Process for the preparation of pyrazolo[1,5-a]pyrimidines and salts thereof

Information

  • Patent Grant
  • 11091486
  • Patent Number
    11,091,486
  • Date Filed
    Thursday, October 26, 2017
    6 years ago
  • Date Issued
    Tuesday, August 17, 2021
    2 years ago
Abstract
In some embodiments, provided herein are processes for preparing a compound of Formula C or a salt thereof, as disclosed herein. In some embodiments, provided herein is a compound of Formula I or a pharmaceutically acceptable salt, solvate or hydrate thereof. In some embodiments, provided herein is a solid form of the compound, such as a crystalline form of the compound crystalline Form I.
Description
FIELD OF THE INVENTION

Provided herein are processes and intermediates useful for the preparation of a compound of Formula C:




embedded image



or a salt thereof.


Provided also herein is (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one:




embedded image



or a pharmaceutically acceptable salt, solvate or hydrate thereof. The compound of Formula I is also referred to herein as “Compound 1”.


Provided also herein are crystalline forms of (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one, salt forms thereof, and crystalline forms of these salts, including methods of preparation thereof. (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one and its forms are useful in the treatment of the Trk-associated disorders such as cancer, pain, inflammation, neurodegenerative diseases and certain infectious diseases.


BACKGROUND

Trk's are high affinity receptor tyrosine kinases activated by a group of soluble growth factors called neurotrophins (NT). The Trk receptor family has three members: TrkA, TrkB and TrkC. Among the neurotrophins are (i) nerve growth factor (NGF) which activates TrkA, (ii) brain-derived neurotrophic factor (BDNF) and neurotrophin-4/5 which activate TrkB and (iii) neurotrophin-3 which activates TrkC. Inhibitors of the Trk/neurotrophin pathway have been demonstrated to be effective in numerous pre-clinical animal models of pain. Overexpression, activation, amplification and/or mutation of Trk kinases are associated with many cancers including neuroblastoma, ovarian and colorectal cancer, melanoma, head and neck cancer, gastric carcinoma, lung carcinoma, breast cancer, glioblastoma, medulloblastoma, secretory breast cancer, salivary gland cancer, papillary thyroid carcinoma, and adult myeloid leukemia. The neurotrophin/Trk pathway has been implicated in inflammatory diseases including asthma, interstitial cystitis, inflammatory bowel diseases including ulcerative colitis and Crohn's disease, and inflammatory skin diseases such as atopic dermatitis, eczema and psoriasis. The neurotrophin/Trk pathway has also been implicated in the etiology of neurodegenerative diseases including multiple sclerosis, Parkinson's disease and Alzheimer's Disease. The TrkA receptor is also involved the disease process in the parasitic infection of Trypanosoma cruzi (Chagas disease) in human hosts. As such, inhibition of Trk kinases will be useful to provide therapeutic benefit to patients suffering from the aforementioned conditions.


New forms of macrocyclic pyrazolo[1,5-a]pyrimidines can be useful in the preparation of pharmaceutical formulations and dosage forms. Furthermore, a need exists for alternative synthetic procedures for the preparation of such pyrazolo[1,5-a]pyrimidines. Such alternative synthetic procedures are provided herein.


SUMMARY

Provided herein in some embodiments is a process for preparing a compound of Formula C:




embedded image



or a salt thereof. In some embodiments, the process comprises:

    • a) treating a compound of formula C-I




embedded image



or a salt thereof, with a hydrogenation system to form a compound of formula C-II




embedded image



or a salt thereof;

    • b) treating the compound of formula C-II or a salt thereof with a first strong base to form a compound of formula C-III




embedded image



or a salt thereof; and

    • c) cyclizing the compound of formula C-III or a salt thereof with a coupling agent to form the compound of Formula C or a salt thereof;


      wherein:


ring A is selected from rings A-1 and A-3 having the structures:




embedded image



wherein the wavy line labeled 1 indicates the point of attachment of ring A to ring B and the wavy line labeled 2 indicates the point of attachment of ring A to the carbon atom of the ethylene linker in formulae C, C-II or C-III, or to the carbon atom of the alkyne linker in formula C-I;


X is N or CH;


Y is H or F;


R1 is H, (1-6C)alkyl, (1-3C)alkoxy or halogen;


ring B is selected from rings B-1 and B-2 having the structures:




embedded image



wherein the wavy line labeled 3 indicates the point of attachment to ring A and the wavy line labeled 4 indicates the point of attachment to the pyrazolo[1,5-a]pyrimidine ring;


R2 and R2a are independently H, F, (1-3 C)alkyl or OH, provided that R2 and R2a are not both OH;


m is 0, 1 or 2;


R3 and R3a are independently H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R4 is H, (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C alkyl) or dihydroxy(2-6C alkyl); and


R5 and R6 are independently H, halogen, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl.


Provided herein in some embodiments is a process for preparing a compound of Formula I




embedded image



or a salt thereof, comprising:


a) treating a compound of formula 13




embedded image



or a salt thereof with a hydrogenation system to form a compound of formula 14




embedded image



or a salt thereof;


b) treating the compound of formula 14 or a salt thereof with a first strong base to form a compound of formula 15




embedded image



or a salt thereof; and


c) cyclizing the compound of formula 15 or a salt thereof with a coupling agent to form the compound of Formula I or a salt thereof.


Provided herein in some embodiments is a process for preparing a compound of Formula II:




embedded image



or a salt thereof, comprising:


a) treating a compound of formula 16




embedded image



or a salt thereof with a hydrogenation system to form a compound of formula 17




embedded image



or a salt thereof;


b) treating the compound of formula 17 or a salt thereof with a first strong base to form a compound of formula 18




embedded image



or a salt thereof; and


c) cyclizing the compound of formula 18 or a salt thereof with a coupling agent to form the compound of Formula II or a salt thereof.


Provided herein in some embodiments is a process for preparing a compound of Formula III:




embedded image



or a salt thereof, comprising:


a) treating a compound of formula 20




embedded image



or a salt thereof with a hydrogenation system to form a compound of formula 21




embedded image



or a salt thereof;


b) treating the compound of formula 21 or a salt thereof with a first strong base to form a compound of formula 22




embedded image



or a salt thereof; and


c) cyclizing the compound of formula 22 or a salt thereof with a coupling agent to form the compound of Formula III or a salt thereof.


Provided herein in some embodiments is a compound of Formula I




embedded image



or a pharmaceutically acceptable salt, solvate or hydrate thereof.


The present disclosure in one embodiment is directed to a solid form of (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one having the following structural formula:




embedded image


The present disclosure is further directed to a crystalline form of Compound 1, such as a crystalline form of Compound 1 having Form I. In some embodiments, Form I has the XRPD peaks, in terms of 2-theta (20), at about 9.1, about 20.2 and about 24.9. In some embodiments, Form I has the XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 20.2 and about 24.9. In some embodiments, Form I has the XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 13.4, about 14.8, about 20.2, and about 29.4. In some embodiments, Form I has the XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 13.4, about 14.8, about 18.3, about 18.6, about 20.2, about 23.6, about 24.9, and about 29.4. As used herein, the term “about” in conjunction with XRPD peaks refers to a variation of ±0.2. Thus, for example, a 2-theta value of “about 9.1” means a 2-theta value of 9.1±0.2.


The present disclosure is further directed to salts of Compound 1.


The present disclosure is further directed to crystalline forms of benzenesulfonic acid salt, citric acid salt, methanesulfonic acid salt, 1,2-ethane disulfonic acid salt, p-toluene sulfonic acid salt, oxalic acid salt, fumaric acid salt, L-malic acid salt, and succinic acid salt of Compound 1.


In one aspect, the present disclosure is directed to a crystalline form of Compound 1 besylate. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 13.4, and about 21.2. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 12.0, about 13.4, and about 21.2. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 12.0, about 13.4, about 19.0, about 19.4, and about 21.2. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 12.0, about 13.4, about 19.0, about 19.4, about 19.9, about 20.1, about 21.2, about 25.5, and about 32.7.


In another aspect, the present disclosure is directed to a crystalline form of Compound 1 citrate, such as crystalline Compound 1 citrate Form A. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 20.7, about 21.6, and about 24.8. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 8.9. 20.7, about 21.6, and about 24.8. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 8.9, about 11.1, about 14.4, about 15.4, about 20.7, about 21.6, and about 24.8. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 8.9, about 11.1, about 13.9, about 14.4, about 15.4, about 19.2, about 20.7, about 21.6, about 24.8, and about 25.6.


The present disclosure is further directed to the hydrochloric acid salt, sulfuric acid salt, naphthalene-2-sulphonic acid salt, 2-hydroxy ethanesulfonic acid salt, L-aspartic acid salt, maleic acid salt, phosphoric acid salt, ethanesulfonic acid salt, L-glutamic acid salt, L-tartaric acid salt, D-glucuronic acid salt, hippuric acid salt, D-gluconic acid salt, DL-lactic acid salt, L-ascorbic acid salt, benzoic acid salt, benzenesulfonic acid salt, citric acid salt, methanesulfonic acid salt, 1,2-ethane disulfonic acid salt, p-toluene sulfonic acid salt, oxalic acid salt, fumaric acid salt, L-malic acid salt, and succinic acid salt of Compound 1.


The present disclosure is further directed to a hydrate or a solvate of Compound 1, or any one of the salts of Compound 1 described herein. In some aspects, the hydrate or the solvate is crystalline.


The present disclosure is further directed to processes for preparing any one of the crystalline forms, solid forms, solvates, hydrates, or salts described herein.


The present disclosure is further directed to pharmaceutical compositions comprising any one of the crystalline forms, solid forms, solvates, hydrates, or salts described herein, and at least one pharmaceutically acceptable carrier.


The present disclosure is further directed to therapeutic methods of using any one of the crystalline forms, solid forms, solvates, hydrates, or salts described herein.


Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present application belongs. Materials are described herein for use in the present application; other, suitable materials known in the art can also be used. The materials and examples are illustrative only and not intended to be limiting. All publications, patent applications, patents, sequences, database entries, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control.


Other features and advantages of the present application will be apparent from the following detailed description and figures, and from the claims.





DESCRIPTION OF DRAWINGS


FIG. 1 is a XRPD diffractogram of Compound 1 (Form I).



FIG. 2 is a TG/DTA thermogram of Compound 1 (Form I).



FIG. 3 is a DSC thermogram of Compound 1 (Form I).



FIG. 4 is a GVS isotherm plot of Compound 1 (Form I).



FIG. 5 is a GVS kinetic plot of Compound 1 (Form I).



FIG. 6 is a DVS isotherm plot of Compound 1 (Form I).



FIG. 7 is a DVS change in mass plot of Compound 1 (Form I).



FIG. 8 is an IR spectrum Compound 1 (Form I).



FIG. 9 is a 1H NMR spectrum of Compound 1 (Form I).



FIG. 10 is an image showing a 3-D view of Compound 1 (Form I) with atom labels.



FIG. 11 is an image showing a ORTEP view of Compound 1 (Form I) with atom labels.



FIG. 12 is an image showing a 3-D view of Compound 1, acetonitrile solvate with atom labels.



FIG. 13 is an image showing a ORTEP view of Compound 1, acetonitrile solvate with atom labels.



FIG. 14 is a XRPD diffractogram of Compound 1 edisylate.



FIG. 15 is a XRPD diffractogram of Compound 1 tosylate.



FIG. 16 is a XRPD diffractogram of Compound 1 mesylate.



FIG. 17 is a XRPD diffractogram of Compound 1 besylate (pattern 1).



FIG. 18 is a XRPD diffractogram of Compound 1 besylate (pattern 2).



FIG. 19 is a XRPD diffractogram of Compound 1 oxalate.



FIG. 20 is a XRPD diffractogram of Compound 1 fumarate.



FIG. 21 is a XRPD diffractogram of Compound 1 citrate (Form A).



FIG. 22 is a XRPD diffractogram of Compound 1 L-malate.



FIG. 23 is a XRPD diffractogram of Compound 1 succinate.



FIG. 24 is a TG/DTA thermogram of Compound 1 tosylate.



FIG. 25 is a TG/DTA thermogram of Compound 1 mesylate.



FIG. 26 is a TG/DTA thermogram of Compound 1 oxalate.



FIG. 27 is a TG/DTA thermogram of Compound 1 fumarate.



FIG. 28 is a TG/DTA thermogram of Compound 1 L-malate.



FIG. 29 is a TG/DTA thermogram of Compound 1 succinate.



FIG. 30 is a XRPD diffractogram of Compound 1 mesylate acetone solvate.



FIG. 31 is a TG/DTA thermogram of Compound 1 mesylate acetone solvate.



FIG. 32 is a DSC thermogram of Compound 1 mesylate.



FIG. 33 is a GVS isotherm of Compound 1 mesylate acetone solvate.



FIG. 34 is a GVS kinetic plot of Compound 1 mesylate acetone solvate.



FIG. 35 is an IR spectrum of Compound 1 mesylate acetone solvate.



FIG. 36 is a 1H NMR spectrum of Compound 1 mesylate acetone solvate.



FIG. 37 is a TG/DTA thermogram of Compound 1 besylate.



FIG. 38 is a DSC thermogram of Compound 1 besylate.



FIG. 39 is a DVS isotherm of Compound 1 besylate.



FIG. 40 is a DVS kinetic plot of Compound 1 besylate.



FIG. 41 is an IR spectrum of Compound 1 besylate.



FIG. 42 is 1H NMR spectrum of Compound 1 besylate.



FIG. 43 is a TG/DTA thermogram of Compound 1 citrate (Form A).



FIG. 44 is a DSC thermogram of Compound 1 citrate (Form A).



FIG. 45 is a DVS isotherm of Compound 1 citrate (Form A).



FIG. 46 is a DVS kinetic plot of Compound 1 citrate (Form A).



FIG. 47 is an IR spectrum of Compound 1 citrate (Form A).



FIG. 48 is a 1H NMR spectrum of Compound 1 citrate (Form A).



FIG. 49 is a XRPD diffractogram of a Compound 1 citrate (Form B).



FIG. 50 is a sequence listing for an exemplary wildtype TrkA polypeptide (SEQ ID NO: 1).



FIG. 51 is a sequence listing for an exemplary wildtype TrkB polypeptide (SEQ ID NO: 2).



FIG. 52 is a sequence listing for an exemplary wildtype TrkC polypeptide (SEQ ID NO: 3).





DETAILED DESCRIPTION

Definitions


Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in the art to which the invention(s) belong. All patents, patent applications, published applications and publications, databases, websites and other published materials referred to throughout the entire disclosure herein, unless noted otherwise, are incorporated by reference in their entirety. In the event that there are a plurality of definitions for terms herein, those in this section prevail. Where reference is made to a URL or other such identifier or address, it is understood that such identifiers can change and particular information on the internet can come and go, but equivalent information can be found by searching the internet. Reference thereto evidences the availability and public dissemination of such information.


As used herein, the term “alkyl” refers to a hydrocarbon chain that can be a straight chain or branched chain, containing the indicated number of carbon atoms. For example, C1-6 indicates that the group can have from 1 to 6 (inclusive) carbon atoms in it. As used herein, recitations such as “C1-6 alkyl”, “(1-6C)alkyl” or “(1-6C alkyl)” are used interchangeably herein to indicate a straight or branched chain alkyl group having from one to six carbon atoms. Examples of such alkyl groups include methyl, ethyl, iso-propyl, tert-butyl, and n-hexyl.


As used herein, “hydrogenation system” refers to a compound or complex capable of catalyzing a hydrogenation reaction, i.e., the reaction of the hydrogen with a hydrogen reactive group, such as a benzyl group or a carbon-carbon double/triple bond. The hydrogenation system includes hydrogen gas at atmospheric or higher pressure and a catalyst. Catalysts useful for hydrogenation include, but are not limited to, metals, such as palladium, platinum, and rhodium and their oxides or hydroxides, preferably supported on a material such as carbon or alumina.


“Coupling agent,” as used herein, refers to a reagent that forms amide or ester bonds, such as by coupling acids and amines or alcohols, respectively. Suitable coupling agents are well known to a person of skill in the art and are commercially available. Coupling agents include, but are not limited to, dicyclohexylcarbodiimide (DCC), N,N′-diisopropylcarbodiimide (DIC), N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide (EDCI). or carbonyl diimidazole (CDI). In some embodiments, one or more than one coupling agent can be used at the same time. A coupling agent may be used in conjunction with a catalyst.


As used herein, “strong base” refers to a basic chemical compound that is able to deprotonate weak acids in an acid-base reaction. A strong base is also able to hydrolyze an ester compound in a hydrolysis reaction to produce the corresponding carboxylic acid compound. Examples of strong bases include, but are not limited to, hydroxides, alkoxides, and ammonia. Common examples of strong bases are the hydroxides of alkali metals and alkaline earth metals, e.g., NaOH. Certain strong bases are even able to deprotonate very weakly acidic C—H groups in the absence of water. Strong bases include, but are not limited to, sodium hydroxide, potassium hydroxide, barium hydroxide, cesium hydroxide, strontium hydroxide, lithium hydroxide and rubidium hydroxide. In certain embodiments, NaOH is used as the strong base.


As used herein, the term “weak base” refers to inorganic and organic bases that are only partially ionized in aqueous solution. Weak bases typically have a pKa of between about 6 and about 11. A large number of such weak bases are known and are exemplified by those listed in the Handbook of Biochemistry and Molecular Biology, Vol. 1, 3rd ed., G. D. Fassman, CRC Press, 1976, pp. 305-347. The weak base may be soluble or insoluble in water. Suitable weak bases include, but are not limited to, alkali metal carbonates and bicarbonates, such as lithium carbonate, sodium carbonate, potassium carbonate, and sodium bicarbonate; ammonia; primary amines; secondary amines; and tertiary amines, such as the trialkylamines, e.g., triethylamine, tripropylamine and tributylamine, benzyldiethylamine, pyridine, quinoline, N-methylmorpholine, and the like.


“Non-nucleophilic base,” as used herein, refers to a base that will not act as a nucleophile, i.e., a base that will not donate an electron pair to an electrophile to form a chemical bond in relation to a reaction. Typically, non-nucleophilic bases are bulky and sterically hindered, such that protons can attach to the basic center, but alkylation and complexation are prevented. Examples of non-nucleophilic bases include, but are not limited to, amines and nitrogen heterocycles, such as triethylamine and pyridine, lithium compounds, and phsophazenes.


The terms “hydrogen” and “H” are used interchangeably herein.


The terms “halogen” or “halo” refer to fluorine (F), chlorine (Cl), bromine (Br), or iodine (I).


As used herein, the term “alkylamine” refers to an amine that contains one or more alkyl groups. An alkylamine can be a primary amine, a secondary amine or a tertiary amine. For example, a secondary alkylamine is an amine that contains two alkyl groups. An example includes diisopropylethylamine.


As used herein, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise.


As used herein, ranges and amounts can be expressed as “about” a particular value or range. About also includes the exact amount. Hence, “about 5 grams” means “about 5 grams” and also “5 grams.” It also is understood that ranges expressed herein include whole numbers within the ranges and fractions thereof. For example, a range of between 5 grams and 20 grams includes whole number values such as 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 and 20 grams, and fractions within the range including, but not limited to, 5.25, 6.5, 8.75 and 11.95 grams.


As used herein, “optional” or “optionally” means that the subsequently described event or circumstance does or does not occur and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, a reaction mixture that “optionally includes a catalyst” means that the reaction mixture contains a catalyst or it does not contain a catalyst.


The compounds disclosed herein include compounds having a sulfoxide group, as shown, by way of example, in the structure of compound 2, below:




embedded image



The sulfur-oxygen bond can also be rendered pictorially as being in ionic form. Thus, for example, compound 2 can also be rendered as shown in the structure below:




embedded image



It is intended throughout this disclosure that the recitation of a given structure for a compound having a sulfoxide group encompasses all representations of the compound, whether the sulfur-oxygen bond is rendered as being an ionic bond, a covalent bond, a dative bond, or in any form that can be envisioned by the skilled artisan.


The term “compound,” as used herein, is meant to include all stereoisomers, geometric isomers, tautomers, and isotopes of the structures depicted. Compounds herein identified by name or structure as one particular tautomeric form are intended to include other tautomeric forms unless otherwise specified. Compounds herein identified by name or structure without specifying the particular configuration of a stereocenter are meant to encompass all the possible configurations at the stereocenter. For example, if a particular stereocenter in a compound of the invention could be R or S, but the name or structure of the compound does not designate which it is, then the stereocenter can be either R or S. The compounds described herein can be asymmetric (e.g., having one or more stereocenters). Compounds of the present application that contain an asymmetrically substituted carbon atom can be isolated in optically active or racemic forms. Methods on how to prepare optically active forms from optically inactive starting materials are known in the art, such as by resolution of racemic mixtures or by stereoselective synthesis. In some embodiments, an asymmetrically substituted carbon atom has the (R)-configuration according to Cahn-Ingold-Prelog nomenclature. In some embodiments, an asymmetrically substituted carbon atom has the (S)-configuration according to Cahn-Ingold-Prelog nomenclature.


“Protecting group”, as used herein, refers to any convenient functional group that allows to obtain chemoselectivity in a subsequent chemical reaction. Protecting groups are described, for example, in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2nd ed. New York; John Wiley & Sons, Inc., 1991. For a particular compound and/or a particular chemical reaction, a person skilled in the art knows how to select and implement appropriate protecting groups and synthetic methods. Examples of amine protecting groups include acyl and alkoxycarbonyl groups, such as t-butoxycarbonyl (BOC), and [2-(trimethylsilyl)ethoxy]methyl (SEM). Examples of carboxyl protecting groups include (1-6C)alkyl groups, such as methyl, ethyl and t-butyl. Examples of alcohol protecting groups include benzyl, trityl, silyl ethers, and the like.


“Leaving group”, as used herein, refers to a molecule or a molecular fragment (e.g., an anion) that is displaced in a chemical reaction as stable species taking with it the bonding electrons. Examples of leaving groups include arylsulfonyloxy group or an alkylsulfonyloxy group, such as a mesylate or a tosylate group. Common anionic leaving groups also include halides such as Cl—, Br—, and I—.


A salt can form from a compound in any manner familiar to the skilled artisan. Accordingly, the recitation “to form a compound or salt thereof” includes embodiments where a compound is formed and the salt is subsequently formed from the compound in a manner familiar to the skilled artisan.


As used herein, the phrase “solid form” refers to Compound 1 or a salt of Compound 1 in either an amorphous state or a crystalline state (“crystalline form” or “crystalline solid”), whereby a compound in a crystalline state may optionally include solvent or water within the crystalline lattice, for example, to form a solvated or hydrated crystalline form.


The term “hydrated,” as used herein, is meant to refer to a crystalline form that includes water molecules in the crystalline lattice.


Different crystalline forms of compounds can be characterized by X-ray powder diffraction (XRPD), differential scanning calorimetry (DSC), differential thermal analysis (DTA), and/or thermogravimetric analysis (TGA). An X-ray powder diffraction (XRPD) pattern of reflections (peaks) is typically considered a fingerprint of a particular crystalline form. It is well known that the relative intensities of the XRPD peaks can widely vary depending on the sample preparation technique, crystal size distribution, various filters used, the sample mounting procedure, and the particular instrument employed. In some instances, new peaks may be observed or existing peaks may disappear depending on the type of instrument or the settings (for example, whether a Ni filter is used or not).


As used herein, the term “peak” refers to a reflection having a relative height/intensity of at least about 5% of the maximum peak height/intensity in the XPRD. Peak assignments, such as those reported herein, can vary by plus or minus 0.2° (2-theta), and the term “substantially” or “about” as used in the context of XRPD herein is meant to refer to the above-mentioned variations. Thus, for example, a 2-theta value of “about 9.1” means a 2-theta value of 9.1±0.2.


As described herein, temperature readings in connection with DSC, TGA, or other thermal experiments can vary by ±4° C. depending on the instrument, particular settings, sample preparation, etc. Accordingly, a crystalline form reported herein having a DSC thermogram “substantially” as shown in any of the Figures is understood to accommodate such variation. An endothermal or exothermic event at “about” a certain temperature is also understood to accommodate this variation.


As used herein, the term “melting point” refers to an endothermal event or endothermal event observed in, e.g., a DSC thermogram. An endothermal event is a process or reaction in which a sample absorbs energy from its surroundings in the form of e.g., heat as in a DSC experiment. An exothermic event is a process or reaction in which a sample releases energy. The process of heat absorption and release can be detected by DSC. In some embodiments, the term “melting point” is used to describe the major endothermal event on a DSC thermogram.


The term “room temperature” or “ambient temperature” as used herein, is understood in the art, and refers generally to a temperature, e.g., a reaction temperature, that is about the temperature of the room in which the reaction is carried out, for example, a temperature from about 20° C. to about 30° C.


In some embodiments, the compounds, salts, and forms described herein are substantially isolated. By “substantially isolated” is meant that the compound, salt, or form is at least partially or substantially separated from the environment in which it was formed or detected. Partial separation can include, e.g., a composition enriched in the compound, salt or form. Substantial separation can include compositions containing at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90%, at least about 95%, at least about 97%, or at least about 99% by weight of the compound, salt or form.


The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.


The phrase “therapeutically effective amount” refers to the amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue, system, animal, individual or human that is being sought by a researcher, veterinarian, medical doctor or other clinician.


As used herein, terms “treat” or “treatment” refer to therapeutic or palliative measures. Beneficial or desired clinical results include, but are not limited to, alleviation, in whole or in part, of symptoms associated with a disease or disorder or condition, diminishment of the extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state (e.g., one or more symptoms of the disease), and remission (whether partial or total), whether detectable or undetectable. “Treatment” can also mean prolonging survival as compared to expected survival if not receiving treatment.


