Solid forms of echinogenesis promoter

文档序号:1909076 发布日期:2021-11-30 浏览:14次 中文

阅读说明:本技术 棘生成促进剂的固体形式 (Solid forms of echinogenesis promoter ) 是由 S·T·撒拉弗 E·B·瓦达斯 于 2020-01-30 设计创作,主要内容包括:本文提供了2-(2-(2-(2-(4-(苯并[d]噻唑-2-基)苯氧基)乙氧基)乙氧基)乙氧基)乙-1-醇(化合物I)的结晶形式:还提供了制造方法和使用该结晶形式的方法。(Provided herein are 2- (2- (2- (2- (4- (benzo [ d ]))]Crystalline forms of thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I): methods of manufacture and methods of using the crystalline forms are also provided.)

1. Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form I) characterized by an X-ray powder diffraction pattern comprising the following peaks (° 2 Θ ± 0.2 ° 2 Θ): about 4.6, about 20.8 and about 23.7, using a wavelength ofIs measured on a diffractometer.

2. Compound I form I according to claim 1, wherein the diffractogram further comprises peaks (° 2 Θ ± 0.2 ° 2 Θ) at about 9.2, about 16.3, about 18.6, and about 19.6.

3. Compound I form I according to claim 1, characterized by a Differential Scanning Calorimetry (DSC) curve that comprises an endotherm at about 70 ℃.

4. Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I, form II), characterized by an X-ray powder diffraction pattern comprising the following peaks (° 2 Θ ± 0.2 ° 2 Θ): about 9.2, about 13.8 and about 16.1, using a wavelength ofIs measured on a diffractometer.

5. Compound I form II according to claim 4, wherein the diffractogram further comprises peaks (° 2 Θ ± 0.2 ° 2 Θ) at about 6.9, about 11.5, and about 18.4.

6. Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I, form III) characterized by an X-ray powder diffraction pattern comprising the following peaks (° 2 Θ ± 0.2 ° 2 Θ): about 7.7, about 10.3 and about 15.3, using a wavelength ofIs measured on a diffractometer.

7. Compound I form III according to claim 6, wherein the diffractogram further comprises peaks (° 2 Θ ± 0.2 ° 2 Θ) at about 5.1, about 12.8, and about 18.0.

8. Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form IV) characterized by an X-ray powder diffraction pattern comprising the following peaks (° 2 Θ ± 0.2 ° 2 Θ): about 5.4, about 22.4 and about 23.3, using a wavelength ofIs measured on a diffractometer.

9. Compound I form IV according to claim 8, wherein the diffractogram further comprises peaks (° 2 Θ ± 0.2 ° 2 Θ) at about 10.8, about 18.9, about 23.9, and about 26.8.

10. Compound I form IV according to claim 8, characterized by a Differential Scanning Calorimetry (DSC) curve that comprises an endotherm at about 48 ℃ and an endotherm at about 59 ℃.

11. Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form V) characterized by an X-ray powder diffraction pattern comprising the following peaks (° 2 Θ ± 0.2 ° 2 Θ): about 4.6, about 9.2 and about 23.1, using a wavelength ofIs measured on a diffractometer.

12. Compound I form V according to claim 11, wherein the diffractogram further comprises peaks (° 2 Θ ± 0.2 ° 2 Θ) at about 13.8 and about 18.6.

13. Compound I form V according to claim 11, characterized by a Differential Scanning Calorimetry (DSC) curve that comprises an endotherm at about 70 ℃.

14. A pharmaceutical composition comprising one or more crystalline forms of any one of the preceding claims and one or more pharmaceutically acceptable carriers.

15. A method of promoting spine formation in a patient comprising administering a therapeutically effective amount of a compound I form according to any one of claims 1-13 or a pharmaceutical composition according to claim 14.

16. The method of claim 15, wherein the patient has a neuronal disorder.

17. The method of claim 16, wherein the neuronal disorder is selected from the group consisting of alzheimer's disease, parkinson's dementia, autism, fragile X syndrome, depression, and traumatic brain injury.

18. The method of claim 17, wherein the neuronal disorder is alzheimer's disease.

19. A process for preparing compound I or a pharmaceutically acceptable salt thereof:

comprising contacting compound a with compound B under first reaction conditions comprising a halide to form compound I:

20. the method of claim 19, wherein the halide is an alkali metal halide.

21. The method of claim 20, wherein the alkali metal halide is an alkali metal iodide.

22. The method of claim 21, wherein the alkali metal iodide is potassium iodide.

23. The method of claim 19, wherein the first reaction conditions further comprise an inorganic base.

24. The process of claim 23, wherein the inorganic base is potassium carbonate.

25. The method of claim 19, wherein the first reaction conditions comprise a temperature of 65 to 120 ℃.

26. The method of claim 19, further comprising contacting compound C with compound D under second reaction conditions comprising a protic acid to form compound a:

27. the method of claim 26, wherein the protic acid is an organic acid.

28. The method of claim 27, wherein the organic acid is acetic acid.

29. The method of claim 26, wherein the protic acid is present at a molar ratio relative to compound C and compound D that is greater than a stoichiometric amount.

30. The method of claim 29, wherein the protonic acid dissolves compound C and compound D.

31. The method of claim 26, further comprising contacting compound E with p-toluenesulfonyl chloride under third reaction conditions comprising a silver salt to form compound B:

32. the method of claim 31, wherein the silver salt is Ag2O。

33. The method of claim 31, wherein the third reaction conditions further comprise an alkali metal iodide.

Technical Field

The present disclosure relates generally to crystalline forms of the compound 2- (2- (2- (2- (4- (benzo [ d ] thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol, referred to herein as compound I, pharmaceutical compositions thereof, their therapeutic use and methods of preparing these forms.

Background

Neurological disorders are diseases of the brain, spinal cord and peripheral nervous system. In epidemiology and individual morbidity, the greatest social costs are caused by neurodegenerative conditions that result in the damage or loss of neurons and synaptic connections between them. The most prominent of these neurological diseases are Alzheimer's disease and Parkinson's disease. Other neurodegenerative conditions include age-related conditions (e.g., parkinson's dementia, vascular dementia, amyotrophic lateral sclerosis), genetic syndromes (e.g., down syndrome), injury-related conditions (e.g., traumatic brain injury, chronic traumatic encephalopathy), and conditions typically considered purely psychiatric in nature, such as schizophrenia and depression.

Researchers have classified hundreds of neurological diseases such as brain tumors, epilepsy, alzheimer's disease, parkinson's disease, and stroke, as well as conditions associated with aging such as dementia. Some of these conditions are caused by the progressive loss of synapses (junction between two different neurons) and eventually the loss of neurons (neurodegeneration). Unfortunately, neurodegenerative diseases are almost completely resistant to treatment. Neurons in the brain communicate with each other by releasing chemicals (neurotransmitters) to synaptic sending neurons, thereby changing the potential of the receiving neuron. The neurotransmitter-releasing part of the neuron is the axon (presynaptic side of the synapse), and the neurotransmitter-affected part of the synapse is called the dendritic spine (postsynaptic side of the synapse). The number, location, and even the change in shape of synaptic junctions is the basis for memory, learning, thinking, and personality. The part of the brain called the hippocampus is closely involved in the formation of memory and suffers from significant loss of synapses and neurons in neurodegenerative diseases. The development of novel methods to restore density of spines in hippocampus may be of great significance for the treatment of many neurodegenerative and developmental cognitive disorders.

Thus, small molecules that promote spine formation have potential utility in ameliorating the cognitive deficits of neuronal diseases, including neurodegenerative diseases such as alzheimer's disease. However, there is a need for highly pure single polymorph forms of compounds that are effective and exhibit improved pharmacokinetic and/or pharmacodynamic characteristics for the treatment of diseases that respond to the promotion of echinogenesis, including neuronal diseases.

Disclosure of Invention

Compound I exhibits activity in promoting echinogenesis and is described, for example, in international publication No. WO2019/028164(2 months and 7 days 2019), which is incorporated herein by reference in its entirety. Compound I has the formula:

compound I can be synthesized according to the methods described herein and the methods described in international publication No. WO2019/028164 (2/7/2019).

The present disclosure provides forms of compound I and salts, co-crystals, hydrates, and solvates thereof. Also described herein are methods of making the forms of compound I, pharmaceutical compositions comprising crystalline forms of compound I, and methods of using such forms and pharmaceutical compositions for treating diseases responsive to echinogenesis. In some embodiments, the disease is a neuronal disease.

Thus, one embodiment is the crystallization of 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form I) characterized by an X-ray powder diffraction pattern comprising the following peaks: about 4.6, about 20.8 and about 23.7 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer.

Another exampleThe embodiment is crystallization of 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I, form II), characterized by an X-ray powder diffraction pattern comprising the following peaks: about 9.2, about 13.8 and about 16.1 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer.

Another embodiment is the crystallization of 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form III) characterized by an X-ray powder diffraction pattern comprising the following peaks: about 7.7, about 10.3 and about 15.3 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer.

Another embodiment is the crystallization of 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form IV) characterized by an X-ray powder diffraction pattern comprising the following peaks: about 5.4, about 22.4 and about 23.3 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer.

Another embodiment is the crystallization of 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form V), characterized by an X-ray powder diffraction pattern comprising the following peaks: about 4.6, about 9.2 and about 23.1 degrees 2 theta + -0.2 degrees 2 theta, e.g. using a wavelength ofIs measured on a diffractometer.

Another embodiment is amorphous 2- (2- (2- (2- (4- (benzo [ d ] thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol.

Some embodiments provided herein relate to a crystalline form or co-crystal of a hydrate of compound I.

Another embodiment relates to a pharmaceutical composition comprising one or more forms of compound I as described herein and one or more pharmaceutically acceptable carriers.

Another embodiment relates to the use of a form of compound I described herein or a pharmaceutical composition described herein for promoting acanthosis in a patient in need thereof.

Another embodiment relates to a method of treating a disease responsive to acanthosis in a patient in need thereof comprising administering a therapeutically effective amount of a compound I form as described herein or a pharmaceutical composition as described herein. In some embodiments, the disease is a neuronal disease. In some embodiments, the disease is selected from alzheimer's disease, parkinson's dementia, autism, fragile X syndrome, and traumatic brain injury.

Another embodiment relates to a method of treating alzheimer's disease in a subject in need thereof comprising administering to the subject a therapeutically effective amount of a compound I form as described herein or a pharmaceutical composition as described herein.

Another embodiment relates to the use of a form of compound I as described herein or a pharmaceutical composition as described herein for the treatment of a disease selected from alzheimer's disease, parkinson dementia, autism, fragile X syndrome and traumatic brain injury. The disease may be alzheimer's disease.

Another embodiment relates to the use of a form of compound I as described herein or a pharmaceutical composition as described herein for the manufacture of a medicament for the treatment of a disease selected from alzheimer's disease, parkinson's dementia, autism, fragile X syndrome and traumatic brain injury. The disease may be alzheimer's disease.

In some embodiments, there is provided a method of preparing compound I, or a pharmaceutically acceptable salt thereof:

comprising contacting compound a with compound B under first reaction conditions comprising a halide to form compound I:

in some embodiments, the method of making compound I further comprises contacting compound C with compound D under second reaction conditions comprising a protic acid to form compound a:

in some embodiments, the method of making compound I further comprises contacting compound E with p-toluenesulfonyl chloride under third reaction conditions comprising a silver salt to form compound B:

drawings

Figure 1 shows the X-ray powder diffraction (XRPD) of compound I form I.

Figure 2 shows a Differential Scanning Calorimeter (DSC) curve and thermogravimetric analysis (TGA) of compound I form I.

Figure 3 shows the X-ray powder diffraction (XRPD) of compound I form II.

Figure 4 shows the x-ray powder diffraction (XRPD) of compound I form III.

Figure 5 shows the X-ray powder diffraction (XRPD) of compound I form IV.

Figure 6 shows a Differential Scanning Calorimeter (DSC) curve and thermogravimetric analysis (TGA) of compound I form IV.

Figure 7 shows the X-ray powder diffraction (XRPD) of compound I form V.

Figure 8 shows a Differential Scanning Calorimeter (DSC) curve and thermogravimetric analysis (TGA) of compound I form V.

Figure 9 shows the interconversion of the crystalline form of compound I obtained during the polymorphic screening process.

Figure 10 shows the XRPD pattern of compound I starting material.

Figure 11 shows the TGA/DSC curve of the compound I starting material.

Figure 12 shows a DVS plot of the compound I starting material.

Figure 13 shows XRPD stacking of compound I starting materials before and after DVS testing.

Figure 14 shows XRPD overlay of compound I after storage at 30 ℃/65% RH/open.

Figure 15 shows XRPD overlay of compound I after storage at 30 ℃/65% RH/block.

Figure 16 shows XRPD overlay of compound I after storage at 40 ℃/75% RH/open.

Figure 17 shows the XRPD overlay of compound I after storage at 40 ℃/75% RH/block.

Detailed Description

The compound 2- (2- (2- (2- (4- (benzo [ d ] thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol, designated herein as compound I, has the formula:

the present disclosure relates to various crystalline forms of compound I, and methods of making these crystalline forms. Also described herein are compounds I in the forms further labeled herein as "compound I form I", "compound I form II", "compound I form III", "compound I form IV", "compound I form V" and "amorphous compound I". In some embodiments, these forms of compound I may be anhydrous. In some embodiments, these forms of compound I may be solvates or hydrates.

Other crystalline forms of compound I are also described further herein. In some embodiments, the crystalline form of compound I may comprise a salt or co-crystal of compound I.

Definition of

The following words and phrases, as used in this specification, are generally intended to have the meanings as set forth below, except where otherwise indicated in the context in which they are used.

The term "fascin" refers to a 54-58kDa protein that is an actin-crosslinking protein. The term "fascin" can refer to the amino acid sequence of human fascin 1. The term "fascin" encompasses the wild-type form of the nucleotide sequence or protein as well as any mutants thereof. In some embodiments, the "fascin" is a wild-type fascin. In some embodiments, the "fascin" is one or more mutant forms. In some embodiments, the fascin is human fascin 1. In some embodiments, fascin is encoded in a nucleic acid sequence that is identical to reference number GI: 347360903 in the corresponding nucleotide sequence. In some embodiments, fascin is encoded in the nucleotide sequence of RefSeq M _ 003088. In some embodiments, the fascin corresponds to the amino acid sequence of RefSeq NP _ 003079.1.

