plasma concentration (C)max) (ii) a Time to Cmax (T)max) (ii) a Extrapolation from zero to infinity plasma concentration-time Curve (AUC)) (ii) a Area under plasma concentration-time curve from zero time to last quantifiable concentration (AUC)t) (ii) a Area under the plasma concentration-time curve from zero time to tau (τ) (where τ is the dosing interval) (AUC τ); terminal elimination half-life (t)1/2,z) (ii) a Apparent total plasma clearance (CL/F) when administered orally; apparent total volume of distribution (V) based on end-stage when orally administeredz/F); based on day 1 and day 14 AUCτThe accumulation Rate (RA).

If exploratory analysis indicates that compound 1 is present in large amounts in urine, validated methods can be used to further quantify the concentration of compound 1 in the urine sample collected in fraction 1. The following PK parameters associated with urinalysis can then be optionally determined: cumulative amount of unchanged excreted drug (Ae) in urine during the collection period from pre-dose (0-hour) to the end of collection; cumulative percentage of administered dose (fe) of unchanged excretion in urine during the collection period from pre-dose (0-hour) to the end of collection; renal Clearance (CL) r)。

A number of references have been cited, the disclosures of which are incorporated herein by reference in their entirety.

140页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:二氨基甲酰胺化合物的固体形式、其组合物及其使用方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!