In one embodiment, the term “preventing” as used herein means the prevention of the onset, recurrence or spread, in whole or in part, of the disease or condition as described herein (e.g., multiple types of pain including inflammatory pain, neuropathic pain, and pain associated with cancer, surgery, and bone fracture), or a symptom thereof.


The terms “effective amount” and “therapeutically effective amount” refer to an amount of compound that, when administered to a mammal in need of such treatment, is sufficient to (i) treat or prevent a particular disease, condition, or disorder, (ii) attenuate, ameliorate, or eliminate one or more symptoms of the particular disease, condition, or disorder, or (iii) prevent or delay the onset of one or more symptoms of the particular disease, condition, or disorder described herein. The amount of a Compound 1, or salt thereof, that will correspond to such an amount will vary depending upon factors such as the particular compound, disease condition and its severity, the identity (e.g., weight) of the mammal in need of treatment, but can nevertheless be routinely determined by one skilled in the art.


The terms “individual” or “patient,” used interchangeably, refer to any animal, including mammals, and most preferably humans. As used herein, the term “mammal” refers to a warm-blooded animal that has or is at risk of developing a disease described herein and includes, but is not limited to, guinea pigs, dogs, cats, rats, mice, hamsters, primates, and humans.


It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable sub-combination.


All combinations of the embodiments pertaining to the aspects described herein are specifically embraced by the present invention just as if each and every combination was individually explicitly recited, to the extent that such combinations embrace possible aspects. In addition, all sub-combinations of the embodiments contained within the aspects described herein, as well as all sub-combinations of the embodiments contained within all other aspects described herein, are also specifically embraced by the present invention just as if each and every sub-combination of all embodiments are explicitly recited herein.


Examples of Embodiments

The present application provides, inter alia, a process for preparing a compound of Formula C, or a salt thereof, as set out, for example, in Scheme 1:




embedded image


embedded image


In some embodiments, provided herein is a process for preparing a compound of Formula C




embedded image


or a salt thereof, comprising:

    • a) treating a compound of formula C-I




embedded image



or a salt thereof, with a hydrogenation system to form a compound of formula C-II




embedded image



or a salt thereof;

    • b) treating the compound of formula C-II or a salt thereof with a first strong base to form a compound of formula C-III




embedded image



or a salt thereof; and

    • c) cyclizing the compound of formula C-III or a salt thereof with a coupling agent to form the compound of Formula C or a salt thereof;


      wherein:


ring A is selected from rings A-land A-3 having the structures:




embedded image



wherein the wavy line labeled 1 indicates the point of attachment of ring A to ring B and the wavy line labeled 2 indicates the point of attachment of ring A to the carbon atom of the ethylene linker in formulae C, C-II or C-III, or to the carbon atom of the alkyne linker in formula C-I;


X is N or CH;


Y is H or F;


R1 is H, (1-3C)alkyl, (1-3C)alkoxy or halogen;


ring B is selected from rings B-1 and B-2 having the structures:




embedded image



wherein the wavy line labeled 3 indicates the point of attachment to ring A and the wavy line labeled 4 indicates the point of attachment to the pyrazolo[1,5-a]pyrimidine ring;


R2 and R2a are independently H, F, (1-3 C)alkyl or OH, provided that R2 and R2a are not both OH;


m is 0, 1 or 2;


R3 and R3a are independently H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl; R4 is H, (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C alkyl) or dihydroxy(2-6C alkyl); and


R5 and R6 are independently H, halogen, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl. In some embodiments, provided herein is a process for preparing a compound of Formula C, or a salt thereof, comprising cyclizing the compound of formula C-III, or a salt thereof, with a coupling agent to form the compound of Formula C, or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula C-III, or a salt thereof, comprising treating the compound of formula C-II, or a salt thereof, with a first strong base to form a compound of formula C-III, or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula C-II, or a salt thereof, comprising treating a compound of formula C-I, or a salt thereof, with a hydrogenation system to form a compound of formula C-II, or a salt thereof.


In some embodiments, the process for preparing a compound of formula C, or a salt thereof, further comprises preparing the compound of formula C-I




embedded image


or a salt thereof, by a process comprising:

    • a) coupling the compound of formula C-VI




embedded image


or a salt thereof;


with a compound of formula D




embedded image



or a salt thereof, in the presence of a catalyst comprising palladium and a catalyst comprising copper to form the compound of formula C-VII




embedded image



or a salt thereof; and

    • b) deprotecting the compound of formula C-VII to obtain the compound of formula C-I or a salt thereof;


      wherein:


P1 is an amino-protecting group; and


L1 is a leaving group.


In some embodiments, provided herein is a process for preparing a compound of formula C-I, or a salt thereof, comprising deprotecting the compound of formula C-VII, or a salt thereof, to form a compound of formula C-I, or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula C-VII, or a salt thereof, comprising coupling a compound of formula C-VI, or a salt thereof, with a compound of formula D or a salt thereof, in the presence of a catalyst comprising palladium and a catalyst comprising copper to form the compound of formula C-VII, or a salt thereof.


In some embodiments, the compound of formula C-VI, or salt thereof, may be prepared as set out in Scheme 1a:




embedded image


In some embodiments, provided herein is a process for preparing a compound of formula C-VI




embedded image



or a salt thereof, comprising treating the compound of formula C-V




embedded image



or a salt thereof, with a reagent comprising a group L2 to form a compound of formula C-VI, wherein L2 is a group that, when bonded to an oxygen atom, forms a leaving group L1.


For example, when the leaving group L1 is trifluoromethanesulfonate (triflate), the group L2 is trifluoromethanesulfonyl (triflyl); when the leaving group L1 is tosylate, the group L2 is tosyl; when the leaving group L1 is mesylate, the group L2 is mesyl; or when the leaving group L1 is nosylate, the group L2 is nosyl. In some more particular embodiments, the reagent comprising a group L2 has a formula L2-Hal, wherein Hal is a halogen (e.g., Cl, Br or I). In some more particular embodiments, the reagent comprising a group L2 has a formula L2-O-L2. In some embodiments, the reagent comprising a group L2 is a reagent comprising a —S(O2)LG moiety, wherein LG is a leaving group such as a halogen or a OS(O2)-alkyl or OS(O2)-aryl group. In some embodiments, the reagent comprising a group L2 is a sulfonic anhydride, such as trifluoromethanesulfonic anhydride, trifluoromethanesulfonyl chloride, tosyl chloride, mesyl chloride or nosyl chloride. In some embodiments, the reagent comprising a group L2 is a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide).


In some embodiments, provided herein is a process for preparing a compound of formula C-V




embedded image



or a salt thereof, comprising treating a compound of formula C-IV




embedded image



or a salt thereof, with a first acid to form a compound of formula C-V.


In some embodiments, the compound of Formula C has formula Ca:




embedded image



or a salt thereof;


the compound of formula C-I has formula C-Ia




embedded image



or a salt thereof;


the compound of formula C-II has formula C-IIa




embedded image



or a salt thereof; and


the compound of formula C-III has formula C-IIIa:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VI has formula C-VIa




embedded image



or a salt thereof;


the compound of formula D has formula D-1




embedded image



the compound of formula C-VII has formula C-VIIa




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-IV has formula C-IVa




embedded image



or a salt thereof; and


the compound of formula C-V has formula C-Va




embedded image



or a salt thereof;


In some embodiments, the compound of formula C has formula Cb




embedded image


In some embodiments, the compound of formula C has formula Cc




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Cd




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Ce




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Cf




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Cg




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Ch




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Ci




embedded image



or a salt thereof.


In some embodiments, the compound of formula C has formula Cj




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ib




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ic




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Id




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ie




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-If




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ig




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ih




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ii




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-I has formula C-Ij




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIb




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIc




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IId




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIe




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIf




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIg




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIh




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIi




embedded image



or a salt thereof.


In some embodiments, compound of formula C-II has formula C-IIj




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIb:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIc:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIId:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIe:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIf:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIg:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIh:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIi:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-III has formula C-IIIj:




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-IV has formula C-IVa




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-IV has formula C-IVb




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-IV has formula C-IVc




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-V has formula C-Va




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-V has formula C-Vb




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-V has formula C-Vc




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VI has formula C-VIb




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VI has formula C-VIc




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIb




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIc




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIId




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIe




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIf




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIg




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIh




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIi




embedded image



or a salt thereof.


In some embodiments, the compound of formula C-VII has formula C-VIIj




embedded image



or a salt thereof.


In some embodiments, the compound of formula D-1 has formula D-1a




embedded image


In some embodiments, the compound of formula D-1 has formula D-1b




embedded image


In some embodiments, the compound of formula D has formula D-2




embedded image


In some embodiments, the compound of formula D-2 has formula D-2a




embedded image


In some embodiments, the compound of formula D-2 has formula D-2b




embedded image


In some embodiments, the compound of formula D has formula D-3




embedded image


In some embodiments, the compound of formula D-3 has formula D-3a




embedded image


In some embodiments, the compound of formula D-3 has formula D-3b




embedded image


In some embodiments, the compound of formula D-3 has formula D-3c




embedded image


In any one of the formulae disclosed herein, ring A is selected from rings A-1 and A-3 having the structures:




embedded image



wherein in the ring of formulae A-1 or A-3 the wavy line labeled 1 indicates the point of attachment of ring A to ring B (e.g., the pyrrolidine ring), and the wavy line labeled 2 indicates the point of attachment of ring A to either i) the aliphatic chain connecting ring A to the NR4 nitrogen, ii) OH, iii) O-(1-6C) alkyl, iv) L1, or v) OL2.


In some embodiments, ring A is ring A-1 having the structure




embedded image


In some embodiments, X is CH. In other embodiments, X is N. In particular embodiments, ring A when represented by structure A-1 include the structures:




embedded image


In some embodiments, ring A is ring A-3 having the structure




embedded image


In any one of the embodiments of ring A, Y is H. In some embodiments, Y is halogen. For example, Y is Cl, F or Br. In some embodiments, Y is F. In any one of the embodiments of ring A, R1 is H. In some embodiments, R1 is (1-3C)alkyl or (1-3C)alkoxy. In one embodiment, R1 is (1-3C)alkoxy. A particular example is methoxy. In one embodiment, R1 is (1-3C)alkyl. A particular example is methyl. In some embodiments, R1 is halogen. In one embodiment, R1 is F.


In some embodiments of any one of the formulae disclosed herein, R4 is H. In some embodiments, R4 is (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C alkyl) or dihydroxy(2-6C alkyl). In one embodiments, R4 is (1-6C)alkyl. Examples include methyl, ethyl, propyl, isopropyl, butyl, and isobutyl. In one embodiment, R4 is fluoro(1-6C)alkyl. Examples include fluoromethyl and 2-fluoroethyl. In one embodiment, R4 is difluoro(1-6C)alkyl. Example include difluoromethyl and 2,2-difluoroethyl. In one embodiment, R4 is trifluoro(1-6C)alkyl. Examples include trifluoromethyl and 2,2,2-trifluoroethyl. In one embodiment, R4 is hydroxy(1-6C alkyl). Examples include hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl and 3-hydroxypropyl. In one embodiment, R4 is dihydroxy(2-6C alkyl). An example includes 2,3-dihydroxypropyl. In one embodiments, R4 is H or (1-6C)alkyl. In one embodiment, R4 is H or methyl.


In some embodiments of any one of the formulae disclosed herein, R2 and R2a are independently H, F, methyl or OH, provided that R2 and R2a are not both OH. In some embodiments, R2 and R2a are each hydrogen. In some embodiments, R2 and R2a are each fluoro. In some embodiments, R2 is hydrogen and R2a is fluoro. In some embodiments, R2 is hydrogen and R2a is OH. In some embodiments, R2 is H and R2a is (1-6C)alkyl. In some embodiments, R2 is H and R2a is methyl. In some embodiments, R2 and R2a are both (1-6 C) alkyl. In some embodiments, R2 and R2a are both methyl.


In some embodiments of any one of the formulae disclosed herein, R3 and R3a are independently H, (1-3C)alkyl or hydroxy(1-3 C)alkyl. In one embodiment, R3a is H. In one embodiment, R3 is H. In some embodiments, R3a is H and R3 is H. In one embodiment, R3a is (1-3C)alkyl. Examples include methyl, ethyl, propyl and isopropyl. In one embodiment, R3 is (1-3C)alkyl. Examples include methyl, ethyl, propyl and isopropyl. In some embodiments, R3a is H, and R3 is (1-3 C)alkyl or hydroxy(1-3 C)alkyl. In one embodiment, R3a is (1-3C)alkyl and R3 is H. In one embodiment, R3a is methyl and R3 is H. In one embodiment, R3 is hydroxy(1-3C)alkyl. Examples include hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, and 3-hydroxypropyl. In one embodiment, R3 is hydroxymethyl, 2-hydroxyethyl, 2-hydroxypropyl, or 3-hydroxypropyl and R3a is H. In some embodiments, R3a is H and R3 is (1-3 C)alkyl. In some embodiments, R3a is H and R3 is methyl. In one embodiment, R3a is (1-3C)alkyl and R3 is H. In one embodiment, R3a is methyl and R3 is H. In some embodiments, R3a and R3 are both (1-3 C)alkyl. In some embodiments, R3 and R3a are both methyl.


In some embodiments, R3 and R3a are different, and the configuration of the carbon atom to which R3 and R3a are attached is (S). In other embodiments, R3 and R3a are different, and the configuration of the carbon atom to which R3 and R3a are attached is (R). In some aspects of these embodiments, when R3a is hydrogen, R3 is other than hydrogen and the configuration of the carbon atom to which R3 is attached is (5). In other aspects of these embodiments, when R3a is hydrogen, R3 is other than hydrogen and the configuration of the carbon atom to which R3 is attached is (R).


In some embodiments of any one of the formulae disclosed herein, R5 and R6 are independently H, halogen, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl. In one embodiment, R5 and R6 are independently H, F, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl. In one embodiment, R5 is H and R6 is H, F, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl. In some embodiments, R5 and R6 are independently H, F, OH, methyl, ethyl, HOCH2— or HOCH2CH2—. In one embodiment, R5 and R6 are independently H, F, OH, (1-3C)alkyl or hydroxy(1-3C)alkyl. In one embodiment, R5 is hydrogen and R6 is H, F, OH, (1-3C)alkyl or hydroxy(1-3C)alkyl. In one embodiment, R5 and R6 are independently H, F, OH, methyl, ethyl, HOCH2— or HOCH2CH2—. In one embodiment, R5 is hydrogen and R6 is H, F, OH, methyl, ethyl, HOCH2— or HOCH2CH2—. In one embodiment, R5 and R6 are independently H, F, or methyl. In one embodiment, R5 is H and R6 is H, F, or methyl. In one embodiment, R5 is H and R6 is F. In one embodiment, R5 is H and R6 is methyl. In one embodiment, R5 and R6 are both H. In one embodiment, R5 and R6 are both F. In one embodiment, R5 and R6 are both methyl. In some embodiments, R5 is hydrogen. In some embodiments, R6 is hydrogen.


In some embodiments of formulae C, C-I, C-II, C-IV, C-V, C-VI and C-VII, ring B when represented by ring B-1 includes the structures:




embedded image



wherein the wavy line labeled 3 indicates the point of attachment to ring A and the wavy line labeled 4 indicates the point of attachment to the pyrazolo[1,5-a]pyrimidine ring.


In some embodiments of formulae C, C-I, C-II, C-IV, C-V, C-VI and C-VII, ring B is ring B-2 having the formula:




embedded image


In some embodiments of any one of the formulae disclosed herein, m is 0, 1 or 2. In one embodiment, m is 0. In one embodiment, m is 1. In one embodiment, m is 2.


In some embodiments, one or more of C-VII, C-I, C-II and C-III are isolated after forming and prior to the respective following step in the process. In some embodiments, one or more of C-VII, C-I, C-II and C-III are not isolated after forming and prior to the respective following step in the process. In some embodiments, C-VII is not isolated after forming and prior to the following step in the process. In some embodiments, C-I is not isolated after forming and prior to the following step in the process. In some embodiments, C-II is not isolated after forming and prior to the following step in the process. In some embodiments, C-III is not isolated after forming after forming and prior to the following step in the process. In some embodiments, C-III is isolated after forming and prior to the following step in the process.


In some embodiments of any one of the formulae disclosed herein, L1 is a leaving group selected from triflate, tosylate, mesylate, nosylate and a halogen. In some aspects of these embodiments, halogen is Cl, Br or I. In some embodiments, L1 is a leaving group selected from triflate, tosylate, mesylate, and a halogen. In some embodiments, L1 is triflate or mesylate. In one embodiment, L1 is triflate. In some embodiments, L1 is Cl or Br. In one embodiment, L1 is Cl.


In some embodiments of any one of the formulae disclosed herein, P1 is an amino-protecting group selected from methoxymethyl, methylthiomethyl, p-methoxybenzyloxymethyl, p-nitrobenzyloxymethyl, t-butoxymethyl, 2-methoxyethoxymethyl, 1-ethoxyethyl, allyl, p-methoxybenzyloxycarbonyl (Moz), p-nitrobenzyloxycarbonyl (PNZ), trimethylsilyl, diethylisopropylsilyl, triphenylsilyl, formyl, chloroacetyl, methanesulfonyl, tosyl, benzyl sulfonyl, methoxymethylcarbonyl, benzyloxycarbonyl, carboxybenzyl (Cbz), t-butyloxycarbonyl (BOC), 9-fluorenylmethylcarbonyl, N-phenylcarbamoyl, and 4,4′-dimethoxytrityl.


In some embodiments, P1 is an amino-protecting group selected from p-methoxybenzyloxymethyl, p-methoxybenzyloxycarbonyl, trimethylsilyl, diethylisopropylsilyl, triphenylsilyl, methanesulfonyl, tosyl, benzyloxycarbonyl, t-butyloxycarbonyl (BOC), 9-fluorenylmethylcarbonyl and 4,4′-dimethoxytrityl.


In some embodiments, P1 is an amino-protecting group selected from p-methoxybenzyloxycarbonyl, trimethylsilyl, benzyloxycarbonyl and t-butyloxycarbonyl (BOC).


In one embodiment, P1 is t-butyloxycarbonyl (BOC).


In some embodiments, in any one of the formulae disclosed herein, ring A is selected from rings A-1 and A-3 having the structures:




embedded image


wherein in the ring of formulae A-1 or A-3 the wavy line labeled 1 indicates the point of attachment of ring A to ring B (e.g., the pyrrolidine ring), and the wavy line labeled 2 indicates the point of attachment of ring A to either i) the aliphatic chain connecting ring A to the NR4 nitrogen, ii) OH, iii) O-(1-6C) alkyl, iv) L1, or v) OL2;


X is N or CH;


Y is H or F;


R1 is H, (1-6C)alkyl, (1-3C)alkoxy or halogen;


m is 0, 1 or 2;


R2 and R2aa are independently H, F, or OH, provided that R2 and R2aa are not both OH;


R3 is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R3a (when present) is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R4 is H, (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C alkyl) or dihydroxy(2-6C alkyl);


R5 and R6 are independently H, halogen, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl;


L1 (when present) is a leaving group selected from triflate, tosylate, mesylate, and a halogen; and


P1 (when present) is an amino-protecting group selected from methoxybenzyloxymethyl, p-methoxybenzyloxycarbonyl, trimethylsilyl, diethylisopropylsilyl, triphenylsilyl, methanesulfonyl, tosyl, benzyloxycarbonyl, t-butyloxycarbonyl (BOC), 9-fluorenylmethylcarbonyl and 4,4′-dimethoxytrityl.


In some embodiments, in any one of the formulae disclosed herein, ring A is ring A-1 represented by the structure




embedded image



wherein in the ring of formulae A-1 the wavy line labeled 1 indicates the point of attachment of ring A to ring B (e.g., the pyrrolidine ring), and the wavy line labeled 2 indicates the point of attachment of ring A to either i) the aliphatic chain connecting ring A to the NR4 nitrogen, ii) OH, iii) O-(1-6C) alkyl, iv) L1, or v) OL2;


X is N or CH;


Y is H or F;


R1 is H, (1-6C)alkyl, (1-3C)alkoxy or halogen;


m is 0, 1 or 2;


R2 and R2aa are independently H, F, or OH, provided that R2 and R2aa are not both OH;


R3 is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R3a (when present) is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R4 is H, (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C alkyl) or dihydroxy(2-6C alkyl);


R5 and R6 are independently H, halogen, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl;


L1 (when present) is a leaving group selected from triflate, tosylate, mesylate, and a halogen; and


P1 (when present) is an amino-protecting group selected from methoxybenzyloxymethyl, p-methoxybenzyloxycarbonyl, trimethylsilyl, diethylisopropylsilyl, triphenylsilyl, methanesulfonyl, tosyl, benzyloxycarbonyl, t-butyloxycarbonyl (BOC), 9-fluorenylmethylcarbonyl and 4,4′-dimethoxytrityl.


In some embodiments, in any one of the formulae disclosed herein, ring A is ring A-3 represented by the structure




embedded image


wherein in the ring of formulae A-3 the wavy line labeled 1 indicates the point of attachment of ring A to ring B (e.g., the pyrrolidine ring), and the wavy line labeled 2 indicates the point of attachment of ring A to either i) the aliphatic chain connecting ring A to the NR4 nitrogen, ii) OH, iii) O-(1-6C) alkyl, iv) L1, or v) OL2;


Y is H or F;


R1 is H, (1-6C)alkyl, (1-3C)alkoxy or halogen;


R2 and R2aa are independently H, F, or OH, provided that R2 and R2aa are not both OH;


R3 is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R3a (when present) is H, (1-3 C)alkyl or hydroxy(1-3 C)alkyl;


R4 is H, (1-6C)alkyl, fluoro(1-6C)alkyl, difluoro(1-6C)alkyl, trifluoro(1-6C)alkyl, hydroxy(1-6C alkyl) or dihydroxy(2-6C alkyl);


R5 and R6 are independently H, halogen, OH, (1-6C)alkyl or hydroxy(1-6C)alkyl;


L1 (when present) is a leaving group selected from triflate, tosylate, mesylate, and a halogen; and


P1 (when present) is an amino-protecting group selected from methoxybenzyloxymethyl, p-methoxybenzyloxycarbonyl, trimethylsilyl, diethylisopropylsilyl, triphenylsilyl, methanesulfonyl, tosyl, benzyloxycarbonyl, t-butyloxycarbonyl (BOC), 9-fluorenylmethylcarbonyl and 4,4′-dimethoxytrityl.


In some embodiments, in any one of the formulae disclosed herein, ring A is ring A-1 having the structure




embedded image


X is N;


Y is H or F;


R1 is H or (1-6C)alkyl;


R4 is H or (1-6C)alkyl;


m is 0;


R3a (when present) is H and R3 is H;


R5 and R6 are each independently H or (1-6C)alkyl;


L1 (when present) is a triflate leaving group; and


P1 (when present) is a t-butyloxycarbonyl (BOC) amino-protecting group.


In some embodiments, in any one of the formulae disclosed herein, ring A is ring A-1 having the structure




embedded image


X is CH or N;


Y is H or F;


R1 is H or (1-6C)alkyl;


R4 is H or (1-6C)alkyl;


m is 0;


R3a (when present) is H and R3 is (1-3 C)alkyl;


R5 and R6 are each independently H or (1-6C)alkyl;


L1 (when present) is a triflate leaving group; and


P1 (when present) is a t-butyloxycarbonyl (BOC) amino-protecting group.


In some embodiments, in any one of the formulae disclosed herein, ring A is ring A-1 having the structure




embedded image


X is CH or N;


Y is H or F;


R1 is H or (1-6C)alkyl;


R4 is H or (1-6C)alkyl;


m is 0;


R3a is present, and R3a and R3 are each (1-3 C)alkyl;


R5 and R6 are each independently H or (1-6C)alkyl;


L1 (when present) is a triflate leaving group; and


P1 (when present) is a t-butyloxycarbonyl (BOC) amino-protecting group.


The present application also provides, inter alia, a process for preparing a compound of Formula I, or a salt thereof, as set out, for example, in Scheme 2:




embedded image


embedded image


In some embodiments, provided herein is a process for preparing a compound of Formula I




embedded image



or a salt thereof, comprising:


a) treating a compound of formula 13




embedded image



or a salt thereof with a hydrogenation system to form a compound of formula 14




embedded image



or a salt thereof;


b) treating the compound of formula 14 or a salt thereof with a first strong base to form a compound of formula 15




embedded image



or a salt thereof; and


c) cyclizing the compound of formula 15 or a salt thereof with a coupling agent to form the compound of Formula I or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 14




embedded image



or a salt thereof, comprising treating a compound of formula 13




embedded image



or a salt thereof, with a hydrogenation system to form the compound of formula 14 or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 14 or a salt thereof with a first strong base to form a compound of formula 15




embedded image



or a salt thereof.


In some embodiments, the process further comprises cyclizing the compound of formula 15 or a salt thereof with a coupling agent to form a compound of Formula I




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 15




embedded image



or a salt thereof, comprising treating a compound of formula 14




embedded image



or a salt thereof, with a first strong base to form the compound of formula 15.


In some embodiments, the process further comprises cyclizing the compound of formula 15 or a salt thereof with a coupling agent to form a compound of Formula I




embedded image



or a salt thereof.


In some embodiments, the process for preparing the compound of Formula I further comprises preparing the compound of formula 13




embedded image



or a salt thereof, by a process comprising:


a) treating a compound of formula 9




embedded image



or a salt thereof with a first acid to form a compound of formula 10




embedded image



or a salt thereof;


b) treating the compound of formula 10 or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form a compound of formula 11




embedded image



or a salt thereof; and


c) coupling the compound of formula 11 or a salt thereof with a compound of formula 12




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and


d) deprotecting the product of the coupling of compound 11 and compound 12 to form the compound of formula 13 or a salt thereof.