The terms "spine formation" and the like refer in a general and customary sense to the development (e.g., growth and/or maturation) of the dendritic spines of a neuron. In some embodiments, the compounds provided herein promote spine formation without affecting spine morphology. Promotion is relative to the absence of administration of the compound.

As used herein, the term "dendrite" refers to a branched extension of a neuronal cell. Dendrites are generally responsible for receiving electrochemical signals transmitted from axons of neighboring neurons. The term "dendritic spine" or "dendritic spine" refers to a protoplasmic process on a neuronal cell (e.g., a neuronal cell on a dendrite). In some embodiments, the dendritic spines can be described as having membranous necks, which can be terminated by small heads (e.g., heads). Dendritic spines are classified according to their shape: headless, thin, short, mushroom, or dendritic. Dendritic spine density refers to the total number of dendritic spines per unit length of neuronal cells. For example, the dendritic spine density may be given as the number of dendritic spines per micron.

The term "dendritic spine formation" and the like refer in a general and customary sense to a process that results in an increase in the number of dendritic spines or an increase in the development of dendritic spines. The terms "dendritic spine morphology" and the like refer in a general and customary sense to the physical characteristics (e.g., shape and structure) of the dendritic spine. An improvement in dendritic spine morphology is a change in morphology (e.g., an increase in length or an increase in width) that results in increased functionality (e.g., an increase in the number of contacts between neurons or a decrease in the space between adjacent neurons (e.g., synaptic clefts)). As known in the art and disclosed herein, an exemplary method for this characterization involves measuring the dimensions (i.e., length and width) of the dendritic spines. Thus, the term "improving the morphology of the dendritic spines" generally refers to increasing the length, width, or both the length and width of the dendritic spines.

"binding" means that at least two different species (e.g., chemical compounds comprising biomolecules or cells) become sufficiently close to react or interact, resulting in the formation of a complex. For example, the association of two different species (e.g., proteins and compounds described herein) may result in the formation of a complex, wherein the species interact through non-covalent or covalent bonds. In some embodiments, the resulting complex is formed when two different species (e.g., proteins and compounds described herein) interact through non-covalent bonds (e.g., electrostatic, van der Waals, or hydrophobic).

As defined herein, the terms "activation", and the like with respect to a protein-activator (e.g., agonist) interaction, mean that the activity or function of a protein is positively affected (e.g., increased) relative to the activity or function of the protein in the absence of an activator (e.g., a compound described herein).

"control" or "control experiment" is used in accordance with its ordinary general meaning and refers to an experiment in which the subject or agent of the experiment is treated as in a parallel experiment, except that the procedures, agents or variables of the experiment are omitted. In some examples, controls are used as a standard of comparison in evaluating the effect of an experiment.

"contacting" is used according to its ordinary meaning and refers to the process of bringing at least two different species (e.g., chemical compounds comprising biomolecules, or cells) into sufficient proximity to interact. The term "contacting" may include allowing two species to react or physically touch, wherein the two species may be a compound, biomolecule, protein, or enzyme as described herein. In some embodiments, contacting comprises allowing a compound described herein to interact with a protein (e.g., fascin) or enzyme.

As defined herein, the terms "inhibit", "inhibiting", and the like will be given their customary meaning to those skilled in the art. With respect to protein-inhibitor (e.g., antagonist) interactions, the terms "inhibit", "inhibiting" and "inhibiting" mean negatively affecting (e.g., decreasing) the functional activity of a protein relative to the functional activity of the protein in the absence of the inhibitor.

As used herein, the term "about" as used in the context of quantitative measurements means the indicated amount ± 10%, or alternatively, the indicated amount ± 5% or ± 1%.

The term "complex" refers to a formation resulting from an interaction between compound I and another molecule.

The term "solvate" refers to a complex formed by combining compound I with a solvent. As used herein, the term "solvate" includes hydrates (i.e., solvates when the solvent is water).

The term "co-crystal" refers to a molecular complex of ionized or non-ionized compound I (or any other form, salt, or compound disclosed herein) and one or more non-ionized co-crystal formers (e.g., pharmaceutically acceptable salts) linked by non-covalent interactions. In certain embodiments, the co-crystal may have improved properties compared to the free form (i.e., free molecule, zwitterion, hydrate, solvate, etc.) or the salt (which includes salt hydrates and solvates). In a further embodiment, the improved property is selected from the group consisting of: increased solubility, increased dissolution, increased bioavailability, increased dose response, reduced hygroscopicity, crystalline forms of generally amorphous compounds, crystalline forms of compounds that are difficult or impossible to salt, reduced form diversity, more desirable morphology, and the like.

The term "co-crystal former" or "co-former" refers to one or more of the pharmaceutically acceptable bases and/or pharmaceutically acceptable acids disclosed herein in association with compound I or any other compound disclosed herein.

Any formulae or structures given herein, including compound I, are also intended to represent unlabeled as well as isotopically labeled forms of these compounds. Isotopically-labeled compounds have the structure depicted by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the present disclosure include: isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as, but not limited to:2h (deuterium, D),3H (tritium),11C、13C、14C、15N、18F、31P、32P、35S、36Cl, and125I. various isotopically-labeled compounds of the present disclosure can be prepared, for example, with incorporation of an isotope therein (e.g.3H、13C and14C) the compound of (1). Such isotopically labeled compounds are useful in metabolic studies, reaction kinetic studies, detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution assays, or in the radiation treatment of patients.

The disclosure also includes compounds I wherein 1 to "n" hydrogens attached to a carbon atom are replaced with deuterium, where n is the number of hydrogens in the molecule. Such compounds exhibit increased metabolic resistance and are therefore useful for increasing the half-life of any compound I when administered to a mammal. See, e.g., Foster, "role of Deuterium isotopes in Drug Metabolism Studies" (Deuterium Isotrope Effects in Studies of Drug Metabolism), "Trends in pharmacology (Trends Pharmacol. Sci.5(12):524-527 (1984). Compounds of this type are synthesized using methods well known in the art, e.g., by using a starting material in which one or more hydrogen atoms have been replaced by Deuterium.

Deuterium labeled or substituted therapeutic compounds of the present disclosure may have improved DMPK (drug metabolism and pharmacokinetics) properties, which are involved in distribution, metabolism and excretion (ADME). Isotopically labeled compounds of the present disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes below or in the examples and preparations below, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent. Further, with heavier isotopes, especially deuterium (i.e. deuterium)2Substitution by H or D) may provide certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half-life or reduced dosage requirements or an increased therapeutic index. Isotopic labels for diagnostic purposes are also contemplated.

The concentration of such isotopes, in particular deuterium, can be defined by the isotopic enrichment factor. In the compounds of the present disclosure, any atom not specifically designated as a particular isotope is intended to mean any isotope of that atom. Unless otherwise indicated, when a position is specifically designated as "H" or "hydrogen," the position is understood to contain hydrogen in its natural abundance isotopic composition. Thus, in the compounds of the present disclosure, any atom specifically designated as deuterium (D) is intended to represent deuterium that is greater than natural abundance.

As used herein, "pharmaceutically acceptable carrier" includes excipients or agents that are not deleterious to the compounds of the invention or the use thereof, such as solvents, diluents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. The use of such carriers and agents for the preparation of compositions of pharmaceutically active substances is well known in the art (see, e.g., Remington's Pharmaceutical Sciences, machine Publishing Co., Philadelphia, Pa., 17 th edition (1985); and Modern pharmaceuticals (Modern pharmaceuticals), Marcel Dekker, 3 rd edition, (G.S. Bank & C.T. Rhodes, Eds.).

The term "therapeutically effective amount" refers to an amount of a compound as described herein that is sufficient to effect a treatment as defined below when administered in one or more doses to a patient (particularly a human) in need of such treatment. The therapeutically effective amount will vary depending on the patient, the disease being treated, the weight and/or age of the patient, the severity of the disease, or the mode of administration determined by a qualified prescriber or caregiver.

The term "treatment" or "treating" refers to the administration of a compound described herein for the following purposes:

(i) delay the onset of the disease, i.e., do not develop or delay the development of clinical symptoms or markers of the disease;

(ii) inhibiting disease, i.e., slowing or arresting the development of clinical symptoms or markers thereof, or the spread of disease;

and/or

(iii) Remission of the disease, i.e., causing regression of clinical symptoms or severity markers.

"prevention" or "preventing" refers to the treatment of any disease or disorder that results in the failure to develop clinical symptoms of the disease or disorder. In some embodiments, the compound may be administered to a subject (including a human) at risk of or having a family history of a disease or condition.

"subject" refers to an animal, such as a mammal (including a human), that has been or will become the subject of treatment, observation or experiment. The methods described herein can be used for human therapy and/or veterinary applications. In some embodiments, the subject is a mammal. In one embodiment, the subject is a human. When the subject is a human, the subject may be referred to as a "patient".

The methods described herein may be applied to a population of cells in vivo or ex vivo. By "in vivo" is meant within a living individual, such as within an animal or human. In this context, the methods described herein may be used therapeutically in an individual. By "ex vivo" is meant outside a living subject. Examples of ex vivo cell populations include in vitro cell cultures and biological samples, including liquid or tissue samples obtained from individuals. Such samples may be obtained by methods well known in the art. Exemplary biological fluid samples include blood, cerebrospinal fluid, urine, and saliva. In this context, the compounds and compositions described herein can be used for a variety of purposes, including therapeutic and experimental purposes. For example, the compounds and compositions described herein can be used ex vivo for a given indication, cell type, individual, and other parameters to determine the optimal schedule and/or dosage for administration of the compounds of the present disclosure. The information gathered from this use can be used for experimental purposes or in the clinic to set up in vivo treatment protocols. Other ex vivo uses to which the compounds and compositions described herein may be suitable are described below or will become apparent to those of skill in the art. The selected compounds may be further characterized by examining a safe or tolerated dose in a human or non-human subject. Such properties can be checked using methods generally known to those skilled in the art.

Further, abbreviations as used herein have the following corresponding meanings:

forms of compound I

As generally described above, the present disclosure provides crystalline forms of compound I and co-crystals thereof. Other forms are discussed further herein.

Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form I) is characterized by an X-ray powder diffraction pattern comprising the following peaks: about 4.6, about 20.8 and about 23.7 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer. The diffraction pattern includes additional peaks at about 9.2, about 16.3, about 18.6, and about 19.6 degrees 2 theta ± 0.2 degrees 2 theta. Compound I form I is also characterized by its full X-ray powder diffraction pattern substantially as shown in figure 1. Compound I form I may be characterized by one or more, e.g., 1, 2, 3, 4, 5, or 6, of the following XRPD peaks:

peak(s)
4.6
9.2
18.6
19.6
20.8
23.7

In some embodiments, compound I form I is characterized by a Differential Scanning Calorimetry (DSC) curve that comprises an endothermic peak at about 70 ℃ ± 2 ℃. Compound I form I is also characterized by its complete DSC curve substantially as shown in figure 2.

Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form II) is characterized by an X-ray powder diffraction pattern comprising the following peaks: about 9.2, about 13.8 and about 16.1 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer. The diffraction pattern includes additional peaks at about 6.9, about 11.5, and about 18.4 ° 2 Θ ± 0.2 ° 2 Θ. Compound I form II is also characterized by its full X-ray powder diffraction pattern substantially as shown in figure 3. In some embodiments, compound I form II is present as a wet material. Compound I form II may be characterized by one or more, e.g., 1, 2, 3, 4, 5, or 6, of the following XRPD peaks:

peak(s)
6.9
9.2
11.5
13.8
16.1
18.4

Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form III) is characterized by an X-ray powder diffraction pattern comprising the following peaks: about 7.7, about 10.3 and about 15.3 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer. The diffraction pattern includes additional peaks at about 5.1, about 12.8, and about 18.0 ° 2 θ ± 0.2 ° 2 θ. Compound I form III is also characterized by its full X-ray powder diffraction pattern substantially as shown in figure 4. In some embodiments of the present invention, the substrate is,compound I form III is present as a wet material. Compound I form III may be characterized by one or more, e.g., 1, 2, 3, 4, 5, or 6, of the following XRPD peaks:

peak(s)
5.1
7.7
10.3
12.8
15.3
18.0

Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form IV) is characterized by an X-ray powder diffraction pattern comprising the following peaks: about 5.4, about 22.4 and about 23.3 degrees 2 theta + -0.2 degrees 2 theta, e.g., using a wavelength ofIs measured on a diffractometer. The diffraction pattern includes additional peaks at about 10.8, about 18.9, about 23.9, and about 26.8 ° 2 Θ ± 0.2 ° 2 Θ. Compound I form IV is also characterized by its full X-ray powder diffraction pattern substantially as shown in figure 5. Compound I form IV may be characterized by one or more, e.g., 1, 2, 3, 4, 5, or 6, of the following XRPD peaks:

peak(s)
5.4
10.8
18.9
22.4
23.3
23.9
26.8

In some embodiments, compound I form IV is characterized by a Differential Scanning Calorimetry (DSC) curve that comprises an endothermic peak at about 48 ℃ ± 2 ℃ and an endothermic peak at about 59 ℃ ± 2 ℃. Compound I form IV is also characterized by its complete DSC curve substantially as shown in figure 6.

Crystalline 2- (2- (2- (2- (4- (benzo [ d ]))]Thiazol-2-yl) phenoxy) ethoxy) ethan-1-ol (compound I form V) is characterized by an X-ray powder diffraction pattern comprising the following peaks: about 4.6, about 9.2 and about 23.1 degrees 2 theta + -0.2 degrees 2 theta, e.g. using a wavelength ofIs measured on a diffractometer. The diffraction pattern includes additional peaks at about 13.8 and about 18.6 ° 2 θ ± 0.2 ° 2 θ. Compound I form V is also characterized by its full X-ray powder diffraction pattern substantially as shown in figure 7. Compound I form IV may be characterized by one or more, e.g., 1, 2, 3, 4, 5, or 6, of the following XRPD peaks:

peak(s)
4.6
7.0
9.2
13.8
18.4
23.1

In some embodiments, compound I form V is characterized by a Differential Scanning Calorimetry (DSC) curve that comprises an endothermic peak at about 70 ℃ ± 2 ℃. Compound I form V is also characterized by its complete DSC curve substantially as shown in figure 8.