In some embodiments step b) is replaced by a step comprising treating the compound of formula 10 or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 10




embedded image



or a salt thereof, comprising treating a compound of formula 9




embedded image



or a salt thereof with a first acid to form the compound of formula 10 or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 10 or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form a compound of formula 11




embedded image



or a salt thereof. In some embodiments, the process further comprises treating the compound of formula 10 or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, the process further comprises coupling the compound of formula 11 or a salt thereof with a compound of formula 12




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; deprotecting the product of the coupling of compound 11 and compound 12 to form the compound of formula 13




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 11




embedded image



or a salt thereof, comprising treating a compound of formula 10




embedded image



or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form the compound of formula 11 or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 11




embedded image



or a salt thereof, comprising treating a compound of formula 10




embedded image


or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, the process further comprises coupling the compound of formula 11 or a salt thereof with a compound of formula 12




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; deprotecting the product of the coupling of compound 11 and compound 12 to form the compound of formula 13




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 13




embedded image



or a salt thereof, comprising coupling a compound of formula 11




embedded image



or a salt thereof with a compound of formula 12




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 12 to form the compound of formula 13 or a salt thereof.


The present application also provides, inter alia, a process for preparing a compound of Formula II, or a salt thereof, as set out, for example, in Scheme 3:




embedded image


embedded image


In some embodiments, one or more of 10, 11, 16, 17 and 18 are isolated after forming and prior to the respective following step in the process. In some embodiments, one or more of 10, 11, 16, 17 and 18 are not isolated after forming and prior to the respective following step in the process. In some embodiments, 10 is not isolated after forming and prior to the following step in the process. In some embodiments, 11 is not isolated after forming and prior to the following step in the process. In some embodiments, 16 is not isolated after forming and prior to the following step in the process. In some embodiments, 17 is not isolated after forming and prior to the following step in the process. In some embodiments, 18 is not isolated after forming after forming and prior to the following step in the process. In some embodiments, 18 is isolated after forming after forming and prior to the following step in the process.


In some embodiments, provided herein is a process for preparing a compound of Formula II




embedded image



or a salt thereof, comprising:


a) treating a compound of formula 16




embedded image



or a salt thereof with a hydrogenation system to form a compound of formula 17




embedded image



or a salt thereof;


b) treating the compound of formula 17 or a salt thereof with a first strong base to form a compound of formula 18




embedded image



or a salt thereof; and


c) cyclizing the compound of formula 18 or a salt thereof with a coupling agent to form the compound of Formula II or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 17




embedded image



or a salt thereof, comprising treating a compound of formula 16




embedded image



or a salt thereof, with a hydrogenation system to form the compound of formula 17 or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 17 or a salt thereof with a first strong base to form a compound of formula 18




embedded image



or a salt thereof.


In some embodiments, the process further comprises cyclizing the compound of formula 18 or a salt thereof with a coupling agent to form a compound of Formula II




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 18




embedded image



or a salt thereof, comprising treating a compound of formula 17




embedded image



or a salt thereof, with a first strong base to form the compound of formula 18.


In some embodiments, the process further comprises cyclizing the compound of formula 18 or a salt thereof with a coupling agent to form a compound of Formula II




embedded image


In some embodiments, the process for preparing the compound of Formula II further comprises preparing the compound of formula 16 or a salt thereof by a process comprising:


a) treating a compound of formula 9




embedded image



or a salt thereof with a first acid to form a compound of formula 10




embedded image



or a salt thereof;


b) treating the compound of formula 10 or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form a compound of formula 11




embedded image



or a salt thereof;


c) coupling the compound of formula 11 with a compound of formula 19




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and


d) deprotecting the product of the coupling of compound 11 and compound 19 to form the compound of formula 16 or a salt thereof.


In some embodiments step b) is replaced by a step comprising treating the compound of formula 10 or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 10




embedded image



or a salt thereof, comprising treating a compound of formula 9




embedded image



or a salt thereof with a first acid to form the compound of formula 10 or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 10 or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form a compound of formula 11




embedded image



or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 10 or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, the process further comprises coupling the compound of formula 11 or a salt thereof with a compound of formula 19




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 19 to form a compound of formula 16




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 11




embedded image



or a salt thereof, comprising treating a compound of formula 10




embedded image



or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form the compound of formula 11 or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 11




embedded image



or a salt thereof, comprising treating a compound of formula 10




embedded image


or a salt thereof with sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, the process further comprises coupling the compound of formula 11 or a salt thereof with a compound of formula 19




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 19 to form a compound of formula 16




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 16




embedded image



or a salt thereof, comprising coupling a compound of formula 11




embedded image



or a salt thereof with a compound of formula 19




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 19 to form the compound of formula 16 or a salt thereof.


The present application also provides, inter alia, a process for preparing a compound of Formula III, or a salt thereof, as set out, for example, in Scheme 4:




embedded image


embedded image


In some embodiments, provided herein is a process for preparing a compound of formula III




embedded image



or a salt thereof, comprising:


a) treating a compound of formula 20




embedded image



or a salt thereof with a hydrogenation system to form a compound of formula 21




embedded image



or a salt thereof;


b) treating the compound of formula 21 or a salt thereof with a first strong base to form a compound of formula 22




embedded image



or a salt thereof; and


c) cyclizing the compound of formula 22 or a salt thereof with a coupling agent to form the compound of Formula III or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 22




embedded image



or a salt thereof, comprising treating a compound of formula 20




embedded image



or a salt thereof, with a hydrogenation system to form the compound of formula 21 or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 21 or a salt thereof with a first strong base to form a compound of formula 22




embedded image



or a salt thereof.


In some embodiments, the process further comprises cyclizing the compound of formula 22 or a salt thereof with a coupling agent to form a compound of Formula III




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 22




embedded image



or a salt thereof, comprising treating a compound of formula 21




embedded image



or a salt thereof, with a first strong base to form the compound of formula 22.


In some embodiments, the process further comprises cyclizing the compound of formula 22 or a salt thereof with a coupling agent to form a compound of Formula III




embedded image


In some embodiments, the process for preparing the compound of Formula III further comprises preparing the compound of formula 20 or a salt thereof by a process comprising:


a) treating a compound of formula 9




embedded image



or a salt thereof with a first acid to form a compound of formula 10




embedded image



or a salt thereof;


b) treating the compound of formula 10 or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form a compound of formula 11




embedded image



or a salt thereof;


c) coupling the compound of formula 11 with a compound of formula 23




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and


d) deprotecting the product of the coupling of compound 11 and compound 23 to form the compound of formula 20 or a salt thereof.


In some embodiments step b) is replaced by a step comprising treating the compound of formula 10 or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 10




embedded image



or a salt thereof, comprising treating a compound of formula 9




embedded image



or a salt thereof with a first acid to form the compound of formula 10 or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 10 or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form a compound of formula 11




embedded image



or a salt thereof.


In some embodiments, the process further comprises treating the compound of formula 10 or a salt thereof with a sulfonimide, such as N-Phenyl-bis(trifluoromethanesulfonimide), to form a compound of formula 11 or a salt thereof.


In some embodiments, the process further comprises coupling the compound of formula 11 or a salt thereof with a compound of formula 23




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 23 to form a compound of formula 20




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 11




embedded image



or a salt thereof, comprising treating a compound of formula 10




embedded image



or a salt thereof with a reagent comprising a —S(O2)LG moiety, such as a sulfonic anhydride, such as trifluoromethanesulfonic anhydride to form the compound of formula 11 or a salt thereof.


In some embodiments, the process further comprises coupling the compound of formula 11 or a salt thereof with a compound of formula 23




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 23 to form a compound of formula 20




embedded image



or a salt thereof.


In some embodiments, provided herein is a process for preparing a compound of formula 20




embedded image



or a salt thereof, comprising coupling a compound of formula 11




embedded image



or a salt thereof with a compound of formula 23




embedded image



in the presence of a catalyst comprising palladium and a catalyst comprising copper; and deprotecting the product of the coupling of compound 11 and compound 23 to form the compound of formula 20 or a salt thereof.


In some embodiments, provided herein is a process for the preparation of the compound of formula C, wherein the process does not comprise a chromatographic purification step following the formation of the compound of formula C.


In some embodiments, provided herein is a process for the preparation of the compound of Formula I, wherein the process does not comprise a chromatographic purification step following the formation of the compound of Formula I.


In some embodiments, provided herein is a process for the preparation of the compound of Formula II, wherein the process does not comprise a chromatographic purification step following the formation of the compound of Formula II.


In some embodiments, provided herein is a process for the preparation of the compound of Formula III, wherein the process does not comprise a chromatographic purification step following the formation of the compound of Formula III.


In some embodiments, provided herein is a compound of formula C-I




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula C-II




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula C-III




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 13




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 14




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 15




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 16




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 17




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 18




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 20




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 21




embedded image



or a salt thereof.


In some embodiments, provided herein is a compound of formula 22




embedded image



or a salt thereof.


In some embodiments, the following compounds are prepared in a manner analogous to the ones disclosed above for compounds of Formulae I-III using suitable starting materials:




embedded image


embedded image


In some embodiments, the following compounds of formulae XVa-XVp are prepared in a manner analogous to the ones disclosed above for compounds of Formulae I-III using suitable starting materials:




embedded image


embedded image


embedded image


In some embodiments, the following compounds of formulae XVIa-XVIp are prepared in a manner analogous to the ones disclosed above for compounds of Formulae I-III using suitable starting materials:




embedded image


embedded image


embedded image


In some embodiments, the following compounds of formulae XVIIa-XVIIp are prepared in a manner analogous to the ones disclosed above for compounds of Formulae I-III using suitable starting materials:




embedded image


embedded image


embedded image


Parameters of Processes for Preparation of any one of Formulae Disclosed herein


In some embodiments of any one the processes provided herein, the hydrogenation system comprises hydrogen (H2) and a catalyst that comprises a metal. In some embodiments, the catalyst comprises a metal selected from gold, ruthenium, sodium, indium, nickel, palladium, and platinum. In some embodiments, the catalyst is selected from gold, ruthenium, sodium sulfide, indium, nickel, palladium, and platinum. In some embodiments, the catalyst comprises a metal selected from nickel, palladium, and platinum. In some embodiments, the metal is palladium. In some embodiments, the catalyst is palladium on carbon (Pd/C). In some embodiments, the hydrogenation system comprises hydrogen (H2) and palladium on carbon (Pd/C).


In some embodiments of any one of the processes provided herein, the first strong base is selected from sodium hydroxide, lithium hydroxide, potassium hydroxide and calcium hydroxide. In some embodiments, the first strong base is sodium hydroxide.


In some embodiments of any one of the processes provided herein, the cyclizing is performed with a coupling agent that comprises one or more of a carbodiimide, an additive, a phosphonium reagent, an aminium/uranium-imonium reagent, and miscellaneous reagents. In some embodiments, the carbodiimide is selected from dicyclohexylcarbodiimide (DCC), diisopropylcarbodiimide (DIC), and N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide (EDCI). In some embodiments, the carbodiimide is dicyclohexylcarbodiimide (DCC). In some embodiments, the carbodiimide is diisopropylcarbodiimide (DIC). In some embodiments, the carbodiimide is N-(3-dimethylaminopropyl)-N′-ethylcarbodiimide (EDCI). In some embodiments, the additive is selected from 1-hydroxybenzotriazole (HOBt), hydroxy-3,4-dihydro-4-oxo-1,2,3-benzotriazine (HOOBt), N-hydroxysuccinimide (HOSu), 1-hydroxy-7-aza-1H-benzotriazole (HOAt), 4-(N,N-dimethylamino)pyridine (DMAP), and ethyl-2-cyano-2-(hydroxyimino)acetate. In some embodiments, the phosphonium reagent is selected from benzotriazol-1-yloxy-tris(dimethylamino)-phosphonium hexafluorophosphate (BOP), benzotriazol-1-yloxy-tripyrrolidino-phosphonium hexafluorophosphate (PyBOP), bromo-tripyrrolidino-phosphonium hexafluorophosphate (PYBrOP), 7-aza-benzotriazol-1-yloxy-tripyrrolidino-phosphonium hexafluorophosphate (PyAOP), ethyl cyano(hydroxyimino)acetate-O2-tri-(1-pyrrolidinyl)-phosphonium hexafluorophosphate (PyOxim), and 3-(diethoxy-phosphoryloxy)-1,2,3-benzo[d]triazin-4(3H)-one (DEPBT). In some embodiments, the aminium/uranium-imonium reagent is selected from 2-(1H-benzotriazol-1-yl)-N,N,N′,N′-tetramethylaminium tetrafluoroborate (TBTU), 2-(1H-benzotriazol-1-yl)-N,N,N′,N′-tetramethylaminium hexafluorophosphate (HBTU), 2-(6-chloro-1H-benzotriazol-1-yl)-N,N,N′N′-tetramethylaminium hexafluorophosphate (HCTU), N-[(5-chloro-1H-benzotriazol-1-yl)-dimethylamino-morpholino]-uronium hexafluorophosphate N-oxide (HDMC), 2-(7-aza-1H-benzotriazol-1-yl)-N,N,N′N′-tetramethylaminium hexafluorophosphate (HATU), 1-[1-(cyano-2-ethoxy-2-oxoethylideneaminooxy)-dimethylamino-morpholino]-uronium hexafluorophosphate (COMU), 2-(1-oxy-pyridin-2-yl)-1,1,3,3-tetramethylisothiouronium tetrafluoroborate (TOTT), and tetramethylfluoroformamidinium hexafluorophosphate (TFFH). In some embodiments, the miscellaneous reagent is selected from N-ethoxycarbonyl-2-ethoxy-1,2-dihydroquinoiline (EEDQ), 2-propanephosphonic acid anhydride (T3P), 4-(4,6-dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium (DMTMM) salts, bis-trichloromethylcarbonate (BTC; phosgene), and 1,1′-carbonyldiimidazole (CTI). In some embodiments, the coupling agent comprises EDCI and DMAP.


In some embodiments of any one of the processes provided herein, the first acid is selected from sulfuric acid and hydrochloric acid. In some embodiments, the first acid is hydrochloric acid (HCl).


In some embodiments of any one of the processes provided herein, treatment of a compound with a first acid is conducted in the presence of a solvent. In some embodiments, the solvent is a non-protic solvent. In some embodiments, the solvent is dioxane. In some embodiments, the solvent is a protic solvent. In some embodiments, the solvent is an aliphatic alcohol.


In some embodiments, the deprotecting, or removing the protecting group P1 is conducted using any convenient synthetic method known in the art. The chemistry of protecting groups is described, for example, in in Greene & Wuts, eds., “Protecting Groups in Organic Synthesis”, 2nd ed. New York; John Wiley & Sons, Inc., 1991. For example, when the protecting group P1 is a t-butoxycarbonyl (BOC), the deprotecting is carried out using an acid such as trifluoroacetic acid or hydrochloric acid. In some embodiments, the deprotecting is conducted at room temperature.


In some embodiments, the catalyst comprising palladium is a zerovalent palladium complex selected from Pd(PPh3)2Cl2, Pd(dppe)Cl, Pd(dppp)Cl2, and Pd(dppf)Cl2. In some embodiments, the catalyst comprising palladium is Pd(PPh3)2Cl2.


In some embodiments, the catalyst comprising copper is a halide salt of copper(I) selected from copper iodide, copper bromide, and copper chloride. In some embodiments, the catalyst comprising copper is copper iodide (CuI).


In some embodiments, the catalyst comprising palladium is Pd(PPh3)2Cl2 and the catalyst comprising copper is CuI.


In some embodiments, in any one of the processes provided herein the coupling is performed in the presence of a base. In some embodiments, the base is an alkylamine. In some embodiments, the alkylamine is diisopropylamine (DIA).


In some embodiments, in any one of the processes provided herein the coupling is followed by treating with a second acid. In some embodiments, the second acid is selected from hydrochloric acid, trifluoroacetic acid and sulfuric acid. In some embodiments, the second acid is sulfuric acid (H2SO4).


Compound 12 (CAS 118080-82-3), Compound 19 (tert-butyl prop-2-ynylcarbamate, or N-Boc-propargylamine, CAS Registry No. 92136-39-5), and compound 23 (tert-butyl 2-methylbut-3-yn-2-ylcarbamate, N-boc-2-amino-2-methylbut-3-yne, CAS Registry No. 113486-06-9) are commercially available from numerous commercial suppliers. Similarly to 12, 19 and 23, suitably substituted alkynes that may be used in the preparation of any of formulae IV-XVIII above are commercially available and/or readily prepared using routine procedures.


The present application also provides a method of preparing a compound 9 as set out, for example, in Scheme 6




embedded image


In some embodiments, the process for preparing the compound of any one of Formulae I, II or III further comprises preparing the compound of formula 9




embedded image



or a salt thereof, by a process comprising reacting a compound of formula 7




embedded image



or a salt thereof, with a compound of formula 8




embedded image



or a salt thereof, in the presence of a second non-nucleophilic base, to form a compound of formula 9.


In some embodiments, the process for preparing the compound of any one of Formulae I, II or III further comprises preparing the compound of formula 8




embedded image



or a salt thereof, by a process comprising reacting




embedded image



or a salt thereof with POCl3 in an organic solvent to form a compound of formula 8. In some embodiments, the organic solvent is acetonitrile.


In some embodiments, the process for preparing the compound of any one of Formulae I, II or III further comprises preparing




embedded image



or a salt thereof, by a process comprising reacting




embedded image



with




embedded image



in the presence of a phosphate salt to form




embedded image



In some embodiments, the phosphate salt is potassium phosphate.


In some embodiments, the second non-nucleophilic base is selected from triethylamine and diisopropylethylamine. In some embodiments, the second non-nucleophilic base is triethylamine.


In some embodiments, the process for preparing the compound of any one of Formulae I, II or III further comprises preparing the compound of formula 7




embedded image



or a salt thereof, by a process comprising:


a) treating a compound of formula 1




embedded image



or a salt thereof, with (R)-2-methylpropane-2-sulfinamide to form a compound of formula 2




embedded image



or a salt thereof;


b) reacting the compound of formula 2 or a salt thereof with a compound of formula 4




embedded image



to form a compound of formula 5




embedded image



or a salt thereof;


c) treating the compound of formula 5 with a first mixture comprising an acid such as trifluoroacetic acid and water to form a second mixture; and


d) treating the second mixture with a reducing agent, to form the compound of formula 7 or a salt thereof.


In some embodiments, the compound of formula 1 or a salt thereof, is treated with (R)-2-methylpropane-2-sulfinamide in the presence of an activating agent. In some embodiments, the activating agent is selected from cesium carbonate, CuSO4, Ti(OEt)4, other Ti(IV) compounds, sodium carbonate and lithium carbonate. In some embodiments, the activating agent is selected from cesium carbonate, CuSO4, Ti(OEt)4, and other Ti(IV) compounds. In some embodiments, the activating agent is cesium carbonate.


In some embodiments, the reducing agent is a silane. In some embodiments, the reducing agent is triethylsilane.


In some embodiments, the compound of formula 4




embedded image



is prepared by a process comprising treating a compound of formula 3




embedded image



with magnesium.


In some embodiments, the first mixture comprising an acid such as trifluoroacetic acid and water comprises 4:1 trifluoroacetic acid:water.


In some embodiments, the second mixture comprises a compound of formula 6




embedded image


In some embodiments, the process comprises isolating the compound of formula 6 from the second mixture.


In some embodiments, the compound of formula 7 or a salt thereof is prepared by a process comprising:


a) treating a compound of formula 1 or a salt thereof with (R)-2-methylpropane-2-sulfinamide to form a compound of formula 2 or a salt thereof;


b) reacting the compound of formula 2 or a salt thereof with a compound of formula 4 to form a compound of formula 5 or a salt thereof;


c) treating the compound of formula 5 with a first mixture comprising an acid such as trifluoroacetic acid and water to form a compound of formula 6;


d) isolating the compound of formula 6; and


e) treating the compound of formula 6 with triethylsilane, to form the compound of formula 7 or a salt thereof.


In some embodiments, the compound of formula 1 or a salt thereof, is treated with (R)-2-methylpropane-2-sulfinamide in the presence of an activating agent. In some embodiments, the activating agent is selected from cesium carbonate, CuSO4, Ti(OEt)4, other Ti(IV) compounds, sodium carbonate and lithium carbonate. In some embodiments, the activating agent is selected from cesium carbonate, CuSO4, Ti(OEt)4, and other Ti(IV) compounds. In some embodiments, the activating agent is cesium carbonate.


In some embodiments, the compound of formula 4 is prepared by a process comprising treating a compound of formula 3 with magnesium.


In some embodiments, the first mixture comprising an acid such as trifluoroacetic acid and water comprises 4:1 trifluoroacetic acid:water.


Provided herein in some embodiments is a compound of Formula I




embedded image



or a pharmaceutically acceptable salt, solvate or hydrate thereof.


Provided herein in some embodiments is a pharmaceutical composition comprising a compound of Formula I or a pharmaceutically acceptable salt, solvate or hydrate thereof, and a pharmaceutically acceptable carrier.


Provided herein in some embodiments is a method of treating a disease in which one or more Trk kinases (e.g., TrkA, TrkB, and/or TrkC) is activated, e.g., by a soluble growth factor such as a neurotrophin (NT), such as a disease disclosed herein, comprising administering to a subject an effective amount of the compound of Formula I or a pharmaceutically acceptable salt, solvate or hydrate thereof.


In some embodiments of the compound of Formula I, a pharmaceutical composition comprising the compound of Formula I, or a method of treating a disease comprising administering to a subject an effective amount of the compound of Formula I, the compound of Formula I is present in a diastereomeric excess (d.e.) of at least 80% relative to the diastereomeric compound of formula I′:




embedded image


In some embodiments, the compound of Formula I is present in a d.e. of at least 90% relative to the compound of formula I′. In some embodiments, the compound of Formula I is present in a d.e. of at least 92% relative to the compound of formula I′. In some embodiments, the compound of Formula I is present in a d.e. of at least 94% relative to the compound of formula I′. In some embodiments, the compound of Formula I is present in a d.e. of at least 96% relative to the compound of formula I′. In some embodiments, the compound of Formula I is present in a d.e. of at least 98% relative to the compound of formula I′.


In some embodiments, the compound of Formula I is prepared from a mixture of the compound of Formula I and the compound of formula I′ by separating the two compounds. In some embodiments, the two compounds are separated by chromatography.


Table 54 provides exemplary properties of the compound of Formula I and of formula I′:











TABLE 54





Compound
formula I′
Formula I

















Aqueous solubility, pH 6.5 buffer (μg/mL)
43
111


Human Microsome Clearance (% ER)
92
79


Rat Microsome Clearance (% ER)
77
51


Human Hepatocyte Clearance (% ER)
74
48


Rat Hepatocyte Clearance (% ER)
83
64


MDRI efflux ratio
5
14


hERG IC50 (μM)
9
27









Referring to the Table 54, the solubility measurement protocol is disclosed in Example 33 hereinbelow. The Microsome and hepatocyte clearance measurement protocol is disclosed in Example 34 hereinbelow. The MDR1 efflux protocol is disclosed in Example 35 hereinbelow. The hERG IC50 values were measured using the ChanTest Fast Patch assay, available from Charles River Laboratories International, Inc. (http://www.criver.com/products-services/drug-discovery/cap abilities/ion-channel/selectivity-profiling).


In one general aspect, the present disclosure is directed to forms of (6R,15R)-9-fluoro-15-methyl-2, 11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Compound 1), the structure of which is shown below:




embedded image


Compound 1 is an inhibitor of Trk kinase useful in the treatment of diseases in which one or more Trk kinases (e.g., TrkA, TrkB, and/or TrkC) is activated, e.g., by a soluble growth factor such as a neurotrophin (NT). Compound 1 may be referred to herein as “Compound 1 free base”. In some embodiments, Compound 1 provided herein is a solid form. In some embodiments, the solid form is crystalline (e.g., Form I). In another general aspect, the present disclosure is directed to salts of Compound 1. In some embodiments, the salt of the present disclosure is a benzenesulfonic acid salt of Compound 1, which is referred to herein as “Compound 1 besylate”. In some embodiments, Compound 1 besylate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a citric acid salt of Compound 1, which is referred to herein as “Compound 1 citrate”. In some embodiments, the Compound 1 citrate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a methanesulfonic acid salt of Compound 1, which is referred to herein as “Compound 1 mesylate”. In some embodiments, the Compound 1 mesylate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a 1,2-ethane disulfonic acid salt of Compound 1, which is referred to herein as “Compound 1 edisylate”. In some embodiments, the Compound 1 edisylate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a p-toluene sulfonic acid salt of Compound 1, which is referred to herein as “Compound 1 tosylate”. In some embodiments, the Compound 1 tosylate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is an oxalic acid salt of Compound 1, which is referred to herein as “Compound 1 oxalate”. In some embodiments, the Compound 1 oxalate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a fumaric acid salt of Compound 1, which is referred to herein as “Compound 1 fumarate”. In some embodiments, the Compound 1 fumarate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a L-malic acid salt of Compound 1, which is referred to herein as “Compound 1 L-malate”. In some embodiments, the Compound 1 L-malate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a succinic acid salt of Compound 1, which is referred to herein as “Compound 1 succinate”. In some embodiments, the Compound 1 succinate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a hydrochloric acid salt of Compound 1, which is referred to herein as “Compound 1 hydrochloride”. In some embodiments, the Compound 1 hydrochloride has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a sulfuric acid salt of Compound 1, which is referred to herein as “Compound 1 sulfate”. In some embodiments, the Compound 1 sulfate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a naphthalene-2-sulphonic acid salt of Compound 1, which is referred to herein as “Compound 1 2-naphthalenesulfonate”. In some embodiments, the Compound 1 2-naphthalenesulfonate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a 2-hydroxy ethanesulfonic acid salt of Compound 1, which is referred to herein as “Compound 1 isethionate”. In some embodiments, the Compound 1 isethionate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a L-aspartic salt of Compound 1, which is referred to herein as “Compound 1 L-aspartate”. In some embodiments, the Compound 1 L-aspartate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a maleic acid salt of Compound 1, which is referred to herein as “Compound 1 maleate”. In some embodiments, the Compound 1 maleate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a phosphoric acid salt of Compound 1, which is referred to herein as “Compound 1 phosphate”. In some embodiments, the Compound 1 phosphate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a ethanesulfonic acid salt of Compound 1, which is referred to herein as “Compound 1 esylate”. In some embodiments, the Compound 1 esylate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a L-glutamic acid salt of Compound 1, which is referred to herein as “Compound 1 L-glutamate”. In some embodiments, the Compound 1 L-glutamate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a L-tartaric acid salt of Compound 1, which is referred to herein as “Compound 1 L-tartrate”. In some embodiments, the Compound 1 L-tartrate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a D-glucuronic acid salt of Compound 1, which is referred to herein as “Compound 1 D-glucuronate”. In some embodiments, the Compound 1 D-glucuronate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a hippuric acid salt of Compound 1, which is referred to herein as “Compound 1 hippurate”. In some embodiments, the Compound 1 hippurate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a D-gluconic acid salt of Compound 1, which is referred to herein as “Compound 1 D-gluconate”. In some embodiments, the Compound 1 D-gluconate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a DL-lactic acid salt of Compound 1, which is referred to herein as “Compound 1 lactate”. In some embodiments, the Compound 1 lactate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a L-ascorbic acid salt of Compound 1, which is referred to herein as “Compound 1 L-ascorbate”. In some embodiments, the Compound 1 L-ascorbate has the following structure:




embedded image


In some embodiments, the salt of the present disclosure is a benzoic acid salt of Compound 1, which is referred to herein as “Compound 1 benzoate”. In some embodiments, the Compound 1 benzoate has the following structure:




embedded image


The salts of the present application can be isolated as one or more solid forms. The solid forms, crystalline forms, solvated forms, hydrated forms of the Compound 1 and the salts of Compound 1 are described below, along with the methods of making the same and using the same for therapeutic purposes.


Compound 1 Free Base


In some embodiments, Compound 1 is in the form of the crystalline free base. In some embodiments, Compound 1 is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% crystalline.


In some embodiments, Form I is substantially free of other forms of Compound 1. In some embodiments, Form I contains less than 10%, such as less than 5%, such as less than 3%, such as less than 1% of other forms of Compound 1. In some embodiments, Form I is substantially free of the amorphous form of Compound 1. In some embodiments, Form I contains less than 10%, such as less than 5%, such as less than 3%, such as less than 1%, of the amorphous form of Compound 1.


In some embodiments, Form I is substantially free of other stereoisomers of Compound 1. In some embodiments, Form I contains less than 10%, such as less than 5%, such as less than 3%, such as less than 1% of other stereoisomers of Compound 1. In some embodiments, Form I has an XRPD pattern substantially as depicted in FIG. 1. In some embodiments, Form I has a XRPD peak, in terms of 2-theta, at about 20.2 degrees. In some embodiments, Form I has XRPD peaks, in terms of 2-theta, at about 9.1, about 20.2 and about 24.9. In some embodiments, Form I has XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 20.2 and about 24.9. In some embodiments, Form I has XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 13.4, about 14.8, about 20.2, and about 29.4. In some embodiments, Form I has XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 13.4, about 14.8, about 18.3, about 18.6, about 20.2, about 23.6, about 24.9, and about 29.4.


In some embodiments, Form I has at least one, at least two or at least three XRPD peaks, in terms of 2-theta, selected from about 9.1, about 11.2, about 13.4, about 20.2, and about 24.9 degrees. In some embodiments, Form I has at least one, at least two or at least three XRPD peaks, in terms of 2-theta, selected from about 9.1, about 11.2, about 13.4, about 14.8, about 16.8, about 18.3, about 18.6, about 20.2, about 21.4, about 22.7, about 23.6, about 24.9, and about 29.4. In some embodiments, Form I has at least one, at least two or at least three XRPD peaks, in terms of 2-theta, selected from about 9.1, about 11.2, about 13.4, about 14.8, about 18.3, about 18.6, about 20.2, about 23.6, about 24.9, and about 29.4. In some embodiments, Form I has at least one, at least two or at least three XRPD peaks, in terms of 2-theta, selected from about 9.1, about 11.2, about 13.4, about 14.8, about 20.2, and about 29.4.


In some embodiments, Form I has a DTA thermogram substantially as depicted in FIG. 2. In some embodiments, Form I has a DTA thermogram characterized by an endothermal event at about 317° C. In some embodiments, Form I has a DSC thermogram substantially as depicted in FIG. 3. In some embodiments, Form I has a DSC thermogram characterized by an endothermal event at about 317° C. In some aspects of the aforementioned embodiments, the endothermal event is a melting point. In some embodiments, Form I has a DSC thermogram characterized by an endothermal event at about 124° C. (e.g., at the second heating cycle). In some aspects of these embodiments, the endothermal event at about 124° C. is a glass transition temperature. Form I of Compound 1 is substantially anhydrous (Form I is not hydrated) and is substantially free of organic solvents (Form I is not solvated).


In some embodiments, Form I has hygroscopicity characterized by a mass uptake of about 0.3% at 90% RH as determined by GVS analysis. In other embodiments, Form I has hygroscopicity characterized by a mass uptake of about 0.7% at 90% RH as determined by DVS analysis. In some embodiments, Form I is substantially pure (e.g., the purity of the compound is at least about 90 wt. %, about 95 wt. %, about 98 wt. %, or about 99 wt. %). Purity values indicate the percentage of the amount of sample that is Form I. Purity values can be determined, for example, by HPLC/UV methods. In some embodiments, Form I is substantially free of impurities, such as organic impurities (e.g., process intermediates), inorganic impurities, and/or residual solvents.


In some embodiments, the crystalline form of Compound 1 exhibits the following single crystal X-ray crystallographic parameters at 120K:


















Crystal system
orthorhombic



Space group
P212121



a/Å
6.91792(3)



b/Å
13.74742(3)



c/Å
19.22580(5)



α/°
90.00



β/°
90.00



γ/°
90.00



Volume/Å3
1828.442(10)



Z, Z′
4



ρcalc g/cm3
1.382










In some embodiments, the crystalline form of Compound 1 is substantially as shown in FIGS. 10 and 11.


In some embodiments, Compound 1 forms a solvate with acetonitrile solvent. In some embodiments, the acetonitrile solvate of Compound 1 is crystalline. In some embodiments, the crystalline form of acetonitrile solvate of Compound 1 exhibits the following single crystal X-ray crystallographic parameters at 120K:


















Crystal system
orthorhombic



Space group
P212121



a/Å
6.03307(4)



b/Å
16.10794(9)



c/Å
23.72624(13)



α/°
90.00



β/°
90.00



γ/°
90.00



Volume /Å3
2305.73(2)



Z, Z′
4



ρcalc g/cm3
1.332










In some embodiments, the crystalline form of acetonitrile solvate is substantially as shown in FIGS. 12 and 13. In some embodiments, the crystalline form of acetonitrile solvate readily desolvates at room temperature to yield the crystalline Form I of Compound 1.


In some embodiments, the present disclosure provides crystalline Form I of Compound 1 prepared as disclosed herein. In one example, the disclosure provides the Form I of Compound 1 prepared by precipitating the solid crystalline form of Compound 1 from a saturated solution of Compound 1 in 1-propanol at about 2° C.


Compound 1 Benzenesulfonic Acid Salt


In some embodiments, provided herein is Compound 1 besylate. In some embodiments, Compound 1 besylate is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% crystalline solid. In some embodiments, crystalline Compound 1 besylate is substantially free of other forms of Compound 1 besylate. In some embodiments, crystalline Compound 1 besylate contains less than 10%, such as less than 5%, such as less than 3% of other forms of Compound 1 besylate. In some embodiments, the crystalline Compound 1 besylate is substantially free of the amorphous form of Compound 1 besylate. In some embodiments, the crystalline Compound 1 besylate contains less than 10%, less than 5%, or less than 3% of the amorphous form of Compound 1 besylate.


In some embodiments, the molar ratio of Compound 1 to the benzenesulfonic acid in the besylate is about 1:1. In some embodiments, Compound 1 besylate is a monobesylate.


In some embodiments, the crystalline Compound 1 besylate has an XRPD pattern substantially as depicted in FIG. 17. In other embodiments, the crystalline Compound 1 besylate has an XRPD pattern substantially as depicted in FIG. 18.


In some embodiments, the crystalline Compound 1 besylate has a XRPD peak, in terms of 2-theta, at about 8.1 degrees. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 13.4, and about 21.2. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 12.0, about 13.4, about 19.0, about 19.4, and about 21.2. In some embodiments, the crystalline Compound 1 besylate has XRPD peaks, in terms of 2-theta, at about 8.1, about 12.0, about 13.4, about 19.0, about 19.4, about 19.9, about 20.1, about 21.2, about 25.5, and about 32.7.


In some embodiments, the crystalline Compound 1 besylate has a XRPD peak, in terms of 2-theta, at about 8.1, about 13.4, or about 21.2. In some embodiments, the crystalline Compound 1 besylate has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 8.1, about 9.2, about 12.0, about 13.4, about 19.0, about 19.4, about 19.9, about 20.1, about 21.2, about 25.5, about 27.0, about 32.0, and about 32.7. In some embodiments, the crystalline Compound 1 besylate has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 8.1, about 12.0, about 13.4, about 19.0, about 19.4, and about 21.2. In some embodiments, the crystalline Compound 1 besylate has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 8.1, about 12.0, about 13.4, about 19.0, about 19.4, about 19.9, about 20.1, about 21.2, about 25.5, and about 32.7.


In some embodiments, the crystalline Compound 1 besylate has a DTA thermogram substantially as depicted in FIG. 37. In some embodiments, the crystalline Compound 1 besylate has a DTA thermogram characterized by an endothermal event at about 248° C. In some aspects of these embodiments, the endothermal event is a melting point. In some embodiments, the crystalline Compound 1 besylate has a DSC thermogram substantially as depicted in FIG. 38. In some embodiments, the crystalline Compound 1 besylate has a DSC thermogram characterized by an endothermal event at about 249° C.


In some embodiments, the crystalline Compound 1 besylate has hygroscopicity characterized by a mass uptake of about 0.7% at 90% RH as determined by DVS analysis. The crystalline Compound 1 besylate is substantially anhydrous (the crystalline form of the besylate is not hydrated) and is substantially free of organic solvents (the crystalline form of the besylate is not solvated).


In some embodiments, the crystalline Compound 1 besylate is substantially pure (e.g., free of organic, inorganic or other impurities). In some embodiments, the purity of the crystalline Compound 1 besylate is 90 wt. % or more, 95 wt. % or more, or 99 wt. % or more. In some embodiments, the crystalline Compound 1 besylate is substantially free of other crystalline forms of Compound 1 besylate.


In some embodiments, the benzenesulfonic acid salt of Compound 1 may form a hydrate. In some aspects of these embodiments, the hydrate is crystalline.


In some embodiments, the present disclosure provides a crystalline Compound 1 besylate prepared as disclosed herein. In one example, the application provides the crystalline Compound 1 besylate prepared by precipitating the solid crystalline form of Compound 1 besylate from a mixture of Compound 1 besylate with THF (e.g., a solution of Compound 1 besylate in THF). In another example, the application provides the crystalline Compound 1 besylate prepared by precipitating the crystalline form of Compound 1 besylate from a mixture of Compound 1 besylate with ethanol (e.g., a solution of Compound 1 besylate in ethanol).


Compound 1 Citric Acid Salt


In some embodiments, provided herein is Compound 1 citrate. In some embodiments, Compound 1 citrate is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% crystalline solid. In some embodiments, crystalline Compound 1 citrate is substantially free of other forms of Compound 1 citrate. In some embodiments, crystalline Compound 1 citrate contains less than 10%, such as less than 5%, such as less than 3% of other forms of Compound 1 citrate. In some embodiments, the crystalline form of Compound 1 citrate is substantially free of the amorphous form of Compound 1 citrate. In some embodiments, the crystalline form of Compound 1 citrate contains less than 10%, less than 5%, or less than 3% of the amorphous form of compound 1 citrate.


In some embodiments, the molar ratio of Compound 1 to the citric acid in the citrate is about 1:1. In some embodiments, Compound 1 citrate is a monocitrate. In some embodiments, crystalline Compound 1 citrate has Form A, which is described below in the Examples. In some embodiments, the Compound 1 citrate Form A has an XRPD pattern substantially as depicted in FIG. 21.


In some embodiments, the Compound 1 citrate Form A has a XRPD peak, in terms of 2-theta, at about 20.7 degrees. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 20.7, about 21.6, and about 24.8. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 8.9, about 11.1, about 14.4, about 15.4, about 20.7, about 21.6, and about 24.8. In some embodiments, Compound 1 citrate Form A has XRPD peaks, in terms of 2-theta, at about 8.9, about 11.1, about 13.9, about 14.4, about 15.4, about 19.2, about 20.7, about 21.6, about 24.8, and about 25.6.


In some embodiments, Compound 1 citrate Form A has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 6.5, about 8.9, about 9.2, about 11.1, about 13.9, about 14.4, about 15.4, about 15.9, about 18.0, about 19.2, about 19.6, about 20.7, about 21.6, about 22.7, about 23.3, about 23.7, about 24.2, about 24.8, about 25.6, about 26.3, about 26.5, about 26.8, about 27.9, about 28.9, about 29.1, about 30.2, about 32.5, and about 33.7. In some embodiments, Compound 1 citrate Form A has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 6.5, about 8.9, about 9.2, about 11.1, about 13.9, about 14.4, about 15.4, about 15.9, about 18.0, about 19.2, about 19.6, about 20.7, about 21.6, about 23.3, about 23.7, about 24.2, about 24.8, about 25.6, about 26.5, and about 27.9. In some embodiments, Compound 1 citrate Form A has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 8.9, about 11.1, about 14.4, about 15.4, about 19.2, about 20.7, about 21.6, about 24.8, and about 25.6.


In some embodiments, Compound 1 citrate Form A has at least one, at least two, or at least three XRPD peaks, in terms of 2-theta, selected from about 6.5, about 8.9, about 9.2, about 11.1, about 13.9, about 14.4, about 15.4, about 15.9, about 18.0, about 19.2, about 19.6, about 20.7, about 21.6, about 22.3, about 22.7, about 23.3, about 23.7, about 24.2, about 24.8, about 25.6, about 26.3, about 26.5, about 26.8, about 27.9, about 28.9, about 29.1, about 30.2, about 30.6, about 31.8, about 32.5, about 33.1, about 33.7, about 34.3, and about 34.5.


In some embodiments, Compound 1 citrate Form A has a DTA thermogram substantially as depicted in FIG. 43. In some embodiments, Compound 1 citrate Form A has a DTA thermogram characterized by an endothermal event at about 194° C. In some embodiments, Compound 1 citrate Form A has a DTA thermogram characterized by an endothermal event at about 318° C. In some embodiments, Compound 1 citrate Form A has a DTA thermogram characterized by an endothermal event at about 194° C. and an endothermal event at about 318° C. In some embodiments, Compound 1 citrate Form A has a DSC thermogram substantially as depicted in FIG. 44. In some embodiments, Compound 1 citrate Form A has a DSC thermogram characterized by an endothermal event at about 205° C. In some embodiments, Compound 1 citrate Form A has a DSC thermogram characterized by an endothermal event at about 194° C. and an endothermal event at about 205° C. In some aspects of these embodiments, the endothermal events are overlapping.


In some embodiments, Compound 1 citrate Form A has hygroscopicity characterized by a mass uptake of around 1.8% at 90% RH as determined by DVS analysis. Compound 1 citrate Form A is substantially anhydrous (Form A is not hydrated) and is substantially free of organic solvents (Form A is not solvated).


In some embodiments, Compound 1 citrate Form A is substantially pure (e.g., free of organic, inorganic or other impurities). In some embodiments, the purity of Compound 1 citrate Form A is 90 wt. % or more, 95 wt. % or more, or 99 wt. % or more. In some embodiments, Compound 1 citrate Form A is substantially free of other crystalline forms of Compound 1 citrate. For example, Compound 1 citrate Form A is substantially free of Compound 1 citrate Form B.


In some embodiments, the citric acid salt of Compound 1 may form a hydrate. In some aspects of these embodiments, the hydrate is crystalline.


In some embodiments, the crystalline Compound 1 citrate has Form B, which has an XRPD pattern substantially as depicted in FIG. 49.


In some embodiments, the present disclosure provides a crystalline form of Compound 1 citrate prepared as disclosed herein. In one example, the present application provides Compound 1 citrate Form A prepared by precipitating Form A from a mixture of Compound 1 citrate with acetone (e.g., a solution of Compound 1 in acetone).


Compound 1 Methanesulfonic Acid Salt


In some embodiments, provided herein is Compound 1 mesylate. In some embodiments, the Compound 1 mesylate is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% crystalline solid. In some embodiments, the crystalline form of Compound 1 mesylate is substantially free of the amorphous form of Compound 1 mesylate. In some embodiments, the crystalline form of Compound 1 mesylate contains less than 10%, less than 5%, or less than 3% of the amorphous form of compound 1 mesylate.


In some embodiments, the molar ratio of the Compound 1 to the methanesulfonic acid in the mesylate is about 1:1. In some embodiments, the Compound 1 mesylate is a monomesylate.


In some embodiments, the crystalline form of Compound 1 mesylate has an XRPD pattern substantially as depicted in FIG. 16. In some embodiments, the crystalline solid of the Compound 1 mesylate has a DTA thermogram substantially as depicted in FIG. 25. In some embodiments, the crystalline solid of the Compound 1 mesylate has a DTA thermogram characterized by an endothermal event at about 232° C. (e.g., a melting point of the mesylate). In some embodiments, the crystalline Compound 1 mesylate has a DSC thermogram substantially as depicted in FIG. 32. In some embodiments, the crystalline Compound 1 mesylate has a DSC thermogram characterized by an endothermal event at about 233° C. The crystalline form of the mesylate is substantially anhydrous (the crystalline form is not hydrated) and is substantially free of organic solvents (the crystalline form is not solvated). In some embodiments, the crystalline form of the mesylate is substantially pure (e.g., purity is 90 wt. % or more, 95 wt. % or more, or 99 wt. % or more). In some embodiments, the crystalline form of Compound 1 mesylate is substantially free of other crystalline forms of Compound 1 mesylate.


Compound 1 mesylate can be prepared as an acetone solvate. In some embodiments, the acetone solvate of the mesylate is a solid form (e.g., an amorphous solid, a crystalline solid, or a mixture thereof). In some embodiments, the acetone solvate of the mesylate is crystalline. In some embodiments, the crystalline form of the acetone solvate of the mesylate salt of Compound 1 is has an XRPD pattern substantially as depicted in FIG. 30. In some embodiments, the crystalline acetone solvate has a DTA thermogram substantially as depicted in FIG. 31. In some embodiments, the crystalline acetone solvate has a DTA thermogram characterized by an endothermal event at about 125° C. and an endothermal event at about 232° C. (melting point). The endothermal event at about 125° C. is likely associated with the desolvation of the material. In some embodiments, the crystalline acetone solvate has a DSC thermogram characterized by an endothermal event at about 233° C. at the first heating cycle, a solidification event at about 181° C. at the first cooling cycle, and an endothermal event at about 229° C. at the second heating cycle. In some embodiments, the acetone solvate readily desolvates upon heating to produce crystalline form of the Compound 1 mesylate.


In some embodiments, the present disclosure provides a crystalline form of Compound 1 mesylate prepared as disclosed herein. In one example, the application provides the crystalline form of Compound 1 mesylate prepared by precipitating the solid crystalline form of Compound 1 mesylate from a mixture of Compound 1 mesylate in 2-propanol (e.g., a solution of Compound 1 in isopropanol).


Other Salts


In some embodiments provided herein is a salt of Compound 1 which is Compound 1 edisylate, Compound 1 tosylate, Compound 1 oxalate, Compound 1 fumarate, Compound 1 L-malate or Compound 1 succinate. In some embodiments, each of Compound 1 edisylate, Compound 1 tosylate, Compound 1 oxalate, Compound 1 fumarate, Compound 1 L-malate and or Compound 1 succinate can be prepared as a solid form, e.g., as an amorphous solid, as a crystalline solid, or as a mixture thereof. In some aspects of these embodiments, any of the aforementioned salts of Compound 1 is at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 99% crystalline solid. In other aspects of these embodiments, the crystalline salt of Compound 1 is substantially free of the amorphous form of the salt. For example, Compound 1 salt contains less than 10%, less than 5%, or less than 3% of the amorphous form of the salt.


In some embodiments, the present disclosure provides crystalline forms of Compound 1 edisylate, Compound 1 tosylate, Compound 1 oxalate, Compound 1 fumarate, Compound 1 L-malate or Compound 1 succinate prepared as disclosed herein.


In some embodiments, the crystalline Compound 1 edisylate has an XRPD pattern substantially as depicted in FIG. 14.


In some embodiments, the crystalline Compound 1 edisylate has XRPD peaks, in terms of 2-theta, at about 20.0, about 20.6, and about 23.3. In some embodiments, the crystalline Compound 1 edisylate has XRPD peaks, in terms of 2-theta, at about 18.1, about 18.3, about 20.0, about 20.6, about 23.3, and about 25.3. In some embodiments, the crystalline Compound 1 edisylate has XRPD peaks, in terms of 2-theta, at about 11.6, about 15.5, about 17.0, about 18.1, about 18.3, about 20.0, about 20.6, about 23.3, about 24.9, and about 25.3.


In some embodiments, the crystalline Compound 1 tosylate has an XRPD pattern substantially as depicted in FIG. 15.


In some embodiments, the crystalline Compound 1 tosylate has XRPD peaks, in terms of 2-theta, at about 6.6, about 16.9, and about 21.2. In some embodiments, the crystalline Compound 1 tosylate has XRPD peaks, in terms of 2-theta, at about 6.6, about 8.2, about 15.0, about 16.9, about 21.2, and about 21.6. In some embodiments, the crystalline Compound 1 tosylate has XRPD peaks, in terms of 2-theta, at about 6.6, about 8.2, about 11.8, about 15.0, about 16.9, about 21.2, about 21.6, about 21.9, about 24.2 and about 24.9.


In some embodiments, the crystalline Compound 1 tosylate has a DTA thermogram substantially as depicted in FIG. 24. In some embodiments, the crystalline Compound 1 tosylate has a DTA thermogram characterized by an endothermal event at about 90° C.


In some embodiments, the crystalline Compound 1 oxalate has an XRPD pattern substantially as depicted in FIG. 19.


In some embodiments, the crystalline Compound 1 oxalate has XRPD peaks, in terms of 2-theta, at about 20.2, about 20.5, and about 24.9. In some embodiments, the crystalline Compound 1 oxalate has XRPD peaks, in terms of 2-theta, at about 11.2, about 18.6, about 20.2, about 20.5, about 23.5, and about 24.9. In some embodiments, the crystalline Compound 1 oxalate has XRPD peaks, in terms of 2-theta, at about 11.2, about 18.6, about 20.0, about 20.2, about 20.5, about 21.1, about 22.9, about 23.5, about 24.9, and about 27.0.


In some embodiments, the crystalline Compound 1 oxalate has a DTA thermogram substantially as depicted in FIG. 26. In some embodiments, the crystalline Compound 1 oxalate has a DTA thermogram characterized by an endothermal event at about 317° C. (a melting point).


In some embodiments, the crystalline Compound 1 fumarate has an XRPD pattern substantially as depicted in FIG. 20.


In some embodiments, the crystalline Compound 1 fumarate has XRPD peaks, in terms of 2-theta, at about 9.3, about 21.6, and about 27.1. In some embodiments, the crystalline Compound 1 fumarate has XRPD peaks, in terms of 2-theta, at about 9.3, about 14.8, about 21.6, about 22.2, about 27.1, and about 27.9. In some embodiments, the crystalline Compound 1 fumarate has XRPD peaks, in terms of 2-theta, at about 6.4, about 9.3, about 14.8, about 19.4, about 19.8, about 20.4, about 21.6, about 22.2, about 27.1, and about 27.9.


In some embodiments, the crystalline Compound 1 fumarate has a DTA thermogram substantially as depicted in FIG. 27. In some embodiments, the crystalline Compound 1 fumarate has a DTA thermogram characterized by an endothermal event at about 166° C. In some embodiments, the crystalline Compound 1 fumarate has a DTA thermogram characterized by an endothermal event at about 191° C. In some embodiments, the crystalline Compound 1 fumarate has a DTA thermogram characterized by an endothermal event at about 201° C. In some embodiments, the crystalline Compound 1 fumarate has a DTA thermogram characterized by an endothermal event at about 312° C. In some embodiments, the crystalline Compound 1 fumarate has a DTA thermogram characterized by an endothermal event at about 166° C., an endothermal event at about 191° C., an endothermal event at about 201° C., and an endothermal event at about 312° C.


In some embodiments, the crystalline Compound 1 L-malate has an XRPD pattern substantially as depicted in FIG. 22.