Some embodiments relate to compositions comprising a form of compound I as described herein without any other form of compound I. In some embodiments, the composition comprises greater than 95% of a form of compound I as described herein relative to other forms of compound I. In some embodiments, the compositions comprise greater than 97% of a form of compound I described herein relative to other forms of compound I. In some embodiments, the compositions comprise greater than 99% of a compound I form described herein, relative to other forms of compound I.

Some embodiments relate to other forms of compound I compositions comprising crystalline compound I form I as described herein. In some embodiments, the composition comprises greater than 95% crystalline compound I form I as described herein relative to other forms of compound I. In some embodiments, the composition comprises greater than 97% of crystalline compound I form I described herein relative to other forms of compound I. In some embodiments, the compositions comprise greater than 99% of crystalline compound I form I described herein, relative to other forms of compound I.

In some embodiments, the composition comprises crystalline compound I form I in greater than 95% purity. In some embodiments, the composition comprises crystalline compound I form I in a purity of greater than 97%. In some embodiments, the composition comprises crystalline compound I form I in a purity of greater than 99%.

Some embodiments relate to methods of preparing a form of compound I as described herein. In some embodiments, the method is as described in the examples provided herein.

In some embodiments, the method of preparing compound I form I is selected from solid vapor diffusion, anti-solvent addition, liquid vapor diffusion, slow cooling, slurry inversion, temperature cycling, and slow evaporation. The solid vapor diffusion can be performed at room temperature with a solvent selected from acetone, THF, EtOH, H2O, EtOAc, dioxane, toluene, DCM and acetonitrile. The anti-solvent addition may be carried out by adding an anti-solvent selected from IPA, MTBE, CPME and heptane to a solution of compound I in a solvent selected from THF, MeOH, DCM and EtOAc. Slow cooling may be carried out by cooling a solution of compound I in a solvent or solvent system selected from MEK, dioxane: toluene (optionally at a ratio of about 1:1 v/v), EtOAc: acetonitrile (optionally at a ratio of 1:1 v/v), DCM: IPA (optionally at a ratio of about 4:1 v/v) and THF: heptane (optionally at a ratio of about 4:1 v/v). Can be prepared by reacting compound I in a solvent selected from THF, EtOAc, MEK, acetonitrile and dioxane H2Slow evaporation is carried out by forming a solution in a solvent or solvent system of O (optionally at a ratio of about 9:1 v/v).

In some embodiments, compound I form I is stable at 30 ℃/65% RH (closed or open) and 40 ℃/75% RH (closed or open) for at least 4 weeks. In some embodiments, compound I form I retains at least 99% purity after storage for at least 4 weeks, optionally at 30 ℃/65% RH (closed or open) or 40 ℃/75% RH (closed or open).

Pharmaceutical compositions and dosages

The form of compound I as described herein may be administered in the form of a pharmaceutical composition. Thus, also provided herein are pharmaceutical compositions containing one or more of the forms of compound I described herein, together with one or more pharmaceutically acceptable vehicles selected from carriers, adjuvants and excipients. Suitable pharmaceutically acceptable vehicles may include, for example, inert solid diluents and fillers, diluents including sterile aqueous solutions and various organic solvents, penetration enhancers, solubilizing agents and adjuvants. Such compositions are prepared in a manner well known in the pharmaceutical art. See, for example, Remington's Pharmaceutical Sciences, meis Publishing company, Philadelphia, Pa, 17 th edition (1985); and Modern pharmaceuticals (Modern pharmaceuticals), Marcel Dekker, 3 rd edition (edited by g.s.banker and c.t.rhodes). The pharmaceutical composition may be administered alone or in combination with other therapeutic agents.

The pharmaceutical composition may be administered in a single dose or in multiple doses. The pharmaceutical compositions may be administered by a variety of methods, including, for example, rectal, buccal, intranasal, and transdermal routes. In certain embodiments, the pharmaceutical composition may be administered by intra-arterial injection, intravenously, intraperitoneally ("i.p."), parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.

One means for use is parenterally, for example by injection. The pharmaceutical compositions described herein may be incorporated into a form for administration by injection, including, for example, aqueous or oil suspensions or emulsions with sesame, corn, cottonseed, or peanut oil, as well as elixirs, mannitol, dextrose, or sterile aqueous solutions, and similar pharmaceutical vehicles.

Oral administration may be another route of administration of one or more of the compound I forms described herein. Administration can be by way of, for example, capsules or enteric-coated tablets. In preparing a pharmaceutical composition comprising one or more compounds described herein, the active ingredient is typically diluted by an excipient and/or enclosed within a carrier, which may be in the form of a capsule, sachet, paper or other container. When an adjuvant agent is used as a diluent, it may take the form of a solid, semi-solid, or liquid material that acts as an excipient, carrier, or medium for the active ingredient. Thus, the compositions may take the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10% by weight of the active ingredient, soft and hard gelatin capsules, sterile injectable solutions, and sterile packaged powders.

Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methyl cellulose. The formulation may additionally comprise: lubricants, such as talc, magnesium stearate and mineral oil; a wetting agent; emulsifying and suspending agents; preservatives, such as methyl and propyl hydroxybenzoate; a sweetener; and a flavoring agent.

Compositions comprising one or more of the compound I forms described herein can be formulated to provide rapid, sustained, or delayed release of the active ingredient upon administration to a subject by using procedures known in the art. Controlled release drug delivery systems for oral administration include osmotic pump systems and dissolution systems comprising polymer coated reservoirs or drug-polymer matrix formulations. Examples of controlled release systems are given in the following: U.S. patent nos. 3,845,770; U.S. Pat. No. 4,326,525; 4,902,514 No; and No. 5,616,345. Another formulation for use in the methods disclosed herein employs a transdermal delivery device ("patch"). Such transdermal patches may be used to provide continuous or intermittent infusion of the compound I form described herein in controlled amounts. The construction and use of transdermal patches for delivering pharmaceutical agents is well known in the art. See, for example, U.S. Pat. nos. 5,023,252, 4,992,445, and 5,001,139. Such patches may be configured for continuous, pulsed, or on-demand delivery of the agent.

To prepare a solid composition, such as a tablet, the primary active ingredient may be mixed with a pharmaceutical excipient to form a solid pre-formulated composition containing a homogeneous mixture of the forms of compound I described herein. Where these preformulation compositions are mentioned as being homogeneous, the active ingredient may be dispersed uniformly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules.

Tablets or pills of compound I as described herein may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action or to protect the stomach from acidic conditions. For example, a tablet or pill may comprise an inner dosage component and an outer dosage component, the latter being in the form of a film over the former. The two components may be separated by an enteric layer that serves to resist disintegration in the stomach and allows the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials may be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate.

Compositions for inhalation or insufflation may comprise solutions and suspensions or mixtures thereof in pharmaceutically acceptable aqueous or organic solvents, as well as powders. The liquid or solid composition may contain suitable pharmaceutically acceptable excipients as described above. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic action. In other embodiments, the composition in a pharmaceutically acceptable solvent may be atomized by using an inert gas. The nebulized solution may be inhaled directly from the nebulizing device, or the nebulizing device may be attached to a mask tent or an intermittent positive pressure ventilator. The solution, suspension or powder composition may be administered, preferably orally or nasally, from a device that delivers the formulation in an appropriate manner.

The form of compound I as described herein may be administered in a pharmaceutically effective amount. For oral administration, each dosage unit may contain 1mg to 2g, 1mg to 1g, or 1mg to 500mg of compound I. In some embodiments, the dose is 1mg to 250mg of compound I. In some embodiments, the dose of compound I ranges from about 20mg twice daily to about 50mg twice daily. In some embodiments, the dose is 2mg, 4mg, 6mg, 8mg, 10mg, 12mg, 14mg, 16mg, 18mg, 20mg, 25mg, 30mg, 35mg, 40mg, 45mg, 50mg, 75mg, 100mg, 200mg, or 500mg of compound I. It will be understood, however, that the amount of the compound actually administered will generally be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration and co-administration of the compound, and, if applicable, the age, weight, response, severity of the patient's symptoms, and the like.

The form of compound I of the present application or a composition thereof may be administered once, twice, three times or four times daily using any suitable mode described above. In addition, the form of compound I of the present application or a composition thereof may be administered once or twice weekly, once every two weeks, once every three weeks, once every four weeks, once every five weeks, or once every six weeks. In some embodiments, a form of compound I of the present application or a composition thereof may be administered once daily for 4 weeks, 8 weeks, 12 weeks, 16 weeks, 20 weeks, 24 weeks, 28 weeks, 32 weeks, 36 weeks, 40 weeks, 44 weeks, 48 weeks, 52 weeks, or longer as needed.

The specific dosage level of the active agent of the present application, e.g., a form of compound I described herein, or a pharmaceutical composition thereof, for any particular subject will depend upon a variety of factors including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, route of administration and rate of secretion, drug combination, and the severity of the particular disease in the subject undergoing therapy. For example, a dose can be expressed as milligrams of a compound described herein per kilogram of the subject's body weight (mg/kg). Dosages between about 0.1 and 150mg/kg may be appropriate. In some embodiments, about 0.1 and 100mg/kg may be suitable. In other embodiments, a dose between 0.5mg/kg and 60mg/kg may be appropriate. Normalization according to the weight of the subject can be particularly useful when adjusting the dose between subjects of widely different sizes, such as occurs when using drugs in both children and adult humans or when converting an effective dose in a non-human subject (such as a dog) to a dose suitable for a human subject.

A daily dose may also be described as the total amount of a compound described herein administered per dose or per day. A daily dose of a compound or salt thereof described herein may be between about 1mg and 4,000mg, between about 2,000 mg to 4,000 mg/day, between about 1mg to 2,000 mg/day, between about 1mg to 1,000 mg/day, between about 10mg to 500 mg/day, between about 20mg to 500 mg/day, between about 50mg to 300 mg/day, between about 75mg to 200 mg/day, or between about 15mg to 150 mg/day.

When administered nasally, the total daily dose for a human subject can be between 1mg and 1,000mg, between about 1,000-2,000 mg/day, between about 10-500 mg/day, between about 50-300 mg/day, between about 75-200 mg/day, or between about 100-150 mg/day. In various embodiments, the daily dose is about 10mg, about 30mg, about 50mg, about 75mg, about 100mg, about 200mg, about 300mg, about 400mg, about 500mg, about 600mg, about 700mg, about 800mg, about 900mg, or about 1000mg or some range of values therebetween.

The form of compound I described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, may be administered once, twice, three times or four times daily using any suitable means described above. Likewise, administration or treatment may continue for several days; for example, for one cycle of treatment, typically treatment will continue for at least 7, 14 or 28 days. Treatment cycles are well known and often alternate between cycles or for the remainder of about 1 to 28 days, usually about 7 or about 14 days. In other embodiments, the treatment cycle may also be continuous. Administration or treatment may continue indefinitely.

In particular embodiments, the methods comprise administering to the subject an initial daily dose of about 1mg to 800mg of a form of a compound described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, and incrementally increasing the dose until clinical efficacy is achieved. Increments of about 5mg, 10mg, 25mg, 50mg or 100mg may be used to increase the dosage. The dose may be increased daily, every other day, twice weekly, or once weekly.

The form of compound I described herein or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof may be administered under fed conditions. The term "eating conditions" or variants thereof refers to the consumption or ingestion of food or any suitable form of calories in solid or liquid form prior to or simultaneously with the administration of the active ingredient. For example, a form of compound I described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, can be administered to a subject (e.g., a patient) within minutes or hours of consuming calories (e.g., having a meal). In some embodiments, a form of compound I described herein, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, can be administered to a subject (e.g., a patient) within 5-10 minutes, about 30 minutes, or about 60 minutes of consuming calories.

Methods of treatment and uses

Fascin

Described herein are methods of regenerating synapses lost to neurodegenerative diseases by targeting cytoskeletal proteins by administering to a subject a form of compound I described herein. Unexpectedly, it was observed that inhibition of the cytoskeletal protein fascin 1(FSCN1) resulted in rapid upregulation of dendritic spines in vivo and in vitro. Dendritic spines contain filamentous actin (F-actin), a cytoskeletal polymer that confers cellular structure and confers subcellular specialization to them. Without wishing to be bound by theory, it is believed that dendritic spines require the formation of highly branched assemblies of F-actin, and the formation of such assemblies can be prevented or significantly reduced by virtue of bundling into parallel arrays by fascin 1.

In some embodiments, there is provided a method of binding fascin, comprising contacting fascin with an effective amount of compound I, or a pharmaceutically acceptable salt thereof, as described herein. In some embodiments, the method inhibits fascin. It is believed that the form of compound I or a pharmaceutically acceptable salt thereof can promote dendritic spines formation by inhibiting fascin.

Fascin is an important actin cross-linker that has no amino acid sequence homology to other actin-binding proteins. Three forms of fascin are found in vertebrates: fascin 1, which is widely found in the nervous system and elsewhere; fascin 2, which is found in retinal photoreceptor cells; and fascin 3, which is found only in the testis. In some embodiments, the fascin is human fascin 1. Fascin has a molecular weight of 55kDa and acts as a monomeric entity and cross-links actin filaments into straight, tight and rigid bundles, giving mechanical rigidity to the actin bundles. Fascin is believed to hold parallel actin filaments together to form filopodia approximately 60-200nm in diameter.

During neuronal development, it is believed that long tracts of f-actin push the membranes of neurons out to form structures such as axons, dendrites, filopodia, and lamellipodia. Fascin is thought to be involved in cytoskeletal reorganization of nascent dendritic processes. Therefore, fascin-bundled actin is generally considered required for the formation and extension of axons and dendrites. It has been surprisingly found that inhibiting fascin activity in actin bundle formation promotes dendritic spine formation, and dendritic cell plasma membrane protrusion.