In some embodiments, the crystalline Compound 1 malate has XRPD peaks, in terms of 2-theta, at about 19.3, about 21.6, and about 24.9. In some embodiments, the crystalline Compound 1 malate has XRPD peaks, in terms of 2-theta, at about 10.7, about 13.4, about 18.8, about 19.3, about 21.6, and about 24.9. In some embodiments, the crystalline Compound 1 malate has XRPD peaks, in terms of 2-theta, at about 6.7, about 10.7, about 13.4, about 18.8, about 19.3, about 19.9, about 21.1, about 21.6, about 23.9, and about 24.9.


In some embodiments, the crystalline Compound 1 L-malate has a DTA thermogram substantially as depicted in FIG. 28. In some embodiments, the crystalline Compound 1 L-malate has a DTA thermogram characterized by an endothermal event at about 162° C. In some embodiments, the crystalline Compound 1 L-malate has a DTA thermogram characterized by an endothermal event at about 313° C. In some embodiments, the crystalline Compound 1 L-malate has a DTA thermogram characterized by an endothermal event at about 162° C. and an endothermal event at about 313° C. In some embodiments, the crystalline form of Compound 1 succinate has pattern 1. In some embodiments, the crystalline Compound 1 succinate has an XRPD pattern substantially as depicted in FIG. 23.


In some embodiments, the crystalline Compound 1 succinate has XRPD peaks, in terms of 2-theta, at about 9.1, about 21.5, and about 26.8. In some embodiments, the crystalline Compound 1 succinate has XRPD peaks, in terms of 2-theta, at about 9.1, about 11.2, about 19.4, about 21.5, about 26.0, and about 26.8. In some embodiments, the crystalline Compound 1 succinate has XRPD peaks, in terms of 2-theta, at about 6.4, about 9.1, about 11.2, about 14.5, about 15.8, about 19.4, about 20.5, about 21.5, about 26.0, about 26.8.


In some embodiments, the crystalline Compound 1 succinate has a DTA thermogram substantially as depicted in FIG. 29. In some embodiments, the crystalline Compound 1 oxalate has a DTA thermogram characterized by an endothermal event at about 151° C. In some embodiments, the crystalline Compound 1 oxalate has a DTA thermogram characterized by an endothermal event at about 315° C. In some embodiments, the crystalline Compound 1 oxalate has a DTA thermogram characterized by an endothermal event at about 151° C. and an endothermal event at about 315° C. Compound 1 hydrochloride, Compound 1 sulfate, Compound 1 2-naphthalenesulfonate, Compound 1 isethionate, Compound 1 L-aspartate, Compound 1 maleate, Compound 1 phosphate, Compound 1 esylate, Compound 1 glutamate, Compound 1 L-tartrate, Compound 1 D-glucuronate, Compound 1 hippurate, Compound 1 D-gluconate, Compound 1 lactate, Compound 1 L-ascorbate, Compound 1 benzoate are provided herein and each of these salts can be prepared by treating Compound 1 with the corresponding acid.


Synthetic Preparations


Compound 1 and its forms


In some embodiments, Compound 1 (free base) may be prepared as described as disclosed herein. The crystalline form of Compound 1 (e.g., Form I as described herein) may be prepared by the method comprising precipitating the crystalline form from a mixture comprising Compound 1 (free base). In some embodiments, the mixture further comprises a solvent. In some embodiments, the method comprises obtaining a mixture of Compound 1 with a solvent. In some embodiments, the mixture is a solution of Compound 1 in a solvent. In some embodiments, the solution is saturated. The solvent may be selected from acetone, acetonitrile, 2-butanone, cyclopropylmethyl ether, 1,2-dimethoxyethane, 1,4-dioxane, ethanol, ethyl acetate, 2-ethoxy ethanol, isobutyl acetate, isopropyl acetate, methanol, MIBK, 2-propanol, 1-propanol and THF.


In some embodiments, the precipitating is carried out at a temperature above 0° C. (e.g., 5° C., 10° C., 20° C., or 30° C.). In some embodiments, the precipitating is carried out below room temperature. In some aspects of these embodiments, the precipitating is carried out below 10° C. In some embodiments, the precipitating is carried out at about 2° C. In some aspects of these embodiments, the solution comprises 2-propanol (e.g., Compound 1 is precipitated from the solution in 2-propanol).


In some embodiments, the precipitating is carried out at a temperature below 0° C. (e.g., −5° C., −10° C., −20° C., or −30° C.). In some aspects of these embodiments, the precipitating is carried out at about −18° C. In other aspects of these embodiments, the solution comprises a solvent selected from 1-butanol, ethanol, 2-propanol and 1-propanol. For example, the Form I of Compound 1 may be precipitated by cooling a saturated solution of Compound 1 in, e.g., 1-butanol, and further collecting the resultant solid.


In some embodiments, the precipitating is carried out for a time period from about 24 hours to about 72 hours (e.g., cooled solution of Compound 1 may be stored at the specified temperature for 24-72 hours).


In some embodiments, the precipitating comprises adding an anti-solvent to the solution of Compound 1. In some aspects of these embodiments, the anti-solvent is miscible with the solvent in which Compound 1 is dissolved. For example, the anti-solvent may be selected from heptane and t-butylmethyl ether (herein also referred to as TBME). In some embodiments, the precipitating is carried out at or above room temperature. In some aspects of these embodiments, the solvent may be acetone, acetonitrile, 2-butanone, 1,2-dimethoxyethane, 1,4-dioxane and ethanol. For instance, a MTBE may be added to the solution of Compound 1 in acetone at room temperature, followed by collection of the precipitated Form I. In other aspects of these embodiments, the precipitating is carried out below room temperature (e.g., at 0° C., 5° C., or 10° C.). In one example, the precipitating is carried out at about 2° C. In some aspects of these embodiments, the solvent may be selected from acetone, acetonitrile, 1-butanol, 2-butanone, 1,2-dimethoxyethane, 1,4-dioxane, ethanol, ethyl acetate, MIBK, 1-propanol and THF. For instance, a heptane may be added to the solution of Compound 1 in ethyl acetate at about 2° C., followed by collection of the precipitated Form I.


In some embodiments, the precipitating may be carried out by evaporating the solvent. In some aspects of these embodiments, the evaporating may be carried out at about room temperature. In other aspects of these embodiments, the solvent is selected from acetone, acetonitrile, 2-butanone, cyclopropylmethyl ether, 1,2-dimethoxyethane, 1,4-dioxane, ethanol, ethyl acetate, 2-ethoxy ethanol, isobutyl acetate, isopropyl acetate, methanol, MIBK, 2-propanol, 1-propanol and THF.


Compound 1 Salts and Crystalline Forms


Generally, the salts of the Compound 1 can be prepared by combining (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Compound 1 free base) with an acid. That is, any one of the salts of Compound 1 described herein may be prepared by combining the Compound 1 with a benzenesulfonic acid, a citric acid, a methanesulfonic acid, a 1,2-ethane disulfonic acid, a p-toluene sulfonic acid, an oxalic acid, a fumaric acid, a L-malic acid, a hydrochloric acid, a sulfuric acid, a naphthalene-2-sulfonic acid, a 2-hydroxy ethanesulfonic acid, a L-aspartic acid, a maleic acid, a phosphoric acid, a ethanesulfonic acid, a L-glutamic acid, a L-tartaric acid, a D-glucuronic acid, a hippuric acid, a D-gluconic acid, a DL-lactic acid, a L-ascorbic acid, or a benzoic acid. In some embodiments, the combining may be carried out in the presence of a solvent, such as, for example, acetone, ethanol, methanol, 2-propanol, TBME or THF. In some embodiments, Compound 1 is combined with a solvent to obtain the first solution, an acid is separately combined with a solvent to obtain the second solution, and the salt of Compound 1 is obtained by combining the first solution with the second solution. In some embodiments, the combining is carried out with the acid in molar excess with respect to the Compound 1 free base. In some aspects of these embodiments, the molar ratio of the acid to the Compound 1 is from about 1:1 to about 1.1:1 (e.g., about 1.05:1). In some embodiments, the combining is carried out from about room temperature to about 40° C. (e.g., the combining is carried out by cycling the temperature between ambient and 40° C. in 4 hour cycles). In some embodiments, the combining is carried out for a time period from 24 hours to 72 hours.


Generally, any one of the crystalline forms of the salts of Compound 1 may be obtained by precipitating the crystalline form from a mixture of the salt with a solvent (e.g., precipitating the crystalline compound from a mixture, such as precipitating the crystalline compound from a solution). In some embodiments, the precipitating is carried out by temperature cycling the reaction mixture from about room temperature to about 40° C. (e.g., 4 hour cycles between room temperature and 40° C.). In some embodiments, the precipitating is carried out by evaporating the solvent from the mixture (e.g., by evaporating the solvent from the solution of Compound 1). In some embodiments, the precipitating is carried out by adding an anti-solvent (e.g., heptane of MTBE) to the solution of Compound 1 in a solvent.


In some embodiments, crystalline Compound 1 besylate may be obtained by precipitating the crystalline form from a mixture of Compound 1 besylate with a solvent selected from THF and t-BME. In some aspects of these embodiments, the mixture is a solution of Compound 1 besylate in THF or t-BME.


In some embodiments, crystalline Compound 1 besylate may be prepared by precipitating the crystalline form from a mixture of Compound 1 besylate with ethanol. In some aspects of these embodiments, the mixture is a solution of Compound 1 besylate in ethanol.


In some embodiments, crystalline Compound 1 citrate Form A may be prepared by precipitating Form A from a mixture of Compound 1 citrate with a solvent selected from acetone and t-BME. In some aspects of these embodiments, the mixture is a solution of Compound 1 citrate in acetone or t-BME.


In some embodiments, crystalline form of Compound 1 mesylate may be prepared by precipitating the crystalline form from a mixture of Compound 1 mesylate with a solvent selected from acetone, methanol and 2-propanol. In some aspects of these embodiments, the mixture is a solution of Compound 1 mesylate in acetone, methanol or 2-propanol.


In some embodiments, crystalline form of Compound 1 edisylate may be prepared by precipitating the crystalline form from a mixture of Compound 1 edisylate with 2-propanol. In some aspects of these embodiments, the mixture is a solution of Compound 1 edisylate in 2-propanol.


In some embodiments, crystalline form of Compound 1 tosylate may be prepared by precipitating the crystalline form from a mixture of Compound 1 tosylate with a solvent selected from acetone and THF. In some aspects of these embodiments, the mixture is a solution of Compound 1 tosylate in acetone or THF.


In some embodiments, crystalline form of Compound 1 oxalate may be prepared by precipitating the crystalline form from a mixture of Compound 1 oxalate with a solvent selected from ethanol and methanol. In some aspects of these embodiments, the mixture is a solution of Compound 1 oxalate in ethanol or methanol.


In some embodiments, a crystalline form of Compound 1 fumarate may be prepared by precipitating the crystalline form from a mixture of Compound 1 fumarate with acetone. In some aspects of these embodiments, the mixture is a solution of Compound 1 fumarate in ethanol or methanol.


In some embodiments, crystalline form of Compound 1 L-malate may be prepared by precipitating the crystalline form from a mixture of Compound 1 L-malate with TBME. In some aspects of these embodiments, the mixture is a solution of Compound 1 L-malate in TBME.


In some embodiments, crystalline form of Compound 1 succinate may be prepared by precipitating the crystalline form from a mixture of Compound 1 succinate with acetone. In some aspects of these embodiments, the mixture is a solution of Compound 1 succinate in acetone.


Methods of Use


Certain compounds which are inhibitors of TrkA and/or TrkB may be useful in the treatment of multiple types of pain including inflammatory pain, neuropathic pain, and pain associated with cancer, surgery, and bone fracture.


In one embodiment, Compound 1 or its solid forms, crystalline forms, solvates or hydrates, or the salts of Compound 1 or their solid forms, crystalline forms, solvates and hydrates as described herein, are useful for treating pain, including chronic and acute pain, in a mammal.


Acute pain, as defined by the International Association for the Study of Pain, results from disease, inflammation, or injury to tissues. This type of pain generally comes on suddenly, for example, after trauma or surgery, and may be accompanied by anxiety or stress. The cause can usually be diagnosed and treated, and the pain is confined to a given period of time and severity. In some rare instances, it can become chronic.


Chronic pain, as defined by the International Association for the Study of Pain, is widely believed to represent disease itself. It can be made much worse by environmental and psychological factors. Chronic pain persists over a longer period than acute pain and is resistant to most medical treatments, generally over 3 months or more. It can and often does cause severe problems for patients.


Compound 1 or its solid forms, crystalline forms, solvates or hydrates, or the salts of Compound 1 or their solid forms, crystalline forms, solvates and hydrates as described herein, are also useful for treating cancer in a mammal. Particular examples include neuroblastoma, ovarian, pancreatic, colorectal and prostate cancer.


Compound 1 or its solid forms, crystalline forms, solvates or hydrates, or the salts of Compound 1 or their solid forms, crystalline forms, solvates and hydrates as described herein, are also useful for treating inflammation in a mammal.


Compound 1 or its solid forms, crystalline forms, solvates or hydrates, or the salts of Compound 1 or their solid forms, crystalline forms, solvates and hydrates as described herein, are also useful for treating certain infectious diseases in a mammal, such as Trypanosoma cruzi infection.


Compound 1 or its solid forms, crystalline forms, solvates or hydrates, or the salts of Compound 1 or their solid forms, crystalline forms, solvates and hydrates as described herein, may also be used to treat neurodegenerative diseases in a mammal. Examples of neurodegenerative disease include demyelination and dysmyelination. Additional examples of neurodegenerative diseases include multiple sclerosis, Parkinson's disease and Alzheimer's disease.


In addition, Compound 1 or its solid forms, crystalline forms, solvates or hydrates, or the salts of Compound 1 or their solid forms, crystalline forms, solvates and hydrates as described herein, may also be used to treat interstitial cystitis (IC), painful bladder syndrome (PBS), urinary incontinence, asthma, anorexia, atopic dermatitis, and psoriasis in a subject (e.g., a mammal such as a human).


Accordingly, another embodiment of the present application provides a method of treating or preventing pain in a subject (e.g., mammal), comprising administering to said mammal Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, in an amount effective to treat or prevent said pain. In one embodiment, the pain is chronic pain. In one embodiment, the pain is acute pain. In one embodiment, the pain is inflammatory pain. In one embodiment, the pain is neuropathic pain. In one embodiment, the pain is pain associated with cancer. In one embodiment, the pain is pain associated with surgery. In one embodiment, the pain is pain associated with bone fracture. In one embodiment, the method comprises a method of treating said pain in a mammal. In one embodiment, the method comprises a method of preventing said pain in a mammal.


Another embodiment of the present disclosure provides a method of treating or preventing inflammation in a subject (e.g., mammal), comprising administering to said mammal Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, in an amount effective to treat or prevent the inflammation. In one embodiment, the method comprises treating the inflammation in a subject. In one embodiment, the method comprises preventing the inflammation in a subject.


Another embodiment of the present application provides a method of treating or preventing a neurodegenerative disease in a mammal, comprising administering to said mammal Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, in an amount effective to treat or prevent said neurodegenerative disease. In one embodiment, the neurodegenerative disease is demyelination. In one embodiment, the neurodegenerative disease is dysmyelination. In one embodiment, the neurodegenerative disease is multiple sclerosis. In one embodiment, the neurodegenerative disease is Parkinson's disease. In one embodiment, the neurodegenerative disease is Alzheimer's disease.


Another embodiment of the present disclosure provides a method of treating or preventing an infectious disease in a subject, comprising administering to the subject Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, in an amount effective to treat or prevent said infectious disease. In one embodiment, the infectious disease is Trypanosoma cruzi infection. In one embodiment, the method comprises treating the neurodegenerative disease in a subject. In one embodiment, the method comprises preventing the neurodegenerative disease in a subject.


Another embodiment of the present disclosure provides a method of treating or preventing cancer in a mammal, comprising administering to said mammal Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, in an amount effective to treat or prevent the cancer. In one embodiment, the cancer is neuroblastoma. In one embodiment, the cancer is ovarian cancer. In one embodiment, the cancer is pancreatic cancer. In one embodiment, the cancer is colorectal cancer. In one embodiment, the cancer is prostate cancer. In one embodiment, the method comprises treating the cancer in a subject. In one embodiment, the method comprises preventing the cancer in a subject.


Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, may be administered alone as a sole therapy or can be administered in addition with one or more other substances and/or treatments that work by the same or a different mechanism of action. Examples include anti-inflammatory compounds, steroids (e.g., dexamethasone, cortisone and fluticasone), analgesics such as NSAIDs (e.g., aspirin, ibuprofen, indomethacin, and ketoprofen), and opioids (such as morphine), and chemotherapeutic agents. These agents may be administered with Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, as part of the same or separate dosage forms, via the same or different routes of administration, and on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art.


In the field of medical oncology, it is normal practice to use a combination of different forms of treatment to treat each patient with cancer. In medical oncology the other component(s) of such conjoint treatment in addition to compositions of the present disclosure may be, for example, surgery, radiotherapy, chemotherapy, signal transduction inhibitors and/or immunotherapy (e.g., monoclonal antibodies).


Accordingly, Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, may be administered in combination with one or more agents selected from mitotic inhibitors, alkylating agents, anti-metabolites, antisense DNA or RNA, intercalating antibiotics, growth factor inhibitors, signal transduction inhibitors, cell cycle inhibitors, enzyme inhibitors, retinoid receptor modulators, proteasome inhibitors, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, cytostatic agents anti-androgens, targeted antibodies, HMG-CoA reductase inhibitors, and prenyl-protein transferase inhibitors. These agents may be administered with one or more Compound 1, its solid form, crystalline form, solvate or hydrate, or a salt of Compound 1, or solid form, crystalline form, solvate or hydrate of the salt as described herein, as part of the same or separate dosage forms, via the same or different routes of administration, and on the same or different administration schedules according to standard pharmaceutical practice known to one skilled in the art.


The term “TRK-associated cancer” as used herein refers to cancers associated with or having a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same. Exemplary TRK-associated cancers are provided herein.


The phrase “dysregulation of a TRK gene, a TRK kinase, or the expression or activity or level of any of the same” refers to a genetic mutation (e.g., a TRK gene translocation that results in the expression of a fusion protein, a deletion in a TRK gene that results in the expression of a TRK protein that includes a deletion of at least one amino acid as compared to the wild-type TRK protein, a mutation in a TRK gene that results in the expression of a TRK protein with one or more point mutations, or an alternative spliced version of a TRK mRNA that results in a TRK protein having a deletion of at least one amino acid in the TRK protein as compared to the wild-type TRK protein) or a TRK gene amplification that results in overexpression of a TRK protein or an autocrine activity resulting from the overexpression of a TRK gene in a cell that results in a pathogenic increase in the activity of a kinase domain of a TRK protein (e.g., a constitutively active kinase domain of a TRK protein) in a cell. As another example, a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same, can be a mutation in a TRK gene that encodes a TRK protein that is constitutively active or has increased activity as compared to a protein encoded by a TRK gene that does not include the mutation. For example, a dysregulation of a TRK gene, a TRK protein, or expression or activity, or level of any of the same, can be the result of a gene or chromosome translocation which results in the expression of a fusion protein that contains a first portion of TRK that includes a functional kinase domain, and a second portion of a partner protein that is not TRK. In some examples, dysregulation of a TRK gene, a TRK protein, or expression or activity or level of any of the same can be a result of a gene translocation of one TRK gene with another non-TRK gene. Non-limiting examples of fusion proteins are described in Tables 1-3. Additional examples of TRK kinase protein mutations (e.g., point mutations) are TRK inhibitor resistance mutations.


The term “wildtype” or “wild-type” when referring to a TRK nucleic acid or protein describes a nucleic acid (e.g., a TRK gene or a TRK mRNA) or protein (e.g., a TRK protein) that is found in a subject that does not have a TRK-associated disease, e.g., a TRK-associated cancer (and optionally also does not have an increased risk of developing a TRK-associated disease and/or is not suspected of having a TRK-associated disease), or is found in a cell or tissue from a subject that does not have a TRK-associated disease, e.g., a TRK-associated cancer (and optionally also does not have an increased risk of developing a TRK-associated disease and/or is not suspected of having a TRK-associated disease).


In some embodiments, the dysregulation of a TRK gene, a TRK kinase protein, or expression or activity or level of any of the same, includes one or more chromosome translocations or inversions resulting in a TRK gene fusion. In some embodiments, the dysregulation of a TRK gene, a TRK kinase protein, or expression or activity or level of any of the same, is a result of genetic translocations in which the expressed protein is a fusion protein containing residues from a non-TRK partner protein, and includes a minimum of a functional TRK kinase domain. See, for example, Tables 1-3.









TABLE 1







Exemplary TrkA Fusion Proteins and Cancers











Non-limiting Exemplary Trk-




and Synonyms of Associated


Fusion Protein
Non-TrkA Fusion Partner
Cancer(s)





TP53-TrkA1, 11
Tumor Protein P53
Spitzoid Melanoma, Spitz tumors


LMNA-TrkA1, 12
Lamin A/C
Spitzoid Melanoma, Spitz tumors,




Undifferentiated Sarcoma, Adult




Soft Tissue Sarcoma (e.g., Soft




Tissue Sarcoma Metastatic to




Lung), Soft Tissue Fibrosarcoma,




Spindle Cell SarcomaG,




Congenital Infantile




FibrosarcomaH, Pediatric




haemangiopericytoma-like




sarcomaI, Colorectal CancerK,




Pediatric soft tissue tumorM


CD74-TrkA2
MHC class II
Non-Small Cell Lung Cancer



invariant chain
(NSCLC)




Lung adenocarcimona


TFG-TrkA
TRK-Fused Gene
Papillary Thyroid Carcinoma


(TRK-T3)3

(PTC), Soft Tissue Solitary




Fibrous Tumor


TPM3-TrkA3
Tropomyosin 3
Lung Cancer, Papillary Thyroid




Carcinoma (PTC), Acute Myeloid




Leukemia (AML), Sarcoma,




Pediatric Gliomas, Colorectal




Cancer (CRC), Soft Tissue




Schwannoma, Spitzoid




melanocytic tumorsJ


NFASC-TrkA4
Neurofascin
Gliobastoma multiforme (GBM);




Glioblastoma


BCAN-TrkA4
Brevican
Glioblastoma multiforme (GBM)


MPRIP-TrkA5, E
Myosin Phosphatase Rho
Non-small cell lung cancer



Interacting Protein or Rho
(NSCLC), Lung adenocarcinoma



Interacting Protein 3


TPR-TrkA
Translocated Promoter
Papillary Thyroid Carcinoma


(TRK-T1 or
Region, Nuclear Basket
(PTC), Colorectal Cancer (CRC)A,


TRK-T2)3
Protein
Non-small cell lung cancer




(NSCLC)


RFWD2-TrkA6
Ring Finger and WD Repeat
Large Cell Neuroendrocine Cancer



Domain 2
(LCNEC); NSCLC


IRF2BP2-TrkA7
Interferon Regulatory Factor
Thyroid Cancer; Thyroid Gland



2 Binding Protein 2
Carcinoma


SQSTM1-TrkA7
Sequestosome 1
Thyroid Cancer (e.g., Papillary




Thyroid Cancer), Thyroid Gland




Carcinoma, Soft




TissueFibrosarcoma, Non-small-




cell lung cancerL


SSBP2-TrkA7
Single-Stranded DNA
Thyroid Cancer (e.g., Papillary



Binding Protein 2
Thyroid Cancer); Thyroid Gland




Carcinoma


RABGAP1L-
RAB GTPase Activating
Intrahepatic Cholangicarcinoma


TrkA8
Protein 1-Like
(ICC)


C18ORF8-TrkA9
Chromosome 18 Open
Non-Small Cell Lung Cancer



Reading Frame 8
(NSCLC)


RNF213-TrkA9
Ring Finger Protein 213
Non-Small Cell Lung Cancer




(NSCLC)


TBC1D22A-
TBC1 Domain Family,
Non-Small Cell Lung Cancer


TrkA9
Member 22A
(NSCLC)


C20ORF112-
Chromosome 20 Open
Non-Small Cell Lung Cancer


TrkA9
Reading Frame 112
(NSCLC)


DNER-TrkA9
Delta/Notch-Like EGF
Non-Small Cell Lung Cancer



Repeat Containing
(NSCLC)


ARHGEF2-
Rho Guanine Nucleotide
Glioblastoma


TrkA13
Exchange Factor 2


CHTOP-TrkA13
Chromatin Target of PRMT1
Glioblastoma


PPL-TrkA13
Periplakin
Thyroid Carcinoma


PLEKHA6-TrkA
Pleckstrin Homology



Domain-Containing Family A



Member 6


PEAR1-TrkA
Platelet Endothelial



Aggregation Receptor 1


MRPL24-TrkA
39S Ribosomal Protein L24,



Mitochondrial


MDM4-TrkA
Human Homolg of Mouse



Double Minute 4


LRRC71-TrkA
Leucine Rich Repeat



Containing 71


GRIPAP1-TrkA
GRIP1 Associated Protein 1


EPS15-TrkA
Epidermal Growth Factor



Receptor Substrate 15


DYNC2H1-
Dynein, Cytoplasmic 2,
Sarcoma


TrkAB
Heavy Chain 1


CEL-TrkA
Carboxyl Ester Lipase
Pancreatic adenocarcinoma




sampleD


EPHB2-TrkAB
EPH Receptor B2
Lower Grade Glioma


TGF-TrkAC
Transforming Growth Factor
Papillary Thyroid Cancer


NELL1-TrkAF
Cytoplasmic Protein That
Non-Small Cell Lung Cancer



Contains Epidermal Growth
(NSCLC)



Factor (Egf)-Like Repeats


EPL4-TrkAF
EPH-Related Receptor
Non-Small Cell Lung Cancer



Tyrosine Kinase Ligand 4/
(NSCLC)



Ephrin-A4 Protein


CTNND2-TrkAF
Catenin (Cadherin-Associated
Non-Small Cell Lung Cancer



Protein), Delta 2
(NSCLC)


TCEANC2-
Transcription Elongation
Non-Small Cell Lung Cancer


TrkAF
Factor A (Sll) N-Terminal
(NSCLC)



And Central Domain


SCYL3-TrkAN
SCY1 Like Pseudokinase 3
Colorectal Cancer






ACréancier et al., Cancer Lett. 365(1): 107-111, 2015. J




BU.S. patent application Pub. No. 2015/0315657.




CU.S. patent application Pub. No. 2015/0283132.




DEgren et al., Cancer Res. 75(15 Supplement): 4793, 2015.




EU.S. patent application Pub. No. 2015/0073036.




FP.C.T. Patent Application Publication No. WO2015184443A1.




GHaller et al., The Journal of pathology 238.5 (2016): 700-710.




HWong et al., J Natl Cancer Inst 2016; 108: djv307.




IHaller et al., J. Pathol. 238(5): 700-10.




JWu et al., Mod Pathol. 2016 April; 29(4): 359-69.




KKonicek et al., Cancer research, Vol. 76, No. 14, Supp. Supplement. Abstract Number: 2647; 107th Annual Meeting of the American Association for Cancer Research, AACR 2016. New Orleans, LA; 16-20 Apr. 2016.




LDrilon et al., Cancer research, Vol. 76, No. 14, Supp. Supplement. Abstract Number: CT007; 107th Annual Meeting of the American Association for Cancer Research, AACR 2016. New Orleans, LA; 16-20 Apr. 2016.




MKohsaka et al., Hum. Pathol., August 26. pii: S0046-8177(17)30299-X. doi: 10.1016/j.humpath.2017.08.017, 2017.




NMilione et al., Oncotarget, July 24; 8(33): 55353-55360. doi: 10.18632/oncotarget.19512, 2017.














TABLE 2







Exemplary TrkB Fusion Proteins and Cancers











Non-limiting Exemplary Trk-




and Synonyms of Associated


Fusion Protein
Non-TrkB Fusion Partner
Cancer(s)





NACC2-TrkB10
NACC Family Member 2,
Pilocytic Astrocytoma



BEN and BTB (POZ) Domain



Containing


QKI-TrkB10
QKI, KH Domain Containing,
Pilocytic Astrocytoma



RNA Binding


AFAP1-TrkB7
Actin Filament Associated
Lower-grade Glioma, In vitro



Protein 1
(murine Ba/F3 cells)B, Pilocytic




astrocytoma with anaplasia




(PAA)E


PAN3-TrkB7
PAN3 Poly(A) Specific
Head and Neck Squamous Cell



Ribonuclease Subunit
Carcinoma


SQSTM1-TrkB7
Sequestosome 1
Lower-Grade Glioma


TRIM24-TrkB7
Tripartite Motif Containing 24
Lung adenocarcinoma


VCL-TrkB11
Vinculin
Pediatric gliomas


AGBL4-TrkB11
ATP/GTP Binding Protein-
Pediatric gliomas



Like 4


DAB2IP-TrkB
Disabled Homolog 2-



Interacting Protein


NTRK2-TERTA
Telomerase Reverse
Thyroid Cancer



Transcriptase


TEL-TrkBC
ETS Variant 6
In vitro (murine Ba/F3 cells)


(ETV6)


QKI-TrkBD
Protein Quaking
Astrocytoma


NOS1AP-TrkB F

Anaplastic Astrocytoma


GKAP1-TrkB F

Glioblastoma


KCTD8-TrkB F

Glioblastoma


TBClD2-TrkB F

Glioblastoma


SOSTM1-TrkB F

Glioblastoma


VCAN-TrkB F

Grade II Astrocytoma


SLMAP-TrkBG

Ganglioma






APCT Patent Application Publication No. WO 2015/183836A1




BDrilon et al., Ann Oncol. 2016 May; 27(5): 920-6.




CYuzugullu et al., Cell Discov. 2: 16030, 2016.




DNi et al., Neuro Oncol. 2017 January; 19(1): 22-30.




ELin et al., Neuro-Oncol, Vol. 18, Supp. Supplement 3, pp. iii58, Abstract Number: HG-48; 17th International Symposium on Pediatric Neuro-Oncology, ISPNO 2016. Liverpool, UK, 12 Jun. 2016-15 Jun. 2016.




F Subramaniam et al., J. Clin. Onc., Vol. 35, No. 15, Supp. 1, 2017 Annual Meeting of the American Society of Clinical Oncology, ASCO. Chicago, IL, United States, 2017.




GEllison et al., Neuropathology and Applied Neurobiology., Vol. 42, Supp. 1, pp. 18. Abstract Number: O13, 117th Meeting of the British Neuropathological Society, Royal College of Physicians. London, United Kingdom, 2017.














TABLE 3







Exemplary TrkC Fusion Proteins and Cancers











Non-limiting Exemplary Trk-




and Synonyms of Associated


Fusion Protein
Non-TrkB Fusion Partner
Cancer(s)





ETV6-TrkC11
ETS Variant 6
Salivary Gland Cancer, Secretory


(TEL; or

Breast Carcinoma, Acute Myeloid


chromosomal

Leukemia, Fibrosarcoma,


translocation

Nephroma, Melanoma, Colorectal


t(12; 15)

Cancer (CRC), Breast Cancer,


(p13; q25))J

Pediatric Gliomas, Thyroid




Cancer (e.g., Papillary Thyroid




Cancer), Infantile Fibrosarcoma,




Soft Tissue Hemangioma,




Gastrointestinal Stromal Tumor




(GIST) (e.g., c-kit-negative




GIST), Mammary Carcinoma




(e.g., Mammary Analogue




Secretory Carcinoma, Secretory




Breast Carcinoma (SBSC))K,




Congenital Fibrosarcoma, Acute




Myelogenous Leukemia,




Polymorphous low-grade




adenocarcinomaD, ALK-negative




inflammatory myofibroblastic




tumors (IMT)E, Infantile




Fibrosarcoma (IFS)F,M, Acinic




cell carcinoma (AcCC)G, Cellular




mesoblastic nephromaH,




Promyelocytic leukemiaI, Burkitt




LymphomaI, B-cell lymphomaI,




multiple myelomaI,




medulloblastomaI,




neuroblastomaI, ovarian cancerI,




intestinal cancerI, acute




lymphblastic leukemiaK,




Sinonasal Low-grade Non-




intestinal-type AdenocarcinomaN


BTBD1-TrkC11
BTB (POZ) Domain
Pediatric Gliomas



Containing 1


LYN-TrkC7
V-Yes-1 Yamaguchi Sarcoma
Head and Neck Squamous Cell



Viral Related Oncogene
Carcinoma



Homolog (also known as



Lck/Yes-Related Novel



Protein Tyrosine Kinase)


RBPMS-TrkC7
RNA Binding Protein with
Thyroid Cancer (e.g., Papillary



Multiple Splicing
Thyroid Cancer)


EML4-TrkCA
Echinoderm Microtubule-
Fibrosarcoma (e.g., Pediatric



Associated Protein-Like 4
FibrosarcomaL), GlioblastomaP,




Colon CancerR


HOMER2-TrkC
Homer Protein Homolog 2
Soft Tissue Sarcoma


TFG-TrkC
TRK-Fused Gene
Soft Tissue Solitary Fibrous




Tumor


FAT1-TrkC
FAT Atypical Cadherin 1
Cervical Squamous Cell




CarcinomaB


MYO5A-TrkC
Myosin VA
Spitz tumorC


MYH9-TrkC
Myosin Heavy Chain 9
Spitz tumorC


KANK1-TrkCN
KANK1
Renal Metanephric Adenoma (MA)


SQSTM1-TrkCO
Sequestosome 1
Papillary Thyroid Carcinoma






ATannenbaum et al., Cold Spring Harb. Mol. Case Stud. 1: a000471, 2015.




BU.S. patent application Pub. No. 2015/0315657.




CYeh et al., J Pathol. 240(3): 282-90, 2016




DMontalli et al., J Oral Pathol Med. doi: 10.1111/jop.12491, 2016




EAlassiri et al., Am J Surg Pathol., August; 40(8): 1051-61, 2016.




FNagasubramanian et al., Pediatr Blood Cancer., August; 63(8): 1468-70, 2016.




GChintakuntlawar et al., Oral Surg Oral Med Oral Pathol Oral Radiol. 2016 May; 121(5): 542-549. e1.




HU.S. Pat. No. US9511050B2.




IU.S Pat. No. US9447135B2.




JSkalova et al., Modern Pathology 30, S27-S43, 2017.




KHyrcza et al., Vol. 469, Supp. Supplement 1, pp. S17. Abstract Number: OFP-1997-7; 31st International Congress of the International Academy of Pathology and the 28th Congress of the European Society of Pathology, Cologne, Germany. 25-29 Sep. 2016.




LSims et al., Journal of Immunotherapy of Cancer, Vol. 4, Supp. Supplement 1; Abstract Number: P280; 31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer, SITC 2016. National Harbor, MD; 9-13 Nov. 2016.




KRoberts et al., Blood, Vol. 128, No. 22. Abstract Number: 278, 58th Annual Meeting of the American Society of Hematology, ASH 2016. San Diego, CA, United States. 3 Dec. 2016-6 Dec. 2016.




MPavlick et al., Pediatr Blood Cancer, doi: 10.1002/pbc.26433, 2017.




MAndreasen et al., Am J Surg Pathol., November; 41(11): 1552-1560, 2017.




NCatic et al., Cancer Genet., August; 214-215: 9-15, doi: 10.1016/j.cancergen.2017.03.001, 2017.




OLu et al., Oncotarget, , July 11; 8(28): 45784-45792. doi: 10.18632/oncotarget.17412, 2017.




PSchram et al., Cancer Research, Vol. 77, No. 13, Supp. Supplement 1. Abstract Number: LB-302, American Association for Cancer Research Annual Meeting, Washington, DC, United States, 2017.




RCoebergh et al., Cancer Research, Vol. 77, No. 13, Supp. Supplement 1. Abstract Number: 490, American Association for Cancer Research Annual Meeting, Washington, DC, United States, 2017.







In some embodiments, the dysregulation of a TRK gene, a TRK kinase, or expression or activity or level of any of the same, includes at least one point mutation in a TRK gene that results in the production of a TRK kinase that has one or more amino acid substitutions, insertions, or deletions as compared to the wild-type TRK kinase.


In some embodiments, a TRK-associated cancer has been identified as having one or more TRK inhibitor resistance mutations (that result in an increased resistance to a TRK inhibitor.


In one embodiment, Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, is useful for treating diseases and disorders which can be treated with a TRK inhibitor. Non-limiting examples of cancer (e.g., a Trk-associated cancer) include adenocarcinoma, adrenal gland cortical carcinoma, adrenal gland neuroblastoma, anus squamous cell carcinoma, appendix adenocarcinoma, bladder urothelial carcinoma, bile duct adenocarcinoma, bladder carcinoma, bladder urothelial carcinoma, bone chordoma, bone marrow leukemia lymphocytic chronic, bone marrow leukemia non-lymphocytic acute myelocytic, bone marrow lymph proliferative disease, bone marrow multiple myeloma, bone sarcoma, brain astrocytoma, brain glioblastoma, brain medulloblastoma, brain meningioma, brain oligodendroglioma, breast adenoid cystic carcinoma, breast carcinoma, breast ductal carcinoma in situ, breast invasive ductal carcinoma, breast invasive lobular carcinoma, breast metaplastic carcinoma, cervix neuroendocrine carcinoma, cervix squamous cell carcinoma, colon adenocarcinoma, colon carcinoid tumor, duodenum adenocarcinoma, endometrioid tumor, esophagus adenocarcinoma, eye intraocular melanoma, eye intraocular squamous cell carcinoma, eye lacrimal duct carcinoma, fallopian tube serous carcinoma, gallbladder adenocarcinoma, gallbladder glomus tumor, gastroesophageal junction adenocarcinoma, head and neck adenoid cystic carcinoma, head and neck carcinoma, head and neck neuroblastoma, head and neck squamous cell carcinoma, kidney chromophore carcinoma, kidney medullary carcinoma, kidney renal cell carcinoma, kidney renal papillary carcinoma, kidney sarcomatoid carcinoma, kidney urothelial carcinoma, leukemia lymphocytic, liver cholangiocarcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, lung adenosquamous carcinoma, lung atypical carcinoid, lung carcinosarcoma, lung large cell neuroendocrine carcinoma, lung non-small cell lung carcinoma, lung sarcoma, lung sarcomatoid carcinoma, lung small cell carcinoma, lung small cell undifferentiated carcinoma, lung squamous cell carcinoma, lymph node lymphoma diffuse large B cell, lymph node lymphoma follicular lymphoma, lymph node lymphoma mediastinal B-cell, lymph node lymphoma plasmablastic lung adenocarcinoma, lymphoma follicular lymphoma, lymphoma, non-Hodgkin's lymphoma, nasopharynx and paranasal sinuses undifferentiated carcinoma, ovary carcinoma, ovary carcinosarcoma, ovary clear cell carcinoma, ovary epithelial carcinoma, ovary granulosa cell tumor, ovary serous carcinoma, pancreas carcinoma, pancreas ductal adenocarcinoma, pancreas neuroendocrine carcinoma, peritoneum mesothelioma, peritoneum serous carcinoma, placenta choriocarcinoma, pleura mesothelioma, prostate acinar adenocarcinoma, prostate carcinoma, rectum adenocarcinoma, rectum squamous cell carcinoma, skin adnexal carcinoma, skin basal cell carcinoma, skin melanoma, skin Merkel cell carcinoma, skin squamous cell carcinoma, small intestine adenocarcinoma, small intestine gastrointestinal stromal tumors (GISTs), soft tissue angiosarcoma, soft tissue Ewing sarcoma, soft tissue hemangioendothelioma, soft tissue inflammatory myofibroblastic tumor, soft tissue leiomyosarcoma, soft tissue liposarcoma, soft tissue neuroblastoma, soft tissue paraganglioma, soft tissue perivascular epitheliod cell tumor, soft tissue sarcoma, soft tissue synovial sarcoma, stomach adenocarcinoma, stomach adenocarcinoma diffuse-type, stomach adenocarcinoma intestinal type, stomach adenocarcinoma intestinal type, stomach leiomyosarcoma, thymus carcinoma, thymus thymoma lymphocytic, thyroid papillary carcinoma, unknown primary adenocarcinoma, unknown primary carcinoma, unknown primary malignant neoplasm, unknown primary melanoma, unknown primary sarcomatoid carcinoma, unknown primary squamous cell carcinoma, unknown undifferentiated neuroendocrine carcinoma, unknown primary undifferentiated small cell carcinoma, uterus carcinosarcoma, uterus endometrial adenocarcinoma, uterus endometrial adenocarcinoma endometrioid, uterus endometrial adenocarcinoma papillary serous, and uterus leiomyosarcoma.


Additional examples of cancers (e.g., Trk inhibitor-resistant cancer) include: adrenocortical carcinoma, anal cancer, appendix cancer, atypical teratoid/rhabdoid tumor (e.g., central nervous system atypical teratoid/rhabdoid tumor), B-cell cancer, bile duct cancer, bladder cancer, bone cancer (e.g., osteosarcoma and malignant fibrous histiocytoma), brain cancer (e.g., brain and spinal cord tumor, brain stem glioma, central nervous system embryonal tumors, central nervous system germ cell tumors, craniopharyngioma, and ependymoma), breast cancer, bronchogenic carcinoma, bronchus cancer, cancer of hematological tissues, cancer of the oral cavity or pharynx, carcinoid tumor, cervical cancer, childhood cancers, chordoma, chronic lymphocytic leukemia, chronic myeloproliferative neoplasms, colon cancer, colorectal cancer, cutaneous T-cell lymphoma, ductal carcinoma in situ, embryonal tumor, endometrial cancer, esophageal cancer, esthesioneuroblastoma, extracranial germ cell tumor, extragonadal germ cell tumor, extrahepatic bile duct cancer, eye cancer (e.g., retinoblastoma), fallopian tube cancer, fibrosarcoma, fibrous histiocytoma of bone, gallbladder cancer, gastric cancer, gastrointestinal carcinoid tumor, germ cell tumor, gestational trophoblastic disease, glioblastoma multiforme, glioma (e.g., lower-grade glioma), head and neck cancer, heart cancer, histiocytosis, hypopharyngeal cancer, inflammatory myofibroblastic tumors, intrahepatic cholangiocarcinoma, islet cell tumor, kidney cancer (e.g., renal cell cancer), Langerhans cell histiocytosis, large cell neuroendocrine cancer, laryngeal cancer, leukemia (e.g., acute lymphoblastic leukemia, acute myeloid leukemia, chronic myelogenous leukemia, and hairy cell leukemia), lip cancer, liver cancer, lung cancer, Burkitt lymphoma, Hodgkin's lymphoma, and primary central nervous system lymphoma), medulloblastoma, mesothelioma, mouth cancer, multiple myeloma, myelodysplastic syndromes, nasal cavity and paranasal sinus cancer, nasopharyngeal cancer, neoplasm (e.g., a melanocystic neoplasm), nephroma, neuroblastoma, non-small cell lung cancer, oral cancer, oropharyngeal cancer, ovarian cancer, pancreatic cancer, paraganglioma, parathyroid cancer, pediatric glioma, penile cancer, pharyngeal cancer, pheochromocytoma, pilocytic astrocytoma, pituitary tumor, plasma cell neoplasm, primary peritoneal cancer, prostate cancer, rectum carcinoma, salivary gland cancer, sarcoma (e.g., Ewing sarcoma, rhabdomyosarcoma, uterine sarcoma, and undifferentiated sarcoma), secretory breast carcinoma, Sezary syndrome, skin cancer, small bowel cancer, small cell lung cancer, small intestine cancer, Spitz nevi, Spitz tumors, spitzoid melanoma, stomach cancer, squamous cell carcinoma, squamous neck cancer, testicular cancer, throat cancer, thymoma and thymic carcinoma, thyroid carcinoma, urethral cancer, uterine cancer, urinary bladder cancer, vaginal cancer, vulvar cancer, and Wilms tumor.


In some embodiments, the cancer is a pediatric cancer. In some embodiments, the pediatric cancer is a mesenchymal cancer. For example, the mesenchymal cancer can be selected from the group consisting of: pediatric nephroma, congenital fibrosarcoma (CFS), pediatric high-grade glioma (HGG), mesenchymal cancers (infant fibrosarcoma (IF), congenital mesoblastic nephroma, congenital infantile fibrosarcoma (CIFS); pilocytic astrocytoma, brain tumors, pediatic acute leukemia, Ph-like acute lymphoblastic leukemia, cellular congenital mesoblastic nephroma (CMN); infantile fibrosarcoma, pediatric high-grade glioma (HGG), diffuse intrinsic pontine gliomas (DIPGs), non-brainstem HGGs (NBS-HGGs), anaplastic large cell lymphoma (ALCL), non-Hodgkin's lymphoma (NHL), pediatric papillary thyroid carcinoma, soft tissue sarcoma, spitzoid melanoma, pediatric hemangiopericytoma-like sarcoma, spindle cell sarcoma, NOS with myo/haemangiopericytic growth pattern, lung cancer, advanced pediatric solid tumors, neuroectodermal-derived tumors, pediatric colorectal cancer, adrenal neuroblastoma, and central nervous system tumors.


In some embodiments, the pediatric cancer is a fibrosarcoma such as infantile fibrosarcoma.


In some embodiments, the pediatric cancer is a glioma. For example, the pediatric cancer is selected from the group consisting of: pediatric high-grade glioma (HGG), diffuse intrinsic pontine gliomas (DIPGs), and on-brainstem HGGs (NB S-HGGs).


Provided herein are methods of treating a subject having a cancer (e.g., any of the cancers described herein) that include identifying a subject having a cancer cell that has at least one point mutation in a NTRK gene that results in the expression of a Trk protein including a mutation at one or more of the amino acid positions shown in Tables 4, 5, 6 or 7, and administering to the identified subject Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein.


Also provided herein are methods of treating a subject that include administering a therapeutically effective amount of Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, to a subject having a clinical record that indicates that the subject has a cancer cell that has at least one point mutation in a NTRK gene that results in the expression of a Trk protein including a mutation at one or more amino acid positions (e.g., a mutation at one or more of the amino acid positions shown in Tables 4, 5, 6 or 7).


Also provided herein are methods of treating a subject having a cancer (e.g., any of the cancers described herein or known in the art) that include: identifying a subject having a cancer cell that has at least one point mutation in a NTRK gene that results in the expression of a Trk protein including a mutation at one or more amino acid positions (e.g., a mutation at one or more of the amino acid positions shown in Tables 4, 5, 6 or 7); and administering to the identified subject Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein.


Also provided herein are methods of treating a subject having a cancer (e.g., any of the cancers described herein or known in the art) that include: identifying a subject having a cancer cell that has at least one point mutation in a NTRK gene that results in the expression of a Trk protein including a mutation at one or more amino acid positions (e.g., a mutation at one or more of the amino acid positions shown in Tables 4, 5, 6 or 7); and administering to the identified subject Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, and another anticancer agent (e.g., any one or more of the anticancer agents described herein) or anticancer therapy (e.g., any one or more of the anticancer therapies provided herein.


Also provided herein are methods of treating a subject that include administering a therapeutically effective amount of Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, to a subject having a clinical record that indicates that the subject has a cancer cell that has at least one point mutation in a NTRK gene that results in the expression of a Trk protein including a mutation at one or more amino acid positions (e.g., a mutation at one or more of the amino acid positions shown in Tables 4, 5, 6 or 7).


Also provided herein are methods of treating a subject that include administering a therapeutically effective amount of Compound 1 or a solid form thereof, crystalline form thereof, or solvate or hydrate thereof, or a salt of Compound 1 or solid form thereof, crystalline form thereof, or solvate or hydrate thereof, as described herein, and another anticancer agent (e.g., any one or more of the anticancer agents described herein) or anticancer therapies (e.g., any one or more of the anticancer therapies described herein), to a subject having a clinical record that indicates that the subject has a cancer cell that has at least one point mutation in a NTRK gene that results in the expression of a Trk protein including a mutation at one or more amino acid positions (e.g., a mutation at one or more of the amino acid positions shown in Tables 4, 5, 6 or 7).


In some embodiments, the dysregulation of a NTRK gene, a Trk protein, or expression or activity, or level of the same, includes one or more deletions, insertions, or point mutation(s) in a Trk protein. In some embodiments, the dysregulation of a NTRK gene, a Trk protein, or expression or activity, or level of the same, includes a deletion of one or more residues from the TrkA protein, resulting in constitutive activity of the Trk kinase domain. In some embodiments, the dysregulation of a NTRK gene, a Trk protein, or expression or activity, or level of the same, includes at least one point mutation in a NTRK1 gene that results in the production of a TrkA protein that has one or more amino acid substitutions as compared to the wildtype TrkA protein (see, for example, the point mutations listed in Tables 4 and 5). An exemplary wildtype TrkA polypeptide is SEQ ID NO: 1, an exemplary wildtype TrkB polypeptide is SEQ ID NO: 2, and an exemplary TrkC polypeptide is SEQ ID NO: 3.









TABLE 4







Activating TrkA Point Mutations









Mutation
Pediatric Cancer
Reference













C6773T, C7232T,
TrkA
neuroblastoma
Scaruffi et al.,


C7301T



Int. J. Oncol.






14: 935-938, 1999
















TABLE 5







Activating TrkA Point MutationsA











Exemplary Isoform in




which Mutation is


Point Mutation
Rationale
Present (if known)





R6WI




R33WB

NP_001007793.1F


A336E
Near NGF Binding Site
Reference TrkA sequence


A337T
Near NGF Binding Site
Reference TrkA sequence


R324Q or
Near NGF Binding Site
Unknown


R324W


V420M
Close to Membrane
Reference TrkA sequence


R444Q or
Close to Membrane
Reference TrkA sequence


R444W


G517R or
P-Loop
Reference TrkA sequence


G517V


K538A
Activating
Reference TrkA sequence


V573ME

Reference TrkA sequence


R583HI


F589LE

Reference TrkA sequence


G595R or
Catalytic Domain
Reference TrkA sequence


G667CD


F598LE

Unknown


R649W or
Arginine may stabilize
Reference TrkA sequence


R649L
auto-inhibited



conformation.


R682S
Activation Loop
Reference TrkA sequence


V683G
Activation Loop
Reference TrkA sequence


I699VH


Q627XC,

NP_001012331.1G,


Q597XC,

NP_001007793.1F, and


Q633XC

Reference TrkA sequence,




respectively


R702C
Exposed, may form face-
Reference TrkA sequence



to-face disulfide



linked dimer


R744HI






AReference TrkA sequence is UniProtKB/Swiss-Prot: P04629.4, and can be found at URL: www.ncbi-nlm.nih.gov/protein/94730402?report=genbank&log$=protalign&blast_rank=0&RID=0 (SEQ ID NO: 1)




BZhang et al., Blood 124(21): 1682, 2014. Mutation found in T-cell prolymphocytic leukemia.




CPark et al., Proc. Natl. Acad. Sci. U.S.A. 112(40): 12492-12497, 2015. Mutation found in colorectal cancer.




DRusso et al., Cancer Discov. January; 6(1): 36-44, 2016.




EPCT Application No. WO2016196141A1.