Neuronal diseases and conditions and treatments thereof

A unifying feature of neurodegenerative conditions with cognitive components is the loss of synapses that utilize glutamate amino acids as neurotransmitters ("glutamatergic synapses"), which are believed to be the most types of synapses in humans and other mammals. Importantly, about 90% of glutamatergic synapses involve postsynaptic dendritic spines. The majority of synapses lost in neurodegenerative diseases are synapses where axons contact dendritic spines, so-called "axonal synapses". Normally, changes in the density, shape and protein composition of dendritic spines affect the strength of synaptic transmission and are the basis for several forms of synaptic changes (i.e., "plasticity") involved in learning and memory, cognitive flexibility, adaptability to injury and disease, and other processes. These changes in axonal synapses are believed to be important for memory-encoded function of structures such as the hippocampus. Thus, early and progressive loss of dendritic spines in the hippocampus and other regions is believed to be the driving force for memory loss and cognitive decline in alzheimer's disease and other dementias. The development of novel methods to regenerate spine density may be of great significance for the treatment of many neurodegenerative and developmental cognitive disorders.

Dendritic spines are specialized processes that are responsible for receiving synaptic inputs, providing important functions in communication between neurons. The morphology of dendritic spines and their overall density are related to synaptic function and are strongly involved in memory and learning. Cellular changes in brain cells may contribute to the pathogenesis of neuronal diseases. For example, abnormal levels (e.g., decreased) of dendritic spine density in the brain may contribute to the pathogenesis of neuronal diseases. Thus, it is believed that alterations or dysregulation of dendritic spines affect synaptic function and play an important role in a variety of neurological and psychiatric disorders, such as autism, fragile-X syndrome, Parkinson's Disease (PD) and Alzheimer's Disease (AD). For example, in AD, there is increasing evidence that defects begin with changes in hippocampal synaptic function caused by amyloid beta (a β) protein prior to neuronal loss. Thus, therapeutic strategies targeting initial synaptic loss rather than late-stage disease intervention may provide a better prognosis for the treatment of AD. Furthermore, since most cognitive disorders cause abnormalities in the form and function of dendritic spines, it would be desirable to target them directly using small molecules to alter or mitigate these spine changes. For example, fragile X syndrome is characterized by immature hyperrachidian.

Provided herein are methods useful for promoting dendritic spine production. In some embodiments, the method comprises administering to the subject an effective amount of a form of compound I described herein or a pharmaceutical composition thereof, as described herein, including embodiments. Dendritic spine formation can be observed as an increase in the average number of spines per neuron or in the length of a neuron unit, which may be referred to as an increase in dendritic spine density. Can watchDendritic spine formation was observed as an improvement in dendritic spine morphology. For example, an improvement in dendritic spine morphology can be observed as an increase in the average size of the spine heads. Dendritic spine formation can be observed as an improvement in dendritic spine size, spine plasticity, spine motility, spine density, and/or synaptic function. Dendritic spine formation can be observed as a local spatially averaged increase in membrane potential. It was observed that dendritic spine formation was Ca2+Increase in postsynaptic concentration (e.g., average volume) of (a). Dendritic spine formation can be observed as an increase in the average proportion of mature to immature spines. In some embodiments, a form of compound I described herein or a pharmaceutical composition thereof increases dendritic spine density relative to a control. In some embodiments, a form of compound I or a pharmaceutical composition thereof described herein increases dendritic spine density compared to that observed at the start of treatment. In some embodiments, the increase in density of the dendritic spines results in a reduction in symptoms of the neuronal disease or disorder in the subject or patient. In some embodiments, the increase in density of the dendritic spines is explained by anatomical observation. In some embodiments, an increase in dendritic spine density is observed in primary hippocampal neurons.

In some embodiments, the average dendritic spine density is increased by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 100%, 125%, 150%, 175%, 200%, 250%, 300%, 400%, 500%, 750%, or 1000% or any range between any two of the numbers, inclusive, relative to the time of starting treatment with a form of compound I or a pharmaceutical composition thereof described herein. In some embodiments, the dendritic spine density is increased by about 50% to about 500% relative to the time of treatment initiation with a form of compound I or a pharmaceutical composition thereof described herein. In some embodiments, the dendritic spine density is increased by about 100% to about 300% relative to the time of treatment initiation with a form of compound I or a pharmaceutical composition thereof described herein. In some embodiments, the dendritic spine density is increased by about 200% to about 300% relative to the time of treatment initiation with a form of compound I or a pharmaceutical composition thereof described herein. In some embodiments, the duration of treatment with a form of compound I or a pharmaceutical composition thereof described herein is 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, 1 day, 3 days, 5 days, 7 days, 14 days, 28 days, 90 days, 180 days, or 365 days.

In some embodiments, the method increases the density of the spines by promoting the formation of new spines. In some embodiments, the method increases the average spine density by at least about 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 60%, 70%, 80%, 90%, 100%, 125%, 150%, 175%, 200%, 250%, 300%, 400%, 500%, 750%, or 1000% or any range between any two of the numbers, inclusive, relative to a control (e.g., spine density in the absence of compound). In some embodiments, the method increases the average spine density by about 50% to about 500% relative to a control (e.g., spine density in the absence of compound). In some embodiments, the method increases the spine density by about 100% to about 300% relative to a control (e.g., spine density in the absence of compound). In some embodiments, the method increases the spine density by about 200% to about 300% relative to a control (e.g., spine density in the absence of compound).

In some embodiments, the method increases spine density by increasing neuron length. In some embodiments, the method increases the mean neuron length by about 100nm, 300nm, 500nm, 700nm, 1 micron, 2 microns, 3 microns, 4 microns, 5 microns, 7 microns, 10 microns, 15 microns, 20 microns, 25 microns, or any range between any two of the numbers, inclusive, relative to the time of initiation of treatment with a form of compound I or a pharmaceutical composition thereof described herein. In some embodiments, the method increases the average neuron length by about 500nm to about 25 microns relative to a control (e.g., the neuron length in the absence of the compound). In some embodiments, the method increases the neuron length by about 10% to about 300% relative to a control (e.g., the neuron length in the absence of the compound). In some embodiments, the method increases the neuron length by about 200% to about 300% relative to a control (e.g., the neuron length in the absence of the compound).

In some embodiments, the method increases the average number of spikes per neuron relative to the time at which treatment with a form of compound I or a pharmaceutical composition thereof described herein is initiated. In some embodiments, the average number of spikes per unit length of the neuron is increased by at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 200, 300, 400, 500, 600, 700, 800, 900, or more than about 1000 or any range between any two of the numbers, inclusive. In some embodiments, the time is 1 hour, 2 hours, 4 hours, 8 hours, 1 day, 3 days, 5 days, 7 days, 14 days, 28 days, 90 days, 180 days, or 365 days.

In some embodiments, forms of compound I described herein or pharmaceutical compositions thereof are useful for treating neuronal diseases and disorders. A neuronal disease is a disease or condition in which the function of the nervous system of a subject becomes impaired. The neuronal disease or disorder may be a neurological disease or disorder. The neuronal disease or disorder may be associated with a neurodegenerative disease or disorder.

In one aspect, there is provided a method of treating a neuronal disorder in a patient in need thereof, the method comprising administering to the patient a therapeutically effective amount of a form of compound I described herein or a pharmaceutical composition thereof. In some embodiments, the neuronal disorder is alzheimer's disease. In some embodiments, the neuronal disorder is parkinson's disease. In some embodiments, the neuronal disorder is parkinson's dementia. In some embodiments, the neuronal disease is autism. In some embodiments, the neuronal disease is fragile X syndrome. In some embodiments, the disease or disorder is associated with (e.g., characterized by) the accumulation of amyloid plaques. In some embodiments, the neuronal disease is traumatic brain injury. In some embodiments, a patient suffering from a neuronal disorder has suffered traumatic brain injury before, during, or after the onset of the neuronal disorder. In some embodiments, the neuronal disease comprises neuronal damage. The neuronal damage may comprise an atrophy or other reduction in the effective function of the neuron. For example, alzheimer's disease is known to be manifested as damage to neurons, particularly cortical neurons, such as hippocampal neurons and neurons close to the hippocampus. Loss of synapses may be associated with loss of dendritic spines and neurodegeneration.

In some embodiments, the neuronal disease is associated with abnormal dendritic spine morphology, spine size, spine plasticity, spine motility, spine density, and/or abnormal synaptic function. In some embodiments, the neuronal disease is associated with an abnormal (e.g., decreased) level of density of the dendritic spines.

In some embodiments, the neuronal disorder is alzheimer's disease. In some embodiments, the neuronal disorder is parkinson's disease. In some embodiments, the neuronal disorder is parkinson's disease with dementia. In some embodiments, the neuronal disease is autism. In some embodiments, the neuronal disease is stroke. In some embodiments, the neuronal disease is a post-traumatic stress disorder (PTSD). In some embodiments, the neuronal disease is a Traumatic Brain Disorder (TBD). In some embodiments, the neuronal disease is Chronic Traumatic Encephalopathy (CTE). In some embodiments, the neuronal disorder is schizophrenia. In some embodiments, the neuronal disorder is dementia (e.g., dementia vulgaris). In some embodiments, the neuronal disorder is attention deficit/hyperactivity disorder (ADHD). In some embodiments, the neuronal disease is Amyotrophic Lateral Sclerosis (ALS). In some embodiments, the neuronal disease is frontotemporal lobar degeneration (FTLD) (e.g., FTLD-tau, FTLD-TDP, or FTLD-FUS). In some embodiments, the neuronal disorder is memory loss. In some embodiments, the neuronal disease comprises memory loss. In some embodiments, the neuronal disorder is age-related memory loss. In some embodiments, the neuronal disease comprises age-related memory loss. In some embodiments, the neuronal disease is hypertensive encephalopathy. In some embodiments, the neuronal disorder is chronic stress. In some embodiments, the neuronal disorder comprises chronic stress. In some embodiments, the neuronal disease is FTLD-TDP type I. In some embodiments, the neuronal disease is FTLD-TDP type II. In some embodiments, the neuronal disease is FTLD-TDP type III. In some embodiments, the neuronal disease is FTLD-TDP type IV.

Examples of neuronal diseases that can be treated with the forms of compound I described herein or their pharmaceutical compositions or methods described herein include Alexander's disease, Alper's disease, alzheimer's disease, depression, perinatal asphyxia, parkinson's dementia ("PD dementia"), amyotrophic lateral sclerosis, ataxia telangiectasia, dengue disease (bat disease) (also known as Spielmeyer-Vogt-Sjogren- dengue disease), spongiform encephalopathies (e.g., bovine spongiform encephalopathy (mad cow disease), Kuru disease, Creutzfeldt-Jakob disease), Creutzfeldt-Jakob disease (schutzfeldt-Jakob disease), Canavan fatal disease, Cockayne syndrome (costorsher syndrome), corticostomte degeneration, stratum cruzi syndrome (geckmann-stransler syndrome), schwann-louse-schwanner-louse syndrome (geenkunller syndrome-schwanner-von-schwanner syndrome), alzheimer's dementia, alzheimer's disease, and alzheimer's disease, Huntington's disease, HIV-associated dementia, Kennedy's disease, Klebsiella's disease, Lewy body dementia, Machado-Joseph disease (spinocerebellar ataxia type 3), multiple sclerosis, multiple system atrophy, narcolepsy, neuroborreliosis, Parkinson's disease, Belgium-Morse-Schlem disease (Pelizaus-Merzbacher disease), Pick's disease, Primary lateral sclerosis, prion disease, Ralsworth's disease (Refsum's disease), Sandhoff's disease, Schill's disease (Schilder's disease), secondary to acute subarachnoid degeneration, myelogenous anemia, Spinal muscular atrophy, Steele-Richardson-Olszewski disease, Tabes dorsalis, drug-induced Parkinsonism, progressive supranuclear palsy, corticobasal degeneration, multiple system atrophy, idiopathic Parkinson's disease, autosomal dominant Parkinson disease, familial type 1(PARK 1), Parkinson disease 3, autosomal dominant Lewy body (PARK3), Parkinson disease 4, autosomal dominant Lewy body (PARK4), parkinson's disease 5(PARK5), Parkinson's disease 6, autosomal recessive inheritance early-onset (PARK6), Parkinson's disease 2, autosomal recessive inheritance juvenile (PARK2), Parkinson's disease 7, autosomal recessive inheritance early-onset (PARK7), Parkinson's disease 8(PARK8), Parkinson's disease 9(PARK9), Parkinson's disease 10(PARK10), Parkinson's disease 11(PARK11), Parkinson's disease 12(PARK12), Parkinson's disease 13(PARK13) or mitochondrial Parkinson's disease. In some embodiments, the neuronal disorder is alzheimer's disease, parkinson's dementia, autism, stroke, Post Traumatic Stress Disorder (PTSD), Traumatic Brain Disorder (TBD), Chronic Traumatic Encephalopathy (CTE), schizophrenia, dementia (e.g., dementia of ordinary origin), attention deficit/hyperactivity disorder (ADHD), Amyotrophic Lateral Sclerosis (ALS), frontotemporal lobar degeneration (FTLD) (e.g., FTLD-tau, FTLD-TDP, or FTLD-FUS), memory loss (e.g., age-related memory loss), hypertensive encephalopathy, or chronic stress.

In some embodiments, the neuronal disorder is Alzheimer's Disease (AD). Alzheimer's disease is characterized by symptoms of memory loss in early stages of the disease. Apo epsilon 4 vectors are at greater risk for AD. APO epsilon 4 is generally thought to be less effective at clearing a epsilon than the other isoforms and therefore may be associated with greater amyloid burden, tau phosphorylation, synaptic toxicity and reduced synaptic density. Having experienced Traumatic Brain Injury (TBI) is another risk factor for developing AD, and studies have shown that the risk of developing AD is significantly increased for those experiencing TBI. Cognitive decline has been associated with progressive loss of synapses. As the disease progresses, symptoms include confusion, long-term memory loss, paraphrase, vocabulary loss, aggression, irritability, and/or mood swings. In the more advanced stages of the disease, there is a loss of physical function. Patients with Alzheimer's Disease (AD) exhibit a number of characteristic neuropathies, such as increased oxidative stress, mitochondrial dysfunction, synaptic dysfunction, disruption of calcium homeostasis, deposition of age spots and neurofibrillary tangles, and brain atrophy. Without wishing to be bound by any theory, it is believed that both the cause and effect of these neuropathies are the accumulation of a deleterious form of aggregated amyloid β (a β) peptide in the brain. AD-related disorders include: senile dementia of the AD type (SDAT), frontotemporal dementia (FTD), vascular dementia, Mild Cognitive Impairment (MCI) and age-associated memory impairment (AAMI). In some embodiments, there is provided a method of treating or preventing alzheimer's disease, comprising administering to a patient in need thereof a therapeutically effective amount of a form of compound I described herein or a pharmaceutically acceptable salt thereof. In some embodiments, the patient is an Apo epsilon 2 or Apo epsilon 3 carrier. In some embodiments, the patient has already had TBI. In some embodiments, the patient is an Apo epsilon 4 carrier. In some embodiments, the patient is an Apo epsilon 4 vector with TBI.

In some embodiments, the neuronal disease is Fragile X Syndrome (FXS). As known in the art, FXS is a genetic syndrome associated with a variety of disorders (e.g., autism and hereditary intellectual disability). The range of values that a disability can exhibit ranges from mild to severe. It was observed that men with FXS began to develop more serious problems in performing tasks requiring working memory, which typically began after age 40. This was observed to be particularly true for verbal working memory. In some embodiments, the neuronal disease is autism. As is known in the art, autism is a disorder of neurodevelopment. Without wishing to be bound by any theory, it is believed that autism affects information processing in the brain by altering how nerves and synapses connect and organize.

In additional embodiments, compositions and methods for alleviating, reducing, or reversing the symptoms of a neuronal disease or disorder are provided. The symptom may be any symptom described herein.

The terms "memory" and the like refer in a general and customary sense to the process by which a subject encodes, stores and retrieves information. In the context of memory, the terms "encode," "register," and the like refer in a general and customary sense to receiving, processing, and combining information that affects the sensation caused by a chemical or physical stimulus. In this context, the term "storing" or the like refers in a general and customary sense to the creation of a record of encoded information. In this context, the terms "retrieve," "recall," and the like refer to recall of stored information in a general and customary sense. The retrieval may be a response to a prompt, as is known in the art. In some embodiments, memory loss refers to a reduced ability to encode, store, or retrieve information. In some embodiments, the memory may be a recognition memory or a recall memory. In this context, "cognitive memory" refers to the recall of previously encountered stimuli. As is known in the art, the stimulus may be, for example, a word, scene, sound, smell, etc. A more extensive category of memory is "recall memory," which requires retrieval of previously learned information, such as a list of sequences of actions, words or numbers previously encountered by the subject, and the like. Methods for assessing the level of memory encoding, storage and retrieval exhibited by a subject are well known in the art, including the methods disclosed herein. For example, in some embodiments, the method improves memory in a subject in need thereof, wherein the subject has a neuronal disorder. In some embodiments, the method improves memory in the subject. In some embodiments, the method treats neuronal or cognitive impairment in the subject. In some embodiments, the method treats the subject for neuronal damage. In some embodiments, the method treats cognitive impairment in the subject.

Further, for any aspect disclosed herein, in some embodiments, the subject has brain injury. Types of brain injury include brain injury (i.e., destruction or degeneration of brain cells), traumatic brain injury (i.e., injury due to external forces on the brain), stroke (i.e., a vascular event that temporarily or permanently damages the brain, e.g., by hypoxia), and acquired brain injury (i.e., brain injury not present at birth). In some embodiments, the method improves memory in the subject. In some embodiments, the method improves learning in the subject. In some embodiments, the method treats neuronal or cognitive impairment in the subject. In some embodiments, the method treats the subject for neuronal damage. In some embodiments, the method treats cognitive impairment in the subject.

In some embodiments, there is provided a method for promoting dendritic spine production in a patient in need thereof, the method comprising administering to the patient a form of compound I described herein or a pharmaceutically acceptable salt thereof. In some embodiments, there is provided a method of treating or preventing a neuronal disease or disorder comprising administering to a patient in need thereof a therapeutically effective amount of a form of compound I described herein or a pharmaceutically acceptable salt thereof. In some embodiments, there is provided a form of compound I or a pharmaceutically acceptable salt thereof for use in the treatment of a neuronal disease or disorder. In some embodiments, there is provided a form of compound I or a pharmaceutically acceptable salt thereof for use in the manufacture of a medicament for the treatment of a neuronal disease or disorder. In some embodiments, the neuronal disease or disorder is selected from the group consisting of alzheimer's disease, parkinson's dementia, autism, fragile X syndrome, and traumatic brain injury. In some embodiments, the neuronal disease or disorder is alzheimer's disease. In some embodiments, a form of compound I described herein or a pharmaceutical composition thereof inhibits cross-linking of f-actin. In some embodiments, a form of compound I described herein or a pharmaceutical composition thereof is anti-metastatic.

Combination therapy

In one embodiment, the forms of compound I disclosed herein may be used in combination with one or more additional therapeutic agents for treating and/or developed to treat a neuronal disease or disorder.

When used to treat or prevent the above-mentioned diseases and conditions, the forms of compound I described herein or pharmaceutical compositions thereof may be administered with one or more other therapeutic agents, for example, other therapeutic agents approved for use in treating or preventing the particular disease or condition, more particularly agents deemed to constitute the current standard of care. Where combination therapy is contemplated, the active agents may be administered simultaneously, separately or sequentially in one or more pharmaceutical compositions.

Recent strategies for treating AD involve controlling the production or aggregation state of specific isoforms of a β peptide. Additional strategies include preventing, reducing or removing toxic forms of phosphorylated tau. Other strategies involve small molecule targeted enzymes that play a role in producing a β peptide by processing amyloid precursor protein in an attempt to reduce the abundance of a β peptide in the brain. In addition, there is increasing information on the role of non-starch-like neuropathies (e.g., sporadic inheritance of specific mutations in the tauopathy or apolipoprotein E gene), which stimulates additional strategies to combat neurodegeneration.

The one or more additional therapeutic agents may be tacrine (tacrine), donepezil (donepezil), galantamine (galantamine), rivastigmine (rivastigmine), memantine (memantine), levodopa (levodopa), carbidopa (carbidopa), lisuride (lisuride), rasagiline (rasagiline), tolcapone (tolcapone), entacapone (entacapone), clozapine (clozapine), desipramine (desipramine), citalopram (citalopram), nortriptyline (nortriptyline), paroxetine (paroxitine), atomoxetine (atomoxetine), venlafaxine (venlafaxine), amantadine (amantadine), donepezil (donepezil), rivastigmine (rivastigmine), ziprasine (clavine), meglumine (clavine), pramine (meglumine), meglumine (meglumine), venlafaxine (meglumine (e), meglumine (meglumine), meglumine (e), meglumine (e), meglumine (e), venlafaxine), meglumine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), morphine (e), CEP-1347, isradipine (itradipine), DOPA, lithium, riluzole (riluzole), levetiracetam (levetiracetam), ezocabine (ezogabine), pregabalin (pregabalin), rufmamid (rufmamide), felbamate (felbamate), carbamazepine (carbamazepine), valproate (valproate), sodium valproate (sodium valproate), lamotrigine (lamotrigine), phenytoin (phenoytoin), oxcarbazepine (oxcarbazepine), ethosuximide (ethosuximide), gabapentin (gabapentin), tiagabine (tiagabine), topiramate (topiramate), vigabatrin (vigabatrin), viginomycin (viginomycin), phenylbarbitazone (phenazopyramide), prenamide (phenazophylline), doxylamine (interferon beta), interferon (interferon beta-25 beta-interferon (interferon beta), interferon beta-interferon (interferon beta-interferon), interferon beta-25 interferon (interferon beta-interferon, interferon beta-interferon (interferon beta-interferon, interferon beta-interferon, interferon (interferon beta-interferon, beta (beta-beta (interferon, and a, interferon, and a, Ozanizumab (Ozanezumab), apraclimox (arimoclomol), tirasemidi (tirameriv), dexpramipexole (dexpramipexole), pridopidine (pridopidine) or galantamine (galantamine); or phosphoglycerate kinase (PGK) as described in US 2018/0147263. In some embodiments, the one or more additional therapeutic agents may be an acetylcholinesterase inhibitor (AChEI), such as acotiamide (acaotimide), alpha-pinene (alpha-pinene), amberlonitium chloride (ambenonium), dememmonium (demecarbonium), DFP (diisopropyl fluorophosphate), donepezil (donepezil), epothinium chloride (edrophonium), galantamine (galantamine), huperzine a (huperzine a), lactucan (lactucopicrin), ladostigil (ladostigil), neostigmine (neostigmine), physostigmine (physostigmine), pirstine (pyridostigmine), diispropyrniofluorphosphate (dyflos), diphenoxythiocholine (ecothite), carbapenem (rivilene), peimine (peimine), peimine (pezapine), peimine (peimine), peimine (peimine), peimine, thiasislin (thiacymeserine), SPH 1371 (galantamine +), ER 127528, RS 1259 or F3796. In some embodiments, the one or more additional therapeutic agents may be an amyloid scavenging antibody, such as, for example, bapiduzumab (bapineuzumab), sorafenib (solaneezumab), ganterlizumab (ganteneurumab), kruselizumab (crenezumab), pnelumimab (ponezumab), BAN2401, or aducanura (aducanumab).

The one or more additional therapeutic agents may be a sedative hypnotic agent such as chloral hydrate (chloral hydrate), estazolam (estazolam), flurazepam monohydrochloride (flurazepam hydrochloride), pentobarbital (pentobarbital), pentobarbital sodium (pentobarbital), phenobarbital sodium (phenobarbital sodium), secobarbital sodium (secobarbital sodium), temazepam (temazepam), triazolam (triazolam), zaleplon (zaleplon), or zolpidem tartrate (zolpidem tartrate); anticonvulsants such as acetazolamide sodium (acetazolamide sodium), carbamazepine (carbazepine), clonazepam (clonazepam), chlordiazepoxide (clonazepam), diazepam (diazepam), divalproex sodium (divalproex sodium), ethosuximide (ethosuximde), fosphenytoin sodium (foscarnin sodium), gabapentin (gabapentin), lamotrigine (lamotrigine), magnesium sulfate (magnesulfate), phenobarbital sodium, phenytoin sodium (phenoxyoin sodium), primidone, tiagabine (tiagabine), topiramate (topiramate), sodium valproate (valproate) or provaleric acid (valproic acid); antidepressants such as amitriptyline hydrochloride, amphetamine hydrochloride, citalopram hydrobromide, clomipramine hydrochloride, desipramine hydrochloride, doxepin hydrochloride, fluoxetine hydrochloride, imipramine hydrochloride, mirtazapine hydrochloride, nerfazine hydrochloride, nefazine hydrochloride, triamcinolone hydrochloride, doxetazine hydrochloride, dessertraline hydrochloride, triamcinolone hydrochloride, hydrochloric acid hydrochloride, triamcinolone hydrochloride, trolamine hydrochloride, triamcinolone hydrochloride, hydrochloric acid hydrochloride, trolamine hydrochloride, triamcinolone hydrochloride, or triamcinolone hydrochloride; anxiolytics such as alprazolam, buspirone hydrochloride, chlordiazepoxide hydrochloride, chlordiazepoxide, diazepam, doxepin hydrochloride, hydroxyzine (hydroxyzine hydrochloride), hydroxyzine hydrochloride, lorazepam, meprobazone hydrochloride, meprobamate or oxazepam; antipsychotics, such as chlorpromazine hydrochloride, clozapine (clozapine), fluphenazine decanoate (fluphenazine decanoate), fluphenazine heptanoate (fluphenazine enanthate), fluphenazine hydrochloride (fluphenazine hydrochloride), haloperidol (haloperidol), haloperidol decanoate (haloperidol decanoate), haloperidol lactate (haloperidol lactate), loxapine hydrochloride (loxapine hydrochloride), loxapine succinate (loxapine succinate), mesoridazine besylate (mesoridazine hydrochloride), molindone hydrochloride (molindolone hydrochloride), olzapine (olanzapine), perphenazine hydrochloride (oxyperidine hydrochloride), thiopiperazine (chlorpyripine hydrochloride), thioridone (chlorpyrine hydrochloride), thioridazine (thioridone hydrochloride), thioridazine (thioridone), thioridazine (hydrochloride), thioridazine (thioridone, thioridone hydrochloride), thioridone (thioridazine (thioridone, thioridazine hydrochloride), thioridone, thioridazine (thioridone, thioridazine hydrochloride), thioridone, thioridazine (thioridone, thioridazine, thioridone, thioridazine, thioridone, thio; central nervous system stimulants such as amphetamine sulfate (amphetamine sulfate), caffeine (caffeine), dextroamphetamine sulfate (dextroamphetamine sulfate), doxoram hydrochloride (doxam hydrochloride), methamphetamine hydrochloride (methamphetamine hydrochloride), methylphenidate hydrochloride (methylphenidate hydrochloride), modafinil (modafinil), pimoline (pemoline) or phentermine hydrochloride (phentermine hydrochloride); anti-parkinson's disease agents such as amantadine hydrochloride, benztropine mesylate, biperidine hydrochloride, biperidine lactate, bromocriptine mesylate, carbidopa-levodopa, entacapone, levodopa, pergolide mesylate, pramipexole hydrochloride, ropinirole hydrochloride, selegiline hydrochloride, tolcapone hydrochloride or trihexyphenide hydrochloride; or central nervous system agents such as bupropion hydrochloride (bupropion hydrochloride), donepezil hydrochloride (donepezil hydrochloride), droperidol (droperidol), fluvoxamine maleate (fluvoxamine), lithium carbonate (lithium carbonate), lithium citrate (lithium citrate), naratriptan hydrochloride (naratriptan hydrochloride), nicotine polacrilex (nicotine polacrilex), nicotine system (nicotine transermal system), propofol (propofol), rizatriptan benzoate (rizatriptan benzoate), sibutramine hydrochloride monohydrate (sibutramine hydrochloride), sumatriptan succinate (sulbactriptan succinate), sulbactam succinate (succinate), and fluvoxamine hydrochloride (zolamide); cholinergic groups (e.g. parasympathomimetic groups) such as becholine chloride (betamethacholine chloride), ethidium chloride (ethiophonium chloride), neostigmine bromide (neostigmine bromide), neostigmine methylsulfate (neostigmine methyl sulfate), physostigmine salicylate (physostigmine salicylate) or pyridinimine bromide (pyridinimine bromide); anticholinergics such as atropine sulfate, dibromine hydrochloride, glycopyrrolate, hyoscyamine sulfate, propantheline bromide, scopolamine butylbromide or scopolamine hydrobromide; adrenergic (sympathomimetic) compounds such as dobutamine hydrochloride (dobutamine hydrochloride), dopamine hydrochloride (dopamine hydrochloride), metahydroxylamine bitartrate (metamenol bitartrate), norepinephrine bitartrate (norepinephrine bitartrate), phenylephrine hydrochloride (phenylephrine hydrochloride), pseudoephedrine hydrochloride (pseudoephedrine hydrochloride), or pseudoephedrine sulfate (pseudoephedrine sulfate); adrenergic blockers (sympathogens), such as dihydroergotamine mesylate, ergotamine tartrate, ergotamine maleate, or propranolol hydrochloride; skeletal muscle relaxants, such as baclofen (baclofen), carisoprodol (carisoprodol), chlorzoxazone (chlorzoxazone), cyclobenzaprine hydrochloride (cyclobenzaprine hydrochloride), dantrolene sodium (dantrolene sodium), methocarbamol (methocarbamol) or tizanidine hydrochloride (tizanidine hydrochloride); neuromuscular blockers such as atracurium besylate (atracurium besylate), cisatracurium besylate (cisatracurium besylate), dopamine (doxorubium chloride), mivacurium chloride (mivacurium chloride), pancuronium bromide (pancuronium bromide), pipecuronium bromide (piprocuronium bromide), lapachonium bromide (raparinium bromide), rocuronium bromide (rocuronium bromide), succinylcholine chloride (succinylchloride), tubocurarine chloride (tubocurarine chloride) or vecuronium bromide (vecuronium bromide); or corticosteroids such as betamethasone (betamethasone), betamethasone acetate or betamethasone sodium phosphate (betamethasone acetate or betamethasone sodium phosphate), betamethasone sodium phosphate, cortisone acetate (cortisone acetate), dexamethasone (dexamethasone), dexamethasone acetate (dexamethasone acetate), dexamethasone sodium phosphate (dexamethasone sodium phosphate), fludrocortisone acetate (fludrocortisone acetate), hydrocortisone (hydrocortisone), hydrocortisone acetate (hydrocortisone acetate), hydrocortisone cypionate, hydrocortisone sodium succinate (hydrocortisone sodium succinate), methylprednisolone acetate (prednisolone sodium acetate), prednisolone acetate (prednisolone sodium phosphate), prednisolone acetate (prednisolone sodium acetate), prednisolone sodium phosphate (prednisolone acetate), prednisolone sodium acetate (prednisolone sodium acetate), prednisolone sodium phosphate (prednisolone sodium acetate), prednisolone sodium acetate (prednisolone sodium phosphate), prednisolone sodium acetate (prednisolone sodium acetate), prednisolone sodium acetate (prednisolone sodium phosphate) Prednisolone butyrate, prednisone triamcinolone acetonide, triamcinolone acetonide or triamcinolone acetonide diacetate.