Fwww.ncbi.nlm.nih.gov/protein/56118210?report=genbank&log$=protalign&blast_rank=3&RID=0




Gwww.ncbi.nlm.nih.gov/protein/59889558




HDeihimi et al., Oncotarget. June 20; 8(25): 39945-39962. doi: 10.18632/oncotarget. 18098, 2017.




IIniguez-Ariza et al., Journal of Clinical Oncology, (20 Jun. 2017) Vol. 35, No. 15, Supp. 1, 2017 Annual Meeting of the American Society of Clinical Oncology, ASCO, 2017.







In some embodiments, the dysregulation of a NTRK gene, a Trk protein, or expression or activity, or level of the same, includes a splice variation in a TrkA mRNA which results in an expressed protein that is an alternatively spliced variant of TrkA having at least one residue deleted (as compared to a wild-type TrkA protein) resulting in constitutive activity of the TrkA kinase domain. In some embodiments, an alternatively spliced form of TrkA with constitutive activity has deletions of exons 8, 9, and 11 resulting in an expressed protein missing residues 192-284 and 393-398 relative to TrkA Isoform 2, has a deletion of exon 10 in TrkA, or has a deletion in a NTRK1 gene that encodes a TrkA protein with a 75 amino acid deletion in the transmembrane domain (Reuther et al., Mol. Cell Biol. 20:8655-8666, 2000).


In some embodiments, the dysregulation of a NTRK gene, a Trk protein, or expression or activity, or level of the same, includes at least one point mutation in a NTRK1 gene that results in the production of a TrkB protein that has one or more amino acid substitutions as compared to the wildtype TrkB protein (see, for example, the point mutations listed in Table 6.









TABLE 6







Activating TrkB Point MutationsA













Exemplary Isoform in





which Mutation is



Point Mutation
Rationale
Present (if known)







A13TC

Reference TrkB sequence



E142KC

Reference TrkB sequence



R136HC

Reference TrkB sequence



S167YF



V619MB

Unknown



F633LB

NP_006171.2D





(Corresponding to





position 617 of Reference





TrkB sequence)



G639RB

NP_006171.2D





(Corresponding to





position 623 of Reference





TrkB sequence)



G709C or

NP_006171.2D



G709A or

(Corresponding to



G709SB

position 693 of Reference





TrkB sequence)



P716SE








AReference TrkB sequence is UniProtKB/Swiss-Prot: Q16620.1, and can be found at URL: www.ncbi.nlm.nih.gov/protein/2497560?report=genbank&log$=protalign&blast_rank=0&RID=0 (SEQ ID NO: 2)





BPCT Application No. WO2016196141A1.





CBonanno et al., Journal of Thoracic Oncology, Vol. 11, No. 4, Supp. Suppl. 1, pp S67. Abstract Number: 28P; 6th European Lung Cancer Conference, ELCC 2016, Geneva, Switzerland.





Dwww.ncbi.nlm.nih.gov/protein/NP_006171.2





EDeihimi et al., Oncotarget. Jun. 20; 8(25): 39945-39962. doi: 10.18632/oncotarget.18098, 2017.





FIniguez-Ariza et al., Journal of Clinical Oncology, (20 Jun. 2017) Vol. 35, No. 15, Supp. 1, 2017 Annual Meeting of the American Society of Clinical Oncology, ASCO, 2017.







In some embodiments, the dysregulation of a NTRK gene, a Trk protein, or expression or activity, or level of the same, includes at least one point mutation in a NTRK1 gene that results in the production of a TrkC protein that has one or more amino acid substitutions as compared to the wildtype TrkC protein (see, for example, the point mutations listed in Table 7.









TABLE 7







Activating TrkC Point MutationsA











Exemplary Isoform in




which Mutation is


Point Mutation
Rationale
Present (if known)





V603MC

NP_001007157.1D


F617LC

Reference TrkC sequence


G623RB,C
Steric Hinderance
Reference TrkC sequence


G696C or G696A

Reference TrkC sequence


or G696SC


R745LE


I749MF






AReference TrkC sequence is UniProtKB/Swiss-Prot: Q16288.2, and can be found at URL: www.ncbi.nlm.nih.gov/protein/134035335?report=genbank&log$=protalign&blast_rank=0&RID=0 (SEQ ID NO: 3)




BDrilon et al., Ann Oncol. 2016 May; 27(5): 920-6. doi: 10.1093/annonc/mdw042. Epub 2016 Feb. 15.




CPCT Application No. WO2016196141A1.




Dwww.ncbi.nlm.nih.gov/protein/NP_001007157




EDeihimi et al., Oncotarget. June 20; 8(25): 39945-39962. doi: 10.18632/oncotarget.18098, 2017.




FIniguez-Ariza et al., Journal of Clinical Oncology, (20 Jun. 2017) Vol. 35, No. 15, Supp. 1, 2017 Annual Meeting of the American Society of Clinical Oncology, ASCO, 2017.







ORIGINAL LIST OF REFERENCES (BELIEVED TO BE APPLICABLE TO ALL TABLES)




  • 1 Wiesner et al., Nature Comm. 5:3116, 2014.


  • 2 Vaishnavi et al., Nature Med. 19:1469-1472, 2013.


  • 3 Greco et al., Mol. Cell. Endocrinol. 28:321, 2010.


  • 4 Kim et al., PloS ONE 9(3): e91940, 2014.


  • 5 Vaishnavi et al., Nature Med. 19:1469-1472, 2013.


  • 6 Fernandez-Cuesta et al., “Cross-entity mutation analysis of lung neuroendocrine tumors sheds light into their molecular origin and identifies new therapeutic targets,” AACR Annual Meeting 2014, Abstract, April 2014.


  • 7 Stransky et al., Nature Comm. 5:4846, 2014.


  • 8 Ross et al., Oncologist 19:235-242, 2014.


  • 9 Doebele et al., J. Clin. Oncol. 32:5s, 2014.


  • 10 Jones et al., Nature Genetics 45:927-932, 2013.


  • 11 Wu et al., Nature Genetics 46:444-450, 2014.


  • 12 WO 2013/059740


  • 13 Zheng et al., “Anchored multiplex PCR for targeted next-generation sequencing,” Nature Med., published online on Nov. 10, 2014.


  • 14 Caria et al., Cancer Genet. Cytogenet. 203:21-29, 2010.


  • 15 Frattini et al., Nature Genet. 45:1141-1149, 2013.


  • 16 Martin-Zanca et al., Nature 319:743, 1986.


  • 17 Meyer et al., Leukemia 21: 2171-2180, 2007.


  • 18 Reuther et al., Mol. Cell. Biol. 20:8655-8666, 2000.


  • 19 Marchetti et al., Human Mutation 29(5):609-616, 2008.


  • 20 Tacconelli et al., Cancer Cell 6:347, 2004.


  • 21 Walch et al., Clin. Exp. Metastasis 17: 307-314, 1999.


  • 22 Papatsoris et al., Expert Opin. Invest. Drugs 16(3):303-309, 2007.


  • 23 Van Noesel et al., Gene 325: 1-15, 2004.


  • 24 Zhang et al., Oncology Reports 14: 161-171, 2005.


  • 25 Truzzi et al., J. Invest. Dermatol. 128(8):2031, 2008.


  • 26 Kolokythas et al., J. Oral Maxillofacial Surgery 68(6):1290-1295, 2010.


  • 27 Ni et al., Asian Pacific Journal of Cancer Prevention 13:1511, 2012.



In some embodiments, a TRK-associated cancer has been identified as having one or more TRK inhibitor resistance mutations (that result in an increased resistance to a TRK inhibitor. Non-limiting examples of TRK inhibitor resistance mutations are listed in Tables 8-10.









TABLE 8





Exemplary TrkA Resistance Mutations

















Amino acid position 517 (e.g., G517R)



Amino acid position 542 (e.g., A542V)



Amino acid position 564 (e.g., L5645H2)



Amino acid position 568 (e.g., Q568x)



Amino acid position 573 (e.g., V573M)



Amino acid position 589 (e.g., F589L, F589C)



Amino acid position 595 (e.g., G595S, G595R1, G595L2)



Amino acid position 599 (e.g., D596V)



Amino acid position 600 (e.g., F600L)



Amino acid position 602 (e.g., R602x)



Amino acid position 646 (e.g., F646V, F646I2)



Amino acid position 656 (e.g., C656Y, C656F)



Amino acid position 657 (e.g., L657V)



Amino acid position 667 (e.g., G667C1, G667S)



Amino acid position 676 (e.g., Y676S)



Amino acid position 679 (e.g., D679G2)








1Russo et al., Acquired Resistance to the TRK Inhibitor Entrectinib in Colorectal Cancer, Cancer Discov., January; 6(l): 36-44, 2016.





2Fuse et al., Mechanisms of Resistance to NTRK Inhibitors and Therapeutic Strategies in NTRK1-Rearranged Cancers, Mol. Cancer Ther., . January; 6(1): 36-44, 2016.














TABLE 9





Exemplary TrkB Resistance Mutations

















Amino acid position 545 (e.g., G545R)



Amino acid position 570 (e.g., A570V)



Amino acid position 596 (e.g., Q596E, Q596P)



Amino acid position 601 (e.g., V601G)



Amino acid position 617 (e.g., F617L, F617C, F617I)



Amino acid position 623 (e.g., G623S, G623R)



Amino acid position 624 (e.g., D624V)



Amino acid position 628 (e.g., F628x)



Amino acid position 630 (e.g., R630K)



Amino acid position 639 (e.g., G639R)1



Amino acid position 672 (e.g., F672x)



Amino acid position 682 (e.g., C682Y, C682F)



Amino acid position 683 (e.g., L683V)



Amino acid position 693 (e.g., G693S)



Amino acid position 702 (e.g., Y702x)








1PCT Application No. WO2017155018A1.














TABLE 9





Exemplary TrkC Resistance Mutations

















Amino acid position 545 (e.g., G545R)



Amino acid position 570 (e.g., A570V)



Amino acid position 596 (e.g., Q596x)



Amino acid position 601 (e.g., V601)



Amino acid position 617 (e.g., F617x, F617L)



Amino acid position 623 (e.g., G623R1)



Amino acid position 624 (e.g., D624V)



Amino acid position 628 (e.g., F628x)



Amino acid position 630 (e.g., R630x)



Amino acid position 675 (e.g., F675x)



Amino acid position 685 (e.g., C685Y, C684F)



Amino acid position 686 (e.g., L686V)



Amino acid position 696 (e.g., G696x, G696A)



Amino acid position 705 (e.g., Y705x)








1Drilon et al., What hides behind the MASC: clinical response and acquired resistance to entrectinib after ETV6-NTRK3 identification in a mammary analogue secretory carcinoma (MASC), Ann Oncol. 2016 May; 27(5): 920-6. doi: 10.1093/annonc/mdw042. Epub 2016 Feb. 15.








The letter “x” when used to describe a mutation of an amino acid at a specific amino acid position means (i) a substitution of the amino acid present at the same amino acid position in the corresponding wildtype protein with a different naturally-occurring amino acid, or (ii) a deletion of the amino acid present at the same amino acid position in the corresponding wildtype protein.


Pharmaceutical Compositions, Formulations, Routes of Administration


In some embodiments, provided herein is a process for preparing a pharmaceutical composition comprising mixing (i) a compound of any one of formulae described herein or salt thereof prepared according to any of the processes described herein, and (ii) a pharmaceutically acceptable carrier. Pharmaceutical compositions containing the compound of any one of formulae described herein or a salt thereof as the active ingredient can be prepared by intimately mixing the compound of any one of formulae described herein or a salt thereof with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending upon the desired route of administration (e.g., oral, parenteral). Thus, for liquid oral preparations such as suspensions, elixirs and solutions, suitable carriers and additives include water, glycols, oils, alcohols, flavoring agents, preservatives, stabilizers, coloring agents, and the like; for solid oral preparations, such as powders, capsules and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like. Solid oral preparations can also be coated with substances such as sugars or be enteric-coated so as to modulate major site of absorption. For parenteral administration, the carrier will usually consist of sterile water, and other ingredients can be added to increase solubility or preservation. Injectable suspensions or solutions can also be prepared utilizing aqueous carriers along with appropriate additives.


The pharmaceutical compositions herein contain, per unit dosage unit, e.g., tablet, capsule, suspension, solution, sachet for reconstitution, powder, injection, I.V., suppository, sublingual/buccal film, teaspoonful, and the like, of from about 0.1-1000 mg or any range therein, and may be given at a dosage of from about 0.01-300 mg/kg/day, or any range therein, preferably from about 0.5-50 mg/kg/day, or any range therein. In some embodiments, the pharmaceutical compositions provided herein contain, per unit dosage unit, about 25 mg to about 500 mg of a compound provided herein (for example, about 25 mg to about 400 mg, about 25 mg to about 300 mg, about 25 mg to about 250 mg, about 25 mg to about 200 mg, about 25 mg to about 150 mg, about 25 mg to about 100 mg, about 25 mg to about 75 mg, about 25 mg to a about 50 mg, about 50 mg to about 500 mg, about 100 mg to about 500 mg, about 150 mg to about 500 mg, about 200 mg to about 500 mg, about 250 mg to about 500 mg, about 300 mg to about 500 mg, about 400 mg to about 500 mg, about 50 to about 200 mg, about 100 to about 250 mg, about 50 to about 150 mg). In some embodiments, the pharmaceutical compositions provided herein contain, per unit dosage unit, about 25 mg, about 50 mg, about 100 mg, about 150 mg, about 200 mg, about 250 mg, about 300 mg, about 400 mg, or about 500 mg of Compound 1 or any one of crystalline forms, solid forms, solvates, hydrates or salts described herein. The dosages, however, can be varied depending upon the requirement of the patient, the severity of the condition being treated, and/or (if applicable) the crystalline form, solid form, solvate, hydrate or salt being employed. In some embodiments, the dosages are administered once daily (QD) or twice daily (BID). Preferably, these compositions are in unit dosage forms, such as sterile solutions or suspensions for oral administration.


To prepare the pharmaceutical compositions provided herein, the compound of any one of formulae described herein or a salt thereof as the active ingredient is intimately admixed with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration (e.g., oral or parenteral). Any one of crystalline forms, solid forms, solvates, hydrates or salts described herein can be administered by any convenient route, e.g., into the gastrointestinal tract (e.g., rectally or orally), the nose, lungs, musculature or vasculature, or transdermally or dermally. Any one of crystalline forms, solid forms, solvates, hydrates or salts described herein can be administered in any convenient administrative form, e.g., tablets, powders, capsules, solutions, dispersions, suspensions, syrups, sprays, suppositories, gels, emulsions, patches, etc. Such compositions can contain components that are conventional in pharmaceutical preparations, e.g., diluents, carriers, pH modifiers, sweeteners, bulking agents, and further active agents. If parenteral administration is desired, the compositions will be sterile and in a solution or suspension form suitable for injection or infusion. Such compositions form a further aspect of the present disclosure.


In preparing the compositions in oral dosage form, any of the usual pharmaceutical media can be employed. Thus, for liquid oral preparations, such as, for example, suspensions, elixirs, and solutions, suitable carriers and additives include water, glycols, glycerols, oils, cyclodextrins, alcohols, e.g., ethanol, flavoring agents, preservatives, coloring agents, and the like; for solid oral preparations such as, for example, powders, capsules, caplets, gelcaps and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like. Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.


Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are employed. If desired, tablets can be sugar coated or enteric coated by standard techniques. For parenteral formulations, the carrier will usually comprise sterile water, through other ingredients, for example, for purposes such as aiding solubility or for preservation, can be included. In some embodiments, the carrier is 0.8% saline or a 5% dextrose. Injectable suspensions can also be prepared, in which case appropriate liquid carriers, suspending agents, and the like can be employed. The pharmaceutical compositions herein can contain, per dosage unit, e.g., tablet, capsule, powder, injection, teaspoonful, and the like, an amount of the active ingredient necessary to deliver an effective dose as described above.


In some embodiments, the dosages are administered once daily (QD) or twice daily (BID). Alternatively, the composition can be presented in a form suitable for once-weekly or once-monthly administration. For preparing solid compositions such as tablets, any one of crystalline forms, solid forms, solvates, hydrates or salts described herein is mixed with a pharmaceutical carrier, e.g., conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g., water, to form a solid composition containing any one of crystalline forms, solid forms, solvates, hydrates or salts described herein. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 1000 mg, or any amount or range thereof, of the active ingredient provided herein. The tablets or pills of the composition can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of material can be used for such enteric layers or coatings, such materials including a number of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.


The liquid forms in which the compositions provided herein can be incorporated for administration orally or by injection include aqueous solutions, cyclodextrins, suitably flavored syrups, aqueous or oil suspensions, and flavored emulsions with edible oils, such as cottonseed oil, sesame oil, coconut oil, or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions include synthetic and natural gums, such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinyl-pyrrolidone, and gelatin. For parenteral administration, sterile suspensions and solutions are desired. Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired.


Any one of crystalline forms, solid forms, solvates, hydrates or salts described herein can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of a transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.


Suitable pharmaceutically acceptable carriers are well known in the art. Descriptions of some of these pharmaceutically acceptable carriers may be found in The Handbook of Pharmaceutical Excipients, published by the American Pharmaceutical Association and the Pharmaceutical Society of Great Britain.


Methods of formulating pharmaceutical compositions have been described in numerous publications, such as Pharmaceutical Dosage Forms: Tablets, Second Edition, Revised and Expanded, Volumes 1-3, edited by Lieberman et al.; Pharmaceutical Dosage Forms: Parenteral Medications, Volumes 1-2, edited by Avis et al.; and Pharmaceutical Dosage Forms: Disperse Systems, Volumes 1-2, edited by Lieberman et al.; published by Marcel Dekker, Inc.


Compounds provided herein can be administered in any of the foregoing compositions and according to dosage regimens established in the art whenever treatment of cancer, pain, inflammation, neurodegenerative disease or Trypanosoma cruzi infection is required.


The daily dosage of the compound of any one of formulae described herein or a salt thereof can be varied over a wide range from 1.0 to 10,000 mg per adult human per day, or higher, or any range therein. For oral administration, the compositions are preferably provided in the form of tablets containing 0.01, 0.05, 0.1, 0.5, 1.0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 150, 200, 250 or 500 milligrams of the active ingredient for the symptomatic adjustment of the dosage to the patient to be treated. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.1 mg/kg to about 1000 mg/kg of body weight per day, or any range therein. Preferably, the range is from about 0.5 to about 500 mg/kg of body weight per day, or any range therein. More preferably, from about 1.0 to about 250 mg/kg of body weight per day, or any range therein. More preferably, from about 0.1 to about 100 mg/kg of body weight per day, or any range therein. In an example, the range can be from about 0.1 to about 50.0 mg/kg of body weight per day, or any amount or range therein. In another example, the range can be from about 0.1 to about 15.0 mg/kg of body weight per day, or any range therein. In yet another example, the range can be from about 0.5 to about 7.5 mg/kg of body weight per day, or any amount to range therein. The compound of any one of formulae described herein or a salt thereof can be administered on a regimen of 1 to 4 times per day or in a single daily dose.


Optimal dosages to be administered can be readily determined by those skilled in the art, and can vary with the mode of administration, the strength of the preparation, the mode of administration, and the advancement of the disease condition. In addition, factors associated with the particular patient being treated, including patient age, weight, diet, and time of administration, can result in the need to adjust dosages.


EXAMPLES

Materials and methods for the preparation of compounds, crystalline forms, solid forms, solvates, hydrates, and salts thereof.


Example A

1) Preparation of Compound 1 (the Compound of Formula I)




embedded image


(R,E)-N-((5-fluoro-2-methoxypyridin-3-yl) methylene)-2-methylpropane-2-sulfinamide (2): A flask (equipped with a nitrogen inlet, overhead stirring, and thermocouple) was charged with DCM (3 L, 10 vol). The mixture was agitated, and the mixture was deoxygenated with subsurface nitrogen for 1 h. Next 5-fluoro-2-methoxynicotinaldehyde (1) (300 g, 1934 mmol) and (R)-2-methylpropane-2-sulfinamide (246 g, 2031 mmol) were charged. The Cs2CO3 (441 g, 1354 mmol) was charged in portions, with agitation, over several minutes. The reaction was agitated overnight at ambient temperature under nitrogen. The reaction was sampled and analyzed by HPLC for reaction completion. A 15 wt % solution of the citric acid (in water) was prepared (using 1.5 eq of citric acid based on the Cs2CO3 input). This solution was charged into the reactor with the reaction mixture, using an addition funnel. The charge was done in portions. The biphasic mixture was transferred to a separatory funnel, and the lower DCM layer was removed. The upper aqueous layer was removed and discarded. The DCM layer was transferred back into the separatory funnel, and washed with saturated brine (2 L). Again, the lower DCM layer was removed, and the upper aqueous layer was discarded. The DCM layer was concentrated under vacuum (rotovap) to give the desired product.




embedded image


(S)—N—((S)-3-(1,3-dioxan-2-yl)-1-(5-fluoro-2-methoxypyridin-3-yl)propyl)-2-methylpropane-2-sulfinamide (5): A flask (equipped with a nitrogen inlet, overhead stirring, reflux condenser, thermocouple, and addition funnel) was charged with Mg turnings (565 g, 23.2 moles) followed by THF (24 L, 8 vol) under nitrogen. This mixture was agitated and warmed to ˜30° C. When the internal temperature was 29.9° C., DIBAL (31.2 mL, 0.004 eq.) was added. A separate flask was charged with 2-(2-bromoethyl)-1,3-dioxane (4531 g, 23.2 moles) and THF (15.9 L, 5.3 vol). The mixture was agitated at ambient temperature to dissolve. The reaction flask with the Mg/Dibal-H mixture was slowly charged with the 2-(2-bromoethyl)-1,3-dioxane (3)/THF solution via an addition funnel. The charge was made in portions over ˜5 h. The bromide solution was added so that the internal temperature did not rise above 50° C. The reaction mixture was then held for 45 minutes. After the 45-minute hold, the active Grignard mixture was cooled to −30 to −40° C. (dry ice/acetonitrile). A separate flask was charged with the (R,E)-N-((5-fluoro-2-methoxypyridin-3-yl)methylene)-2-methylpropane-2-sulfinamide (3000 g, 11.6 moles), followed by THF (5.1 L, 1.7 vol). Using an addition funnel, the starting material solution was portion-wise transferred at ambient temperature into the Grignard mixture over ˜2 h and the internal temp was kept at −37.3 to −28.9° C. The reaction mixture was agitated at low temperature and analyzed by HPLC for reaction completion. To quench the reaction, a 15 wt % solution of citric acid (˜11 vol) was charged into a round bottom flask and cooled with an ice bath to ˜10° C. The reaction mixture was transferred into the citric acid solution in portions. When the transfer was complete, the mixture was allowed to stir for ˜15 minutes. MTBE (9 L, 3 vol) was charged into the mixture and then the entire mixture was transferred to a separatory funnel. The reaction flask was rinsed with MTBE (3 L, 1 vol) and transferred to the separatory funnel. The biphasic mixture was agitated for 5 minutes and then the phases were allowed to settle. The layers were separated, and the bottom aqueous layer was back extracted with additional MTBE (16 L, ˜5 vol). After mixing and settling, the layers were separated. The MTBE layers were combined and washed with sat. brine (15 L, 5 vol). After mixing and settling, the aqueous layer was discarded. The MTBE layer was concentrated under vacuum. MTBE (6 L, 2 vol) was charged, and the product was dissolved with agitation at ambient temperature. To the MTBE solution, heptane (30 L, 10 vol) was charged over ˜1 h. The slurry was allowed to agitate at ambient temperature overnight, and then filtered through polypropylene filter cloth. The cake was rinsed with heptane (9 L, 3 vol), and the wet solid 5 was dried in trays under vacuum at ˜50° C. to constant weight.




embedded image


(R)-5-fluoro-2-methoxy-3-(pyrrolidin-2-yl) pyridine (7): A flask (equipped with mechanical stirring, N2 inlet, condenser and J-Kem) was charged with 5 (1993 g, 5322 mmol) 2,2,2-trifluoroacetic acid (7971 mL) and water (1918 mL). The reaction was sampled to monitor completion of the deprotection by HPLC. After the reaction was judged to be complete, the reaction was charged with triethylsilane (2550 mL, 16.0 moles) via addition funnel over ˜1 h. The reaction mixture was stirred at ambient temperature overnight and the solvent was removed under vacuum with heating to 45-50° C. The resulting product was added to a 100 L separatory funnel and was diluted with MTBE (15 L) and water (15 L). The layers were agitated and the separated layers were dropped into tared carboys (Aq 1 and MTBE 1). The MTBE layer was added back to the separatory funnel and was back extracted with 6000 mL 1 M HCl. After mixing, the separated layers were dropped into tared carboys (Aq2 and MTBE 2). The aqueous layers were combined in the separatory funnel. To the aqueous layer was added DCM (16 L). To the mixture was added 50 wt % NaOH (˜900 mL) to reach pH≥12. After mixing, the organic layer was dropped into a tared flask (DCM 1). The aqueous layer was extracted with DCM (16 L) and the organic layer was dropped into a tared flask. The aqueous layer was extracted a third time with DCM (8 L). The organic layer was dropped into a tared flask (DCM 3). The combined organic layers were transferred to the separatory funnel and washed with sat. brine (9 L). The layers were separated and the organic layers were dropped and then the solvent was removed under vacuum to isolated the product.