Reagent kit

Also provided herein are kits comprising a form of compound I described herein or a pharmaceutical composition thereof, optionally a second active agent, and a suitable package. In one embodiment, the kit further comprises instructions for use. In one aspect, a kit comprises a form of compound I described herein or a pharmaceutical composition thereof, and a label and/or instructions for using the pharmaceutical composition in treating an indication comprising a disease or condition described herein.

Also provided herein are articles of manufacture comprising a form of compound I described herein or a pharmaceutical composition thereof in a suitable container. The container may be a vial, jar, ampoule, pre-loaded syringe, nebulizer, aerosol dispensing device, dropper, or iv bag.

Synthesis of

In some embodiments, the present disclosure provides methods for synthesizing compound I.

The methods of the present disclosure can be performed using the methods disclosed herein and conventional modifications thereof, which will be apparent in view of the disclosure herein and methods known in the art. In addition to the teachings herein, conventional and well known synthetic methods may be used. The synthesis of typical compounds described herein, e.g., compound I, compound a, etc., or other compounds of formula or disclosed herein, can be accomplished as described in the examples below. If available, reagents may be purchased commercially from, for example, Sigma Aldrich or other chemical suppliers.

Typical embodiments of the compounds according to the invention may be synthesized using the general reaction schemes described below. A given agent may be defined as a general class or category (e.g., function or structure) that should be construed to include any agent that matches a given descriptor.

The compounds of the present disclosure can be prepared from readily available starting materials using, for example, the following general methods and procedures. It is to be understood that, unless otherwise indicated, other process conditions may also be employed given typical or preferred process conditions (i.e., reaction temperature, time, molar ratios of reactants, solvents, pressures, etc.). Optimal reaction conditions may vary with the particular reactants or solvents used, but such conditions may be determined by one skilled in the art by routine optimization procedures.

In addition, it will be apparent to those skilled in the art that conventional protecting groups may be as described herein. Suitable protecting groups for various functional groups and suitable conditions for protecting and deprotecting particular functional groups are known in the art. For example, a number of protecting groups are described in t.w.greene and g.m.wuts (1999) protecting groups in organic synthesis, third edition, Wiley, New York and references cited therein.

The starting materials for the following reactions are generally known compounds or can be prepared by known methods or obvious modifications thereof. For example, many starting materials are available from suppliers such as: aldrich Chemical Co.) (Milwaukee, Wisconsin, USA, Wisconsin, wi), baheng (torran, California, USA), emmca-kamike or Sigma (Emka-Chemce or Sigma), st louis, Missouri, USA). Other compounds may be prepared by the procedures described in the following standard reference texts or obvious modifications thereof: reagents for Organic Synthesis from Fieser and Fieser's (Fieser and Fieser's Reagents for Organic Synthesis) Vol.1-15 (John Wiley and Sons, 1991), "Roder's Carbon Chemistry (Rodd's Chemistry of Carbon Compounds), Vol.1-5 and" supplements (supplements) (Elsevier Science Publishers, 1989), "Organic Reactions (Organic Reactions), Vol.1-40 (John Willi-Gilg-D., 1991)," high-grade Organic Chemistry (March's Advanced Organic Chemistry), Inc. (Portrait-Gilg-D., Inc., Trans-Gilg-D., 1985, Portrai-D., Inc.), and "9" (Portrai-D. published), Inc. (Portrai-Giraff-D., Inc., 1981).

The term "inert organic solvent" or "inert solvent" refers to a solvent that is inert under the reaction conditions described in connection therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran ("THF"), dimethylformamide ("DMF"), chloroform, methylene chloride (or dichloromethane), diethyl ether, methanol, pyridine, acetic acid, and the like). Unless otherwise indicated, the reaction is carried out under an inert gas such as nitrogen or argon. The "solvent" need not be inert.

In each exemplary embodiment, it may be advantageous to separate the reaction products from each other and/or from the starting materials. The desired product of each step or series of steps is isolated and/or purified (hereinafter isolated) to the desired degree of homogeneity by techniques commonly used in the art. Typically, such separations involve heterogeneous extraction, crystallization from a solvent or solvent mixture, distillation, sublimation, or chromatography. Chromatography may involve a variety of methods including, for example: reverse phase and normal phase; size exclusion; ion exchange; high, medium and low pressure liquid chromatography methods and apparatus; small-scale analysis; simulated Moving Bed (SMB) and techniques for preparing thin or thick layer chromatography, as well as small scale thin layer and flash chromatography.

Another class of separation methods involves treating the mixture with a medium selected to bind or otherwise separate the desired product, unreacted starting materials, reaction byproducts, and the like. Such media include adsorbents or absorbents such as activated carbon, molecular sieves, ion exchange media, and the like. Alternatively, acids (in the case of basic materials) or bases (in the case of acidic materials), binding reagents such as antibodies, binding proteins, selective chelators such as crown ethers, liquid/liquid ion extraction reagents (LIX), and the like.

The choice of a suitable separation method depends on the nature of the materials involved. For example, boiling point and molecular weight in distillation and sublimation, presence or absence of polar functional groups in chromatography, stability of materials in acidic and basic media in heterogeneous extraction, and the like. Those skilled in the art will apply the techniques most likely to achieve the desired separation.

Raw materials and reagents were purchased from Shanghai energy Tantake technologies, Inc. and were used without further purification. The oxygen and moisture sensitive reactions were carried out under a nitrogen atmosphere. The reaction was monitored by Thin Layer Chromatography (TLC), and the thin layer chromatography plate was visualized by exposure to uv light. Elution was monitored at 254 nm. Flash column chromatography is typically performed on silica gel (300-400 mesh). Unless otherwise indicated, yield refers to the isolated chromatographically and spectroscopically homogeneous material.

Scheme 1 represents an exemplary synthesis of compound I and may be performed according to embodiments described herein. It is considered that the exemplary synthesis shown in scheme 1 may be particularly advantageous. For example, the synthesis avoids toxic reagents. The synthesis may also utilize milder reaction conditions and may require fewer purification steps (e.g., avoiding column chromatography). The synthesis may also provide higher yields. The specific reaction conditions and reagents used in scheme 1 are discussed below.

Compound I can be synthesized according to the general scheme, scheme 1:

scheme 1

In some embodiments, there is provided a method of preparing compound I, or a pharmaceutically acceptable salt thereof:

comprising contacting compound a with compound B under first reaction conditions comprising a halide to form compound I:

in some embodiments of the process for preparing compound I, the halide is an alkali metal halide. In some embodiments of the process for preparing compound I, the alkali metal halide is an alkali metal iodide. In some embodiments of the process for preparing compound I, the alkali metal iodide is potassium iodide. In some embodiments of the process for preparing compound I, the first reaction conditions further comprise an inorganic base. In some embodiments of the process for preparing compound I, the inorganic base is potassium carbonate. In some embodiments of the process for preparing compound I, the first reaction conditions comprise a temperature of 65 to 120 ℃.

In some embodiments, a method of making compound a and optionally compound I comprises contacting compound C with compound D under second reaction conditions comprising a protic acid to form compound a:

in some embodiments of the process for preparing compound I, the protic acid is an organic acid. In some embodiments of the process for preparing compound I, the organic acid is acetic acid. In some embodiments of the process for preparing compound I, the protic acid is present in a molar ratio relative to compound C and compound D that is greater than the stoichiometric amount. In some embodiments of the method of making compound I, the protic acid dissolves compound C and compound D. In some embodiments of the process for preparing compound I, acetic acid is a non-inert solvent.

In some embodiments, a method of making compound B and optionally compound I comprises contacting compound E with p-toluenesulfonyl chloride under third reaction conditions comprising a silver salt to form compound B:

in some embodiments of the process for preparing compound I, the silver salt is Ag2And O. In some embodiments of the process for preparing compound I, the third reaction conditions further comprise an alkali metal iodide. In some embodiments of the process for preparing compound I, the alkali metal iodide is potassium iodide.

Examples

Example 1

Experimental procedure

Compound I was subjected to various crystallization conditions including solid vapor diffusion, anti-solvent addition, liquid vapor diffusion, slow cooling, slurry transitions at various temperatures, temperature cycling and slow evaporation. The crystalline form of compound I was analyzed by X-ray powder diffraction (XRPD), Differential Scanning Calorimetry (DSC), and thermogravimetric analysis (TGA), while the metastable form was analyzed by XRPD. XRPD was performed using Panalytical X' Pert3 Powder XRPD with Si zero background scaffold. The 2 θ position was calibrated against a Panalytical Si reference standard disk. The instrument parameters used are listed in Table 1-1.

Tables 1 to 1: parameters for XRPD analysis

TGA data was collected using TA Q500 and Q550 from TA Instruments. DSC was performed using TA Q2000 from TA Instruments. DSC was calibrated using an indium reference standard and TGA was calibrated using a nickel reference standard. Table 1-2 lists the detailed parameters used in the TGA and DSC tests.

Tables 1 to 2: parameters for TGA and DSC testing

TGA DSC
Method Slowly ascending Slowly ascending
Sample plate Platinum, open end Aluminum, crimping
Temperature of RT-desired temperature 25 ℃ to desired temperature
Rate of heating 10℃/min 10℃/min
Purge gas N2 N2

PLM images were captured using a Zeiss Axio scope.a1 microscope. PLM images of samples obtained in single crystal growth experiments were taken at room temperature using Shanghai Zerewing PXS9-T stereomicroscope.

DVS was measured by SMS (surface measurement system) DVS Intrinsic. The relative humidity at 25 ℃ was calibrated according to the deliquescence point of LiCl, Mg (NO3)2 and KCl. The actual parameters of the DVS test are shown in tables 1-3.

Tables 1 to 3: parameters for DVS testing

The detailed chromatographic conditions for purity analysis are listed in tables 1-4 using an Agilent 1260HPLC equipped with a VWD detector.

Tables 1 to 4: HPLC conditions and parameters

Five crystalline forms were observed for compound I, including one anhydrous form (form I), one hydrate/solvate (form IV) and three metastable forms (forms II, III and V). Forms II and III are present only in the wet state and no solids are isolated for thermal analysis. Form V solid was obtained, however, form V changed to form I when dried in a vacuum oven at about 50 ℃. A summary of the isolated forms and conditions are shown in tables 1-5 and tables 1-6.

Tables 1 to 5: isolation conditions for Compound I form

Method Results
Solid vapor diffusion Form I and form V
Addition of anti-solvent Forms I, II, IV and V, amorphous
Liquid vapor diffusion Form I and form V
Slowly cooling Forms I, II and V, amorphous
Slurry conversion Form I and form V
Temperature cycling Form I and form V
Slow evaporation Form I and form III

Tables 1 to 6: isolation and brief description of Compound I form

Crystalline forms Weight loss (%) Endotherm (Peak,. degree.C.) Description of the invention
Form I Can be ignored 70.3 Anhydrous matter
Form II Not applicable to1 Not applicable to1 Metastable state
Form III Not applicable to2 Not applicable to2 Metastable state
Form IV 4.4 48.0,58.9 Solvates/hydrates
Form V 6.6 45.7 (transition), 70.2 Metastable state

Example 2

Forms of compound I

Compound I form I

Compound I form I is the anhydrous form of compound I and it is considered the most thermodynamically stable polymorph of the compound. To date, form I is the only stable anhydrate form identified for compound I.

Compound I form I is obtained by various methods. XRPD results in fig. 1 show that form I is crystalline.

The TGA/DSC curves (fig. 2 and 3) show negligible weight loss and an endothermic peak at 70.3 ℃. From these characterization data, form I is considered to be an anhydrate.

Compound I form I was isolated using the methods provided herein.

Compound I form II

Form II was obtained by various methods. XRPD results showed form II to be crystalline. It exists only as a wet cake and upon storage becomes a mixture of forms II and C or a mixture of forms III and V. To further investigate form II, several attempts to re-prepare were made, resulting in the formation of forms III, IV, V or amorphous forms, indicating that form II is a metastable form. Thus, DSC and TGA data for form II were not obtained.

Compound I form III

Form III was obtained by various methods. XRPD results showed form III to be crystalline. Form III solid obtained by slow evaporation in MeOH was rapidly converted to form I after exposure to dew during XRPD analysis. To further investigate form III, several attempts to re-prepare were made, resulting in the formation of form I or amorphous form, indicating that form III is a metastable form. Thus, DSC and TGA data for form III were not obtained.

Compound I form IV

By adding H to MEK stock solution2Reaction of OThe solvent obtained form IV. XRPD results showed form IV to be crystalline. The TGA/DSC curve shows a weight loss of 4.4% before 150 ℃ and two endothermic peaks at 48.0 and 58.9 ℃. Based on these characterization data, form IV is considered to be a hydrate or solvate.

Compound I form V

Form V is obtained by various methods. XRPD results showed form V to be crystalline. The TGA/DSC curve shows that the weight loss before 150 ℃ is 6.6% and two thermal events occur at 45.7 ℃ (transition, onset) and 70.2 ℃ (melting, peak). XRPD data also showed that the form V solid was converted to form I after drying in vacuo at about 50 ℃. Upon dehydration/desolvation at about 50 ℃, compound I form V is converted to compound I form I. These results indicate that form V is a metastable hydrate/solvate that converts to form I after dehydration/desolvation.

Amorphous compound I

Amorphous compound I was observed under the conditions described herein.

Example 3

Methods of compound I form

Solubility in water

The approximate solubility of compound I in 20 single solvents was determined at room temperature. Approximately 2mg of the powder sample was added to a 3mL glass bottle. The corresponding solvent was added stepwise (50 μ L → 100 μ L → 300 μ L → 1000 μ L) to each vial until the powdered solid was visually completely dissolved or a total volume of 1mL of solvent was added. Approximate solubility values were calculated based on sample mass, solvent volume and experimental observations. The results summarized in Table 3-1 are used to guide solvent selection in the design of subsequent polymorph screening experiments.

Table 3-1: solubility of compound I at room temperature; indicates that the sample precipitated from the clear solution after about 10 minutes at room temperature (S < 40mg/mL) and additional solvent was added to completely dissolve the remaining solid

Solvent(s) Solubility (mg/mL) Solvent(s) Solubility (mg/mL)
DMF S>44 EtOH 22>S>7.3*
DCM S>38 Acetone (II) 18>S>6*
THF S>36 Toluene 18>S>6*
1, 4-dioxane S>32 MIBK 8.3>S>2.5*
MeOH 42>S>21* IPAc 5.7>S>1.7*
EtOAc 40>S>20* CPME S<2.6*
2-MeTHF 40>S>20* Heptane (Heptane) S<2.2*
MEK 36>S>18* MTBE S<2*
DMSO 36>S>18* H2O S<1.8*
Acetonitrile 24>S>8* IPA S<1.9*

Solid vapor diffusion

The solid vapor diffusion experiment was performed using 9 different solvents. Approximately 15mg of starting material was weighed into a 4mL vial and then placed into a 20mL vial containing 3mL of volatile solvent. The 20mL vial was then sealed with a cap and held at room temperature for 7 days to allow the solvent vapor to interact with the sample. If the solid is completely dissolved in the solvent, slow evaporation takes place at room temperature. The obtained solid was analyzed by XRPD. The results summarized in Table 3-2 indicate that form I and form V are observed.

Tables 3-2: summary of solid vapor diffusion experiments for compound I; indicates that the solid powder is completely dissolved in the solvent, thus slowly evaporating to obtain a solid sample

Solvent(s) Solid forms
Acetone (II) Form I
THF Form I
EtOH Form I + form V
H2O Form I
EtOAc Form I
Dioxane(s) Form I
Toluene Form I
DCM Form I
Acetonitrile Form I

Addition of anti-solvent

The anti-solvent addition experiment was performed under 12 conditions. Approximately 20mg of starting material was dissolved in 0.5 or 1.0mL of solvent to obtain a clear solution. The solution was then filtered and the anti-solvent added at a rate of 0.2mL per step with magnetic stirring. The anti-solvent was added until no more solids precipitated or the total solvent volume reached 5.0 mL. If the solution remains clear, the sample is stirred at 5 ℃ to induce precipitation. If no precipitation occurred at 5 ℃, slow evaporation was performed at room temperature to obtain a solid. The solid precipitate was separated by centrifugation for subsequent XRPD analysis. The results in tables 3-3 indicate that forms I, II, III, IV and V as well as amorphous forms are observed.

Tables 3 to 3: summary of antisolvent addition experiments for compound I; indicates that the solid is obtained by slow evaporation at room temperature

1: the sample was retained in the solvent mixture and re-analyzed on day 3; 2: samples were prepared and analyzed immediately on trial 2; 3: samples were prepared and analyzed immediately on trial 3; 4: leave the sample in the solvent mixture and re-analyze on day 2 of the 2 nd trial

Liquid vapor diffusion

The liquid vapor diffusion experiment was performed under 6 conditions. Briefly, about 20mg of starting material was dissolved in 1mL of solvent and filtered into a 5mL vial using a PTFE syringe filter with a pore size of 0.45 μm. The filtrate was then placed in a 20mL vial containing 3mL of volatile solvent. The 20mL vial was sealed with a cap and kept at room temperature to allow the organic solvent vapor to interact with the solution for at least 14 days. The precipitate was then isolated for XRPD analysis. The results summarized in tables 3-4 indicate that form I and form II are observed.

Tables 3 to 4: summary of liquid vapor diffusion experiments for Compound I

Solvent(s) Anti-solvent Solid forms
MEK MTBE No precipitation
DMSO CPME No precipitation
Dioxane(s) MTBE No precipitation
EtOAc IPA Form I + form V
EtOH H2O Form I + form V
2-MeTHF IPA Form I + form V

Slowly cooling

The slow cooling experiments were performed in 7 solvents or solvent combinations. Briefly, approximately 20mg of starting material was dissolved in 0.5 or 1mL of solvent at 50 ℃ and filtered into a new vial using a PTFE syringe filter with a pore size of 0.45 μm. The filtrate was slowly cooled from 50 ℃ to 5 ℃ at a rate of 0.1 ℃/min. The solid obtained was kept at a constant temperature of 5 ℃ before centrifugation and XRPD analysis. If the solution remained clear, slow evaporation was performed to obtain a solid for XRPD analysis. The results are summarized in tables 3-5. The results indicate that forms I, III, V and amorphous forms are observed.

Tables 3 to 5: summary of slow cooling experiments for compound I; indicates that the solution remained clear after cooling, and therefore slowly evaporated

Solvent (v/v) Solid forms
MeOH Form III1
MEK Form I
Dioxane: toluene, 1:1 Form I
Ethyl acetate to acetonitrile, 1:1 Form I
DCM∶IPA,4∶1 Form I + form V
THF to heptane, 4:1 Form I
DMSO∶CPME,4∶1 Gel, amorphous

1: drying to form I

Slurry conversion

Slurry conversion experiments were performed in 17 solvent systems at room temperature or 60 ℃. Approximately 20mg of the starting material was suspended in 0.35mL of solvent at the target temperature for 7 days. The remaining solid was isolated for XRPD analysis. If the sample is completely dissolved, additional solids are added until a total of about 100mg of powdered solids are added. The solid was then separated by centrifugation and analyzed by XRPD. The results summarized in tables 3-6 indicate that form I and form V are observed.

Tables 3 to 6: summary of slurry conversion experiments for Compound I

1: additional solids were added (< 100 mg); 2: after addition of-100 mg of solid the solution remained clear and after 7 days the solution was kept at 5 ℃ to obtain a solid for analysis

Temperature cycling

Temperature cycling experiments were performed under 6 conditions. Briefly, about 20mg of starting material was dissolved in 0.35mL of solvent and magnetically stirred. The suspension was kept at 50 ℃ for 2 hours, cooled to 5 ℃ at a rate of 0.05 ℃/min, kept at 5 ℃ for 2 hours, then raised to 50 ℃ at a rate of 3 ℃/min, kept at the same temperature for 2 hours, and then cooled to 5 ℃ at a rate of 0.05 ℃/min. Samples were stored at 5 ℃ prior to centrifugation. The results are summarized in tables 3-7. Form I and form V are formed.

Tables 3 to 7: summary of temperature cycling experiments for Compound I

Solvent (v/v) Solid forms
EtOAc∶CPME,1∶4 Form I
THF∶MTBE,1∶4 Form I
EtOH n-heptane, 1: 2 Form I
DCM∶MTBE,1∶2 Form I
DMSO∶H2O1∶4 Form V
MeOH∶IPA,1∶4 Form I

*: drying to form I

Slow evaporation

The slow evaporation experiments were performed under 7 conditions. Approximately 20mg of the starting material was dissolved in 0.5 or 1.0mL of solvent and filtered using a PTFE syringe filter with a pore size of 0.45 μm. Then using a needle with 3 pinholesThe filtrate was covered and stored at RT. The resulting solid was collected for XRPD analysis. The results are summarized in tables 3-8. Indicating that form I and form III crystalline forms are observed.

Tables 3 to 8: summary of slow evaporation experiments for compound I; indicates that the solution remained clear after cooling, and therefore slowly evaporated

Solvent (v/v) Solid forms
THF Form I
DCM Form III + additional peak1
MeOH Form III1
EtOAc Form I
MEK Form I
Acetonitrile Form I
1, 4-dioxane: H2O,9∶1 Form I

1: after drying, converting to form I

Example 4

Solid state stability of Compound I

Solid state stability of Compound IEvaluation was performed at 30 ℃/65% RH (open petri dish and closed vessel) and 40 ℃/75% RH (open petri dish and closed vessel) for 4 weeks. Detailed procedures for stability evaluation are listed: 1) weigh approximately 10mg of compound I starting material into a 3mL glass vial; 2) for open petri dish conditions, useCover the vial and stab 6 wells. For closed conditions, the vials were each sealed with a cap and kept under the respective conditions; for the 40 ℃/75% RH conditions, the vial is placed in a 40 ℃/75% RH stabilization chamber; for the condition of 30 ℃/65% RH, the small bottle is placed in a closed atmosphere generated by biochemical saturated potassium iodide aqueous solution at 30 ℃; 4) samples were collected after 3 days, 1 week, 2 weeks and 4 weeks for XRPD, PLM and HPLC purity.

According to XRPD results, no change in form was observed after storage under all the above conditions. The PLM characterization results indicated that the compound I sample remained as irregular, flaky crystals with agglomeration. HPLC analysis showed that no significant decrease in HPLC purity was observed after 4 weeks at 30 ℃/65% RH (open and closed) and 40 ℃/75% RH (open and closed).

Solid state stability evaluations of compound I form I starting materials were performed at 30 ℃/65% RH (closed and open) and 40 ℃/75% RH (closed and open). X-ray powder diffraction (XRPD), polarization microscope (PLM) and HPLC purity were characterized at 4 sampling points (3 days, 1 week, 2 weeks, 4 weeks). No formal change and significant HPLC purity reduction of compound I form I was observed within 4 weeks at both 30 ℃/65% RH and 40 ℃/75% RH. The solid state stability evaluation results are summarized in Table 4-1.

For single crystal structure determination, 83 single crystal growth experiments were performed by different methods including slow evaporation, liquid vapor diffusion, slow cooling, heating-cooling, and solvent-thermal synthesis. The stability evaluation experiments were performed using a stability chamber at 40 ℃/75% RH, whereas the 30 ℃/65% RH conditions were established in a 30 ℃ biochemical incubator with saturated aqueous potassium iodide solution.

Table 4-1: summary of solid state stability evaluation of Compound I form I (HPLC purity)

The compound I starting material was the delivered sample. The starting material was characterized by XRPD, thermogravimetric analysis (TGA), Differential Scanning Calorimetry (DSC), PLM and Dynamic Vapor Sorption (DVS).

TGA/DSC results of the starting material of Compound I show a weight loss of 0.45% before 150 ℃ and

a significant endotherm at 68.2 ℃ (starting temperature) occurs, probably due to melting. The PLM results show that the starting material exhibits an irregular morphology and agglomerates. The DVS plot (FIG. 12) shows a water uptake of 0.64% at 25 ℃/80% RH and 21.43% at 25 ℃/95% RH. No change in form was observed after DVS testing, as shown in figure 13. Sample information and characterization results are summarized in Table 4-2.

Tables 4-2: summary of characterization of the starting Material of Compound I

Weight (D) Weight loss (%, up to 150 ℃ C.) Endotherm (. degree.C., onset)
1.08g 0.45 68.2

Example 5

Growth of single crystal

A total of 83 single crystal growth experiments of compound I were performed by different methods including slow evaporation, liquid vapor diffusion, slow cooling, heat-cooling and solvent-thermal synthesis. However, only lamellar and overlapping crystal samples were obtained, which were too thin for SCXRD characterization. Typical experimental procedures and details are described.

Slow evaporation

A total of 40 slow evaporation experiments were performed and some single crystal samples with a flake-like morphology were obtained. Typical experimental procedure: compound I starting material was weighed into a 3mL glass vial, and the selected solvent was added to the vial to dissolve the solid (accelerated dissolution using a vortex shaker or sonicator). The solution was filtered into a 4mL shell vial (44.6 mm. times.14.65 mm) using a PTFE filter (0.45 μm) and a disposable syringe. In part of the experiments, a small amount of compound I starting material was added as seed. Subsequently, the shell vials were covered with PE-Plug with pinholes on top and left to evaporate slowly at the corresponding temperature. After several days, the samples were observed by PLM. The detailed experimental information is shown in tables 5-1, 5-2, 5-3 and 5-4.

Table 5-1: slow Evaporation Single Crystal growth experiment of Compound I

a: compound I; b: a small amount of compound I is added in the experiment as seed crystal; c: the experimental results were observed after 4 days of slow evaporation

Tables 5-2: slow Evaporation Single Crystal growth experiment of Compound I

a: compound I; b: a small amount of compound I is added in the experiment as seed crystal; c: the experimental results were observed after 3 days of slow evaporation

Tables 5 to 3: slow Evaporation Single Crystal growth experiment of Compound I

a: compound I starting material; b: a small initial amount of compound I was seeded in this batch of experiments; c: observing the experimental result after slowly evaporating for 1 day; d: the experimental results were observed after 4 days of slow evaporation

Tables 5 to 4: slow Evaporation Single Crystal growth experiment of Compound I

a: compound I starting material; b: no seed crystal is added in the experiment group; c: observing the experimental result after slowly evaporating for 1 day

Slow evaporation

A total of 22 liquid vapor diffusion experiments were performed to obtain some single crystal samples having a flake morphology.

Typical experimental procedure: compound I starting material was weighed into a 3mL glass vial, and the selected solvent was added to the vial to dissolve the solid (dissolution could be accelerated using a vortex shaker or sonicator). The solution was filtered into a 4mL shell vial (44.6 mm. times.14.65 mm) using a PTFE filter (0.45 μm) and a disposable syringe. In some experiments, a very small amount of compound I starting material was added as seed to induce crystallization. Subsequently, the shell vial was covered with PE-Plug with a pinhole on top. The 4ml shell vial was then placed into a 20ml glass vial containing 3.0ml of anti-solvent, sealed and stored at the corresponding temperature for liquid vapor diffusion. After several days, the samples were observed by PLM. The detailed experimental information is shown in tables 5-5, tables 5-6 and tables 5-7.

Tables 5 to 5: liquid vapor diffusion single crystal growth experiment of Compound I

a: compound I starting material; b: a small amount of compound I is added in the experiment as seed crystal; c: observation of the results of the experiment after 3 days of liquid vapor diffusion

Tables 5 to 6: liquid vapor diffusion single crystal growth experiment of Compound I

a: compound I starting material; b: a small amount of compound I is added in the experiment as seed crystal; c: observation of the results of the experiment after 4 days of liquid vapor diffusion

Tables 5 to 7: liquid vapor diffusion single crystal growth experiment of Compound I

a: compound I starting material; b: seed crystals are not added in the batch experiment; c: EtOAc was saturated with H2O; d: observation of the results of the experiment after 2 days of liquid vapor diffusion

Slowly cooling

A total of 6 slow cooling experiments were performed to obtain some single crystal samples with a platelet morphology.

Typical experimental procedure: compound I starting material was weighed into a 3mL glass vial and 0.5mL of the selected solvent was added. After accelerating the dissolution process by means of a vortex shaker or an ultrasonic device, the suspension is kept in an oven at 50 ℃ for about 0.5 hour. The hot solution was then filtered into another 3ml glass vial (0.45 μm) with a PTFE filter and a 2.0ml disposable syringe (the PTFE filter, disposable syringe and 3ml glass vial were preheated at 50 ℃) and a small amount of starting material was added to the vial as seeds. The vial was then sealed and placed in a biochemical incubator for slow cooling (cooling program: 50 ℃ → 5 ℃, 0.01 ℃/min).

Tables 5 to 8: slow Cooling Single Crystal growth experiment of Compound I

a: compound I starting material; b: and (3) cooling procedure: 50 ℃→ 5 ℃, 0.01 ℃/min; c: the results of the experiment were observed after slow cooling for 4 days (end of cooling)

Heating-cooling

A total of 9 heating-cooling experiments were performed to obtain some single crystal samples with a lamellar morphology.

Typical experimental procedure: compound I starting material was weighed into a 3mL glass vial and 0.5mL of the selected solvent was added. After accelerating the dissolution process by means of a vortex shaker or an ultrasonic device, the suspension is kept in an oven at 50 ℃ for about 0.5 hour. The hot solution was then filtered into another 3ml glass vial (0.45 μm) with a PTFE filter and a 2.0ml disposable syringe (the PTFE filter, disposable syringe and 3ml glass vial were preheated at 50 ℃). A small amount of starting material was added as seed to the vial. The vial was then sealed and placed in a biochemical incubator for heat-cooling (heating-cooling program: 50 ℃→ 5 ℃, 0.05 ℃/min, 5 cycles).

Tables 5 to 9: heating-Cooling Single Crystal growth experiment of Compound I

a: compound I starting material; b: and (3) cooling procedure: 50 ℃→ 5 ℃, 0.05 ℃/min, 5 cycles; c: the results of the experiment were observed after slow cooling for 4 days (end of cooling)

Solvent-heat

A total of 6 solvent-thermal experiments were performed to obtain a number of single crystal samples with needle-like morphology. Typical experimental procedure: compound I starting material was weighed into a 3mL glass vial and 0.4mL of the selected solvent was added. Subsequently, the vial was placed in a hydrothermal reactor, sealed and placed in an oven for a solvent-thermal experiment (temperature program: 25 ℃ → 80 ℃ → 25 ℃).

TABLE 5-10 solvent-thermal Single Crystal growth experiments for Compound I

a: compound I starting material; b: temperature program: 25 ℃→ 80 ℃→ 25 ℃; c: the results of the experiment were observed after 3 days of reaction (end of cooling)

Example 6

Synthesis of

Tosylation of Compound E to form Compound B

Compound E (99.8g) was dissolved in anhydrous DCM (2.0L) and stirred at room temperature. After 5 minutes, potassium iodide (18.1g), Ag2O (179.8g) and p-toluenesulfonyl chloride (108.5g) were added to the solution in this order. The reaction mixture is stirred under N2Followed by vigorous stirring overnight. After removal of the solids by filtration over celite, the filtrate was concentrated and purified by column chromatography (PE: EA ═ 1:1 to PE: EA ═ 1: 2 to DCM: MeOH ═ 25: 1) to give compound B (110.0g, 61.4% yield) as a colourless oil.1H NMR (FIG. 18) (400)

MHz,CDCl3)δ7.75(d,J=8.0Hz,2H),7.34(d,J=8.0Hz,2H),4.18-4.15(m,2H),3.72-3.59(m,14H)。

Condensing compound C and compound D to form compound A

In a 3-L flask, compound C (160.3g) and compound D (172.0g) were dissolved in AcOH (1).5L). The mixture was refluxed at 105 ℃ for 3.5 hours. After cooling to room temperature, the solution was poured into ice water and then filtered. The filtrate was concentrated and purified by recrystallization using EtOH (-1.4L) to yield compound a as a dark gray solid (105.7g, 36.3% yield). And (3) recrystallization procedure: the crude product was dissolved in 1.4LEtOH to form a solution at 80 ℃. The solution was refluxed for a few minutes and then slowly cooled to room temperature. The solid was collected and washed with EtOH. The solid was dried under vacuum at 50 ℃ for about 8 hours.1H NMR (FIG. 19) (400MHz, CD)3OD)δ7.95-7.91(m,4H),7.51-7.47(m,1H),7.40-7.36(m,1H),6.93-6.91(m,2H)。

Nucleophilic addition of Compound B to Compound A to form Compound I

Compound I is synthesized by condensation of compound a and compound B. First, the condensation of compound a and compound B was attempted on a small scale. A flask was charged with Compound B (5.0g), Compound A (3.3g), and K2CO3(4.0g), KI (0.2g) and dry DMF (60 mL). The resulting mixture is stirred under N2Then, the mixture was heated at 80 ℃ overnight. After cooling to room temperature, the product was extracted into EA by washing the aqueous layer with EA. The EA solvent was removed under reduced pressure and the residue was purified by column chromatography (PE: EA ═ 3: 1) to give a white powder (4.3g, 74.0% yield). The preparation of compound I is then scaled up to several hundred grams. Compound B (105.0g), compound A (68.5g), K2CO3The flask was charged with (83.4g), KI (5.0g) and dry DMF (1.1L). The resulting mixture is stirred under N2Then, the mixture was heated at 80 ℃ overnight. After cooling to room temperature, the aqueous layer (about 5L H) was washed by EA (about 7L)2O) extraction of the product into EA. The solvent of EA was removed under reduced pressure and the residue was purified twice by column chromatography (PE: EA ═ 3: 1) to give a white powder (-110.0 g). Two batches of compound I were mixed into DCM to form a solution. The DCM solvent was evaporated under reduced pressure to give pure compound I (103.0g, 80.8% yield). The structure of the compound I is shown in the specification1Verification by H NMR (FIG. 20) and MS (FIG. 21)。MS(m/z):[M+H]+=404.1。1H NMR (400MHz, CDCl3) δ 8.18(d, J ═ 8.0Hz, 1H), 8.11(d, J ═ 8.0Hz, 2H), 7.82(d, J ═ 4.0Hz, 1H), 7.49-7.45(m, 1H), 7.38-7.34(m, 1H), 7.01-6.98(m, 2H), 4.18-4.15(m, 2H), 3.84-3.81(m, 2H), 3.67-3.61(m, 10H), 3.55-3.53(m, 2H). The HPLC purity of Compound I was also tested (HPLC, 230nm and 254nm UV). The HPLC purity of Compound I was determined to be 99.8 area% when measured at 230nm and 99.7 area% by HPLC at 254nm (tables 6-1 and 6-2, respectively). The XRPD pattern of compound I prepared is consistent with form I.

Table 6-1: HPLC purity of Compound I at 230nm

Peak numbering RRT Area%
1 1.00 99.82
2 1.49 0.18

Table 6-2: HPLC purity of Compound I at 254nm

Peak numbering RRT Area%
1 1.00 99.68
2 1.06 0.09
3 1.49 0.24

The material sources are in accordance with tables 6 to 3

Tables 6 to 3: sources of materials

Example 7

In vitro spine formation using benzothiazole compounds

To demonstrate the efficacy of the compounds in promoting echinogenesis, the effect of the compounds described herein on the synaptic contacts and synapses of mouse cortical neurons was investigated.

Primary mouse cortical neurons were treated with 5 μ M test compound at DIV 15. As a control, primary mouse cortical neurons were treated with vehicle only (10% DMSO, 90% Phosphate Buffered Saline (PBS)). After 24 hours, DIV 16 neurons were fixed, immunostained using presynaptic vesicular protein synapsin (P38), counterstained with the nuclear dye DAPI (4', 6-diamidino-2-phenylindole) and counted. The immunolabeled neurons were imaged on a Leica confocal microscope (Leica confocal microscope). The number of P38 immunopositive bumps was analyzed using FIJI with Squash insert.

In a similar experiment, primary mouse cortical neurons were treated with 1 μ M test compound at DIV 15 using the same DMSO/PBS buffer as above as the control vehicle. After 24 hours, the DIV 16 neurons were fixed, immuno-labeled with synaptophysin, stained with DAPI, and counted as described above.

Example 8

In silico fascin binding

To assess the ability of the compounds described herein to bind fascin and thereby inhibit its ability to bundle actin fibrils, a computer-simulated study was conducted using the available crystal structures of the test compound and human fascin 1. Binding sites were identified on the surface of each fascin crystal structure, and test compounds were virtually docked in each pocket to determine favorable binding conformations. See international publication No. WO2019/028164(2019, 2, 7).

Analysis and preparation of fascin crystal structure

All available fascin crystal structures were downloaded from PDB and prepared for structural analysis (see Sedeh, r.s.et.j.mol.biol.400, 589-. The structure was analyzed by eye and by a standard automated protocol embedded in the ICM-Pro software of MolSoft. Hydrogen atoms are added to the structure and are considered with respect to: the correct orientation of the Asn and gin side chains, ligand and protein charge, histidine orientation and protonation state, and any crystallographic quality markers, such as high b-factor or low occupancy.

■ pocket identification

The ICMPacketFinder algorithm of MolSoft was used to identify potential ligand binding pockets and cavities in all available fascin crystal structures (see An, J., et al. genome Inform. int. Conf. genome Inform.15, 31-41 (2004); Kufareva, I., et al. nucleic Acids Res.40, D535-540 (2012)). First, search for crystalsPocket in the active chain A of the bulk Structure 3LLP, since the structure was found to have the highest resolutionThe four "drug-like" pockets are defined as having properties suitable for binding small molecules.

■ ligand docking and scoring

The head and head + tail of the test compound were docked to each of the four pockets using the ICM-Docking software by MolSoft, version 3.8-6a (Abagyan, R. & Totrov, M.J.mol.biol.235, 983-1002 (1994)). A docking score is determined for each pocket. The lower the docking score, the better the "compound-fascin binding pocket" interaction.

Pocket B at actin binding site 1 was considered to result in the lowest docking score. Binding pocket B was further studied in other fascin crystal structures. The docked head group is used as an anchor point, followed by docking of the tail group to create the final energetically favorable compound posture.

The scope of the invention is not limited by the specific embodiments disclosed in the examples, which are intended to be illustrative of several embodiments of the invention, nor is it limited by any functionally equivalent embodiments within the scope of the disclosure. Indeed, various modifications of the disclosure in addition to those shown and described herein will become apparent to those skilled in the art and are intended to fall within the scope of the appended claims. For this reason, it should be noted that one or more hydrogen atoms or methyl groups may be omitted from the drawn structures consistent with the accepted shorthand notation of such organic compounds, and their presence will be readily understood by those skilled in the art of organic chemistry.

63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:用于治疗肾脏疾病的新化合物及其药物组合物

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!