embedded image


Ethyl 5-hydroxypyrazolo[1,5-a]pyrimidine-3-carboxylate: To a reactor was charged K3PO4 (4104 g granular, 19.3 moles), ethyl 3-amino-1H-pyrazole-4-carboxylate (2000 g, 12.9 moles), and DMF (18.8 kg) and the mixture was agitated. After 20 min, (E)-ethyl 3-ethoxyacrylate, (2230 g, 15.5 moles) was added and the mixture was heated to 110-115° C. internal temperature (IT). After the reaction was judged to be complete based on consumption of starting material, heating was ceased. The mixture was allowed to stir and cool overnight. Aqueous hydrochloric acid (3 M, 13 L) was added over ˜2 h. DI water (6 L) was added and the mixture was allowed to stir overnight. The mixture was filtered through polypropylene filter cloth (PPFC) and the residue was washed with water (3×5 vol, 3×10 L). The solid was placed in trays and oven dried under vacuum at 55° C. for 3 days and then 45° C. for 4 days to constant weight of (2553 g 95.6%).




embedded image


Ethyl 5-chloropyrazolo[1,5-a]pyrimidine-3-carboxylate (8): To a flask, under nitrogen, outfitted with mechanical stirring, J-Kem temperature probe, and condenser was added ethyl 5-hydroxypyrazolo[1,5-a]pyrimidine-3-carboxylate (2319 g, 11.2 moles), acetonitrile (9200 mL), and phosphoryl trichloride (1230 mL, 13.4 mmol). The reaction mixture was heated to ˜74° C. (IT) until it was judged complete by HPLC. The reaction was cooled to ˜30° C. While cooling, a separate flask was outfitted with mechanical stirring and a J-Kem temperature probe. Water (37 L) was added to this and the water was cooled to below 15° C. The reaction mixture was added portion-wise producing a mixture. The chlorination reactor was rinsed with 4:1 water/MeCN (2 L) and the rinse was added to the mixture. To the mixture was added MeCN (1 L) The transfer line was rinsed with 4:1 water/MeCN (2 L), and the rinse was added to the mixture. The mixture was cooled back to below 20° C. and a solution of tribasic phosphate (2312 g, 10.9 mol) in water (4.0 L) was added portion-wise at a rate to keep the IT below 25° C. The slurry was stirred at ambient temperature overnight. The slurry was filtered (PPFC) and rinsed with 4:1 water/MeCN (6 L). The cake was rinsed a second time with water (7.0 L). The solid was placed in trays and dried in a vacuum oven at 50° C. for 36-72 h to give 8.




embedded image


Ethyl (R)-5-(2-(5-fluoro-2-methoxypyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylate (9): Combined triethylamine (1187 mL, 8518 mmol), (R)-5-fluoro-2-methoxy-3-(pyrrolidin-2-yl) pyridine (7) (889 g, 4259 mmol) in EtOH (200 proof, 5 mL/g, 4.4 L) and then ethyl 5-chloropyrazolo[1,5-a]pyrimidine-3-carboxylate (8)(1001 g, 4259 mmol) were added. The reaction was stirred overnight at ambient temp (19 h). The next day, water (10 mL/g, 8.9 L) was added and after stirring at room temperature for 2 h it was filtered through polypropylene filter cloth (PPFC), 23° C. and washed with 2:1 water:EtOH (2×1.8 L) then heptane (1.8 L). The product was placed in trays and dried under vacuum (with N2 bleed) at 55° C. to give 9.




embedded image


Ethyl (R)-5-(2-(5-fluoro-2-oxo-1,2-dihydropyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylate (10): A solution of 4 M HCl in dioxane (1.0 L) was added to a flask containing (R)-ethyl 5-(2-(5-fluoro-2-methoxypyridin-3-yl) pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylate (9) (2500 g, 6486.9 mmol). The mixture was heated to 60° C. with an outlet at the top of the condenser (not under nitrogen). Once complete by HPLC, it was put under nitrogen and allowed to cool to room temperature with stirring overnight. The next day 20% K3PO4 (aq) (19 L, 7.5 mL/g—made by diluting 3800 g of K3PO4 to 19 kg total with water), was added. Once the temp was <35° C., EtOAc (12.5 L, 5 mL/g) was added and stirring continued for another 30 min. The mixture was pumped into a separatory vessel, and the aqueous layer dropped. The organic layer was concentrated under vacuum (rotovap) and the product was dried on vacuum pump at ambient temp to give 10.




embedded image


ethyl (R)-5-(2-(5-fluoro-2-(((trifluoromethyl) sulfonyl)oxy)pyridin-3-yl) pyrrolidin-1-yl) pyrazolo[1,5-a]pyrimidine-3-carboxylate (11): To a DCM solution of 10 was added triethylamine (1467 mL, 105.2 mol) and the mixture cooled to <5° C. Trifluoromethanesulfonic anhydride (1930 g, 684.0 mol) was added in portions maintaining temp <15° C. After 1 h reaction time sat. NaHCO3 (5 mL/g, 11 L) was added The mixture was stirred for 1 h and was then transferred to a separatory vessel with DCM and the layers were separated. The organic layer was washed with NaHCO3 (11 L). The organic layer was concentrated to minimum volume and solvent-swapped to MeOH (target MeOH volume about 10 L). The MeOH solution was added to a flask containing 1:1 MeOH:water (20 L), the suspension was stirred at room temperature for 2 h, filtered, and washed with 1:2 water:MeOH (2×2 mL/g). The solid was oven dried under vacuum at 55° C. until constant weight, to give 11.


N-Phenyl-bis(trifluoromethanesulfonimide) may be used instead of Trifluoromethanesulfonic anhydride to provide 11.




embedded image


ethyl 5-((R)-2-(2-((R)-3-aminobut-1-yn-1-yl)-5-fluoropyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylate (13): Toluene (16 L) was deoxygenated by N2 bubbling for ˜2 h. To a separate flask equipped with a heat source and reflux condenser were charged (R)-ethyl 5-(2-(5-fluoro-2-(((trifluoromethyl)sulfonyl)oxy)pyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylate 11 (1440 g, 2860 mmol), copper(I) iodide (105 g, 551.3 mmol), Pd catalyst (398 g, 567.0 mmol), and the deoxygenated toluene. Diisopropylamine (810 ml, 5779 mmol) was added and the mixture was heated to 60° C. After ˜1 h, the reaction temp was 60° C. and commercially available (R)-tert-butyl but-3-yn-2-ylcarbamate (12) (728 g, 4302 mmol) was added in three portions. After ˜1 h, the mixture was cooled with an ice/water bath and then water (14 L) was added. When the reaction temp reaches ˜35° C., it was filtered (PPFC) and washed with water (2×3.5 L). The filtrate was transferred to a separatory vessel and the aqueous layer was washed with toluene (2×3.5 L). The aqueous layer was transferred to a separate flask and added DCM (14 L) was added. The mixture was cooled to <15° C., then sat. NH4OH (2.4 L) was added. The solution was transferred to a separatory vessel and then washed with DCM (7 L). The DCM layers were allowed to stand at ambient temperature overnight and then they were combined and washed with brine (7 L). The organic layer was then concentrated, MeOH (5 L) was added and the mixture was concentrated to give 13.




embedded image


Ethyl 5-((R)-2-(2-((R)-3-aminobutyl)-5-fluoropyridin-3-yl) pyrrolidin-1-yl) pyrazolo[1,5-a] pyrimidine-3-carboxylate (14): Palladium on carbon (235 g, 104 mmol, 4.7 wt %), a 1285 g methanolic solution of ethyl 5-((R)-2-(2-((R)-3-aminobut-1-yn-1-yl)-5-fluoropyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylate (13) (472 g, 1117 mmol) and MeOH (2.5 L˜4 L total volume) were charged into a 8 L Parr reactor. The mixture was stirred at 50 psi H2 until it was judged complete. The hydrogen atmosphere was replaced with nitrogen and the reaction mixture was allowed to stand overnight. The next day it was filtered through GF/F filter paper. The solution was concentrated to give 14.




embedded image


5-((R)-2-(2-((R)-3-aminobutyl)-5-fluoropyridin-3-yl) pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylic acid (15): A methanol solution of 14 (861 g, 2019 mmol) was combined with IPA (4 L) and then concentrated to 2.2 kg under vacuum. The concentrate was transferred to a reactor (with reflux condenser) with further dilution in IPA (10 L). The mixture was heated to 75° C. (IT). Sodium hydroxide (184 mL, 2631 mmol) was added and the reaction continues until it was judged complete by HPLC. The heat was removed and the mixture was allowed to cool to ambient temp overnight. Concentrated hydrochloric acid (214 mL, 2632 mmol) was added. The mixture was concentrated under vacuum with external heating to 45° C. to ˜5 mL/g. Heptane (12 L) was added and the suspension was allowed to cool to ambient temp and then stirred for ˜1 h. The suspension was filtered (PPFC) and washed with 3:1 heptane:IPA (2×1600 mL). The wet cake was placed in trays and dried under vacuum at 55° C. to constant weight to give 15.




embedded image


(13E,14E,22R,6R)-35-fluoro-6-methyl-7-aza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(3,2)-pyridina-2(1,2)-pyrrolidinacyclooctaphan-8-one(Compound 1): To a flask containing EDCI (157 g, 819 mmol) and DMAP (133 g, 1091 mmol) in DCM (50 mL/g, 125 mL) was added 5-((R)-2-(2-((R)-3-aminobutyl)-5-fluoropyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylic acid (15) (302 g, 546 mmol) in 8 portions (37.8 g each). The portions were added ˜60 min apart. The reaction mixture was stirred overnight at ambient temperature. The mixture was transferred to a separatory funnel with minimal DCM and washed with sat. NaHCO3 (2×3 L), and 0.25 M citric acid (2×3 L, pH 5.5). The combined aqueous layers were washed with DCM (3 L, 10 mL/g) and then concentrated under vacuum (rotovap). The concentrate was dissolved in 3% MeOH in DCM and loaded onto a flash column (3 kg, SiO2) and eluted with 3% MeOH in DCM (40 L total). The fractions containing the product were concentrated to give Compound 1 Combined lots of solid Compound 1 were triturated in IPAc (2.5 L, ca. 5 mL/g) at room temperature for 2 h. The mixture was heated to 40-45° C. for 10 minutes, then triturated at room temperature. The suspension was filtered and washed with IPAc (2×250 mL, ca. 2×0.5 mL/g) to give, after oven drying at 55° C., Compound 1.




embedded image


(13E,14E,22R,6R)-35-fluoro-6-methyl-7-aza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(3,2)-pyridina-2(1,2)-pyrrolidinacyclooctaphan-8-one(Compound 1) (alternative preparation): To a flask containing EDCI (1091 g, 5.7 mol, 1.7 eq) and DMAP (941 g, 7.71 mol, 2.3 eq) in DCM (38 L) were added the amino-acid 15 [5-((R)-2-(2-((R)-3-aminobutyl)-5-fluoropyridin-3-yl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidine-3-carboxylic acid] (1900 g, 3.35 mol) in 6 portions (added at least one hour apart), and the reaction was stirred at room temperature overnight. Once the reaction was complete it was transferred to a separatory funnel and washed with sat'd NaHCO3 solution (2×19 L). The DCM layer was then washed with 0.25 M citric acid (38 L). The combined, citric acid aqueous layers were back-extracted with DCM (19 L), and the organic phases were added back to the 100 L round-bottomed flask. Charcoal (2.01 kg) and silica gel (2.01 kg) were added, and the suspension stirred at room temperature overnight. The next day, the suspension was filtered, and the charcoal cake was washed with DCM (3×19 L). The DCM filtrates were filtered a second time. The pale yellow solution was concentrated to minimum volume. Isopropyl acetate (28.5 L) was added and concentrated to 10 to 20 L. The suspension was heated overnight at 75° C., and the mixture was allowed to cool to room temperature. The solids were collected by filtration and washed with isopropyl acetate (2×1.9 L). The crude product was transferred to trays and dried in a vacuum oven 55° C. until constant mass was achieved.


To a flask was charged [(13E,14E, 22R, 6R)-35-fluoro-6-methyl-7-aza-1(5,3)-pyrazolo[1,5-a]pyrimidina-3(3,2)-pyridina-2(1,2)-pyrrolidinacyclooctaphan-8-one] followed by 2-butanone (6.3 L). The slurry was agitated at 75° C. for 2 days and then the product was collected by filtration, and the product cake was washed with 2-butanone (2×950 mL g). The product was transferred to trays and dried in a vacuum oven at 55° C. until constant mass was achieved to provide Compound 1.


The average purity of Compound 1 was 98.8% as determined by HPLC-UV. The structure of Compound 1 was confirmed using 1H NMR.


2) Preparation of the Compound of Formula II


Compound of Formula II is prepared using methods and procedures similar to those used to prepare the compound of Formula I using tert-butyl prop-2-ynylcarbamate (compound 19) instead of tert-butyl (R)-but-3-yn-2-ylcarbamate (compound 12)




embedded image


(6R)-9-fluoro-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Formula II)

MS (apci) m/z=367.3 (M+H).


3) Preparation of the Compound of Formula III


Compound of Formula III is prepared using methods and procedures similar to those used to prepare the compound of Formula I, using tert-butyl 2-methylbut-3-yn-2-ylcarbamate (compound 23) instead of tert-butyl (R)-but-3-yn-2-ylcarbamate (compound 12).




embedded image


(6R)-9-fluoro-15,15-dimethyl-2,11,16,20,21,24-hexaazapentacyclo [16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Formula III

MS (apci) m/z=395.1 (M+H).


Preparation of Crystalline Forms and Salts of Compound 1


General methods for preparation and characterization of Compound 1 salts


Approximately 20 mg Compound 1 was weighed into 2 mL vials. Acid counterions were weighed into separate vials and stock solutions prepared for the liquid counterions (1.05 eq.). Table 11 shows acid weights and volumes.











TABLE 11









Neat Addition



Amounts










By
By











Acid

pKa
Weight
Volume













No.
Acid
1
2
3
(mg)
(μL)
















1
Hydrochloric acid 37
−6.10


5.45
4.6



wt. % (12M)


2
Sulfuric acid
−3.00
1.92

5.71
3.1


3
1-2-Ethane disulfonic
−2.10
−1.50

12.94



acid


4
p-Toluene sulfonic acid
−1.34


10.84


5
Methane sulfonic acid
−1.20


5.31
3.6


6
Naphthalene-2-sulfonic
0.17


14.14



acid


7
Benzene sulfonic acid
0.70


8.92


8
Oxalic acid
1.27
4.27

5.08


9
2-Hydroxy
1.66


8.19



ethanesulfonic acid


10
L-Aspartic acid
1.88
3.65

7.36


11
Maleic acid
1.92
6.23

6.48


12
Phosphoric acid
1.96
7.12
12.32
5.42


13
Ethane sulfonic acid
−2.05


6.41
4.7


14
L-Glutamic acid
2.19
4.25

8.13


15
L-Tartaric acid
3.02
4.36

8.34


16
Fumaric acid
3.03
4.38

6.48


17
Citric acid
3.13
4.76
6.40
10.67


18
D-Glucuronic acid
3.18


10.73


19
L-Malic acid
3.46
5.10

7.49


20
Hippuric acid
3.55


10.1


21
D-Gluconic acid (50%
3.76


21.68
17.6



in water)


22
DL-Lactic acid (85%
3.86


5.86
4.8



aq. solution)


23
L-Ascorbic acid
4.17
11.57

9.73


24
Benzoic acid
4.19


6.82


25
Succinic acid
4.21
5.64

6.59









Preparation of samples of salts of these acids and the Compound 1 in selective solvents (acetone, ethanol, methanol, 2-propanol, TBME and THF) is described in the Examples. In the Examples 8-32, solids observed post-temperature cycling were collected and analyzed by XRPD. Samples in which solid was not observed had anti-solvent additions made to saturated solutions and the resultant solids were analyzed by XRPD.


Anti-Solvent Additions


Approximately 1 mL of anti-solvent (heptane or TBME depending on miscibility) was added dropwise to saturated salt solutions of Compound 1 free base. Any resulting solid was analyzed by XRPD.


Salt Stability Studies


Recovered salts were placed in an oven at 40° C./75% RH for 1 week, and the resultant materials were analyzed by XRPD to determine any changes to form or crystallinity.


Thermodynamic Solubility


Thermodynamic solubility studies were carried out as follows: 10 mg of prepared salts were suspended in pH 1, 4.5, 6.8 and un-buffered water (300 μL). The pH of the slurries was measured and adjusted accordingly using either 0.2M HCl solution or 0.2M sodium hydroxide solution. The slurries were agitated for 24 hours at ambient temperature using an incubator shaker. The resulting slurries were filtered, any solids recovered were analyzed by XRPD and filtrate pH measured and submitted for UPLC analysis. pH 1.0 Buffer: 67 mL 0.1M hydrochloric acid solution was added to 12.5 mL 0.2M potassium chloride solution and diluted to 100 mL using de-ionized water and adjusted accordingly. pH 4.5 Buffer: 7.0 mL 0.2M sodium hydroxide solution was added to 25 mL 0.2 potassium hydrogen phthalate solution and diluted to 100 mL using de-ionized water and adjusted accordingly. pH 6.8 Buffer: 11.2 mL 0.2M sodium hydroxide solution was added to 25 mL 0.2M potassium phosphate mono-basic and diluted to 100 mL using de-ionized water and adjusted accordingly.


Salt Disproportionation Studies


Salt disproportionation studies were carried out as follows: 20 mg of prepared salts were weighed into a vial and 0.5 mL of deionized water was added. The samples were then agitated for 24 h at ambient temperature. The pH of the samples was taken pre- and post-agitation. Any solids recovered were submitted for XRPD analysis to determine any changes to form.


Hydration Screen


Hydration screen was carried out as follows: 10 mg of prepared salts were suspended in several acetone/water mixtures of various water activities (low: aw=0.281, medium: aw=0.776 and high: aw=0.919) and agitated at ambient temperatures for 24 hr. Any recovered solids were submitted for XRPD analysis to determine any changes to form.


Analytical Methods


X-ray Powder Diffraction (XRPD)


XRPD analysis was carried out on a Panalytical X′pert pro, scanning the samples between 3 and 35° 2θ. The material was gently ground and loaded onto a multi-well plate with Kapton or mylar polymer film to support the sample. The multi well plate was then loaded into a Panalytical diffractometer running in transmission mode, using Cu K radiation, and analyzed. The experimental conditions are shown in Table 12.












TABLE 12







Raw Data Origin:
XRD measurement









Scan Axis:
Gonio



Start Position [°2θ]:
3.0066



End Position [°2θ]:
34.9866



Step Size [°2θ]:
0.0130



Scan Step Time [s]:
18.8700



Scan Type:
Continuous



PSD Mode:
Scanning



PSD Length [°2θ]:
3.35



Offset [°2θ]:
0.0000



Divergence Slit Type:
Fixed



Divergence Slit Size [°]:
1.0000



Measurement Temperature
25.00



Anode Material:
Cu



K-Alpha 1 [Å]:
1.54060



K-Alpha2 [Å]:
1.54443



K-Beta [Å]:
1.39225



K-A2/K-A1 Ratio:
0.50000



Generator Settings:
40 mA, 40 kV



Goniometer Radius [mm]:
240.00



Dist. Focus-Diverg. Slit [mm]:
91.00



Incident Beam Monochromator:
No



Spinning:
No











Single Crystal X-ray Analysis (SXRD)


SXRD analysis was conducted on a Agilent Technologies (Dual Source) SuperNova diffractometer using monochromated Cu Kα (λ=1.54184 Å) radiation. The diffractometer was fitted with an Oxford Cryosystems low temperature device to enable data collection to be performed at 120(1) K and the crystal encased in a protective layer of Paratone oil. The data collected were corrected for absorption effects based on Gaussian integration over a multifaceted crystal model, implemented as a part of the CrysAlisPro software package (Agilent Technologies, 2014).


The structure was solved by direct methods (SHELXS97) (Sheldrick, G. M. Acta Cryst. Sect. A 2008, 64, 112.) and developed by full least squares refinement on F2 (SHELXL97) interfaced via the OLEX2 software package. Images were produced using OLEX2 (Dolomanov, O. V. et al. J Appl. Cryst. 2009, 42, 339-341).


Polarized Light Microscopy (PLM)


The presence of crystallinity (birefringence) was determined using an Olympus BX50 polarizing microscope, equipped with a Motic camera and image capture software (Motic Images Plus 2.0). All images were recorded using the 20× objective, unless otherwise stated.


Thermogravimetric Analysis (TGA)/Differential Thermal Analysis (DTA)


Approximately, 5 mg of material was weighed into an open aluminum pan and loaded into a simultaneous thermogravimetric/differential thermal analyzer (TG/DTA) and held at room temperature. The sample was then heated at a rate of 10° C./min from 20° C. to 400° C. during which time the change in sample weight was recorded along with any differential thermal events (DTA). Nitrogen was used as the purge gas, at a flow rate of 300 cm3/min.


Differential Scanning Calorimetry (DSC)


Approximately, 5 mg of material was weighed into an aluminum DSC pan and sealed non-hermetically with a pierced aluminum lid. The sample pan was then loaded into a Seiko DSC6200 (equipped with a cooler) cooled and held at 20° C. Once a stable heat-flow response was obtained, the sample and reference were heated to 350° C. at scan rate of 10° C./min and the resulting heat flow response monitored.


Infrared Spectroscopy (IR)


Infrared spectroscopy was carried out on a Bruker ALPHA P spectrometer. Sufficient material was placed onto the center of the plate of the spectrometer and the spectra were obtained using parameters indicated in Table 13:












TABLE 13









Resolution:
 4 cm−1



Background Scan Time:
16 scans



Sample Scan Time:
16 scans



Data Collection:
4000 to 400 cm−1



Result Spectrum:
Transmittance











Nuclear Magnetic Resonance (NMR)


NMR experiments were performed on a Bruker AVIIIHD spectrometer equipped with a DCH cryoprobe operating at 500.12 MHz for 1H channel Experiments were performed in deuterated DMSO and each sample was prepared to about 10 mM concentration.


Dynamic Vapor Sorption (DVS)


Approximately, 10 mg of sample was placed into a mesh vapor sorption balance pan and loaded into a DVS-1 dynamic vapor sorption balance by Surface Measurement Systems. The sample was subjected to a ramping profile from 40-90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (99.5% step completion). After completion of the sorption cycle, the sample was dried using the same procedure to 0% RH and then a second sorption cycle back to 40% RH. The weight change during the sorption/desorption cycles were plotted, allowing for the hygroscopic nature of the sample to be determined. XRPD analysis was then carried out on any solid retained.


Gravimetric Vapor Sorption (GVS)


Approximately 10-20 mg of sample was placed into a mesh vapor sorption balance pan and loaded into an IGASorp Moisture Sorption Analyzer balance by Hiden Analytical. The sample was subjected to a ramping profile from 40-90% relative humidity (RH) at 10% increments, maintaining the sample at each step until a stable weight had been achieved (98% step completion). After completion of the sorption cycle, the sample was dried using the same procedure to 0% RH, and finally taken back to the starting point of 40% RH. The weight change during the sorption/desorption cycles were plotted, allowing for the hygroscopic nature of the sample to be determined.


High Performance Liquid Chromatography-Ultraviolet Detection (HPLC-UV)


HPLC experiments were performed on Agilent 1100 HPLC instrument with diode array detector (DAD) using parameters indicated in Table 14:












TABLE 14









Column:
ACE3 C181-PFP 50 × 4.6 × 3 μm



Column Temperature:
45.0° C.



Autosampler Temperature:
Ambient



UV wavelength:
265 nm



Injection Volume:
2.00 μL



Flow Rate:
2 mL/min



Mobile Phase A:
95.0% (0.1% TFA/DI-H2O)



Mobile Phase B:
5.0% (0.1% TFA/MeCN)











Gradient program is shown in Table 15:












TABLE 15







Time (minutes)
Solvent B [%]



















0.00
5.0



2.50
60.0



3.20
80.0



3.21
5.0



5.50
5.0










Example 1
Solubility of Compound 1 Free Base

A solid Compound 1 was obtained as follows. A 53 mL of solution containing about 330 mg of Compound 1 in warm 1,4-dioxane was divided between 33, 2 mL glass vials (1.5 mL in each). The solutions were frozen and freeze-dried by lyophilization overnight. The resulting material was then analyzed by XRPD to confirm mostly amorphous material.


Approximately 10 mg of amorphous Compound 1 was produced in 32×2 mL glass vials from freeze drying and 100 μL of the appropriate solvent system was added to the appropriate vial. Between each addition, the mixture was checked for dissolution and if no dissolution was apparent, the mixture was heated to about 40° C. and checked again. This procedure was continued until dissolution was observed or until 2 mL had been added (to the compound concentration of <5 mg/mL). The results of the solubility measurements are shown in Table 16.










TABLE 16





Solvent
Approx. Solubility mg/mL
















Acetone
11.1


Acetonitrile
12.5


Anisole
11.1


1-Butanol
17


2-Butanone
11.1


TBME
<5


Cyclohexane
<5


Cyclopentylmethyl ether
<5


Dichloromethane
>100


Diisopropyl ether
<5


N,N-Dimethylacetamide
>100


1,2-Dimethoxyethane
8.3


Diglyme (bis(2-methoxyethyl ether)
8.3


1,4-Dioxane
8.3


Dimethylformamide
>100


Dimethylsulfoxide
50


Ethanol
20


Ethyl acetate
<5


2-Ethoxy ethanol
50


Heptane
<5


Isobutyl acetate
<5


Isopropyl acetate
<5


Methanol
50


Methylisobutyl ketone
6.25


2-Methyl THF
<5


N-Methylpyrrolidone
>100


2-Propanol
14.3


1-Propanol
>100


Tetrahydrofuran
20


Toluene
<5


TBME:Heptane (60:40 v/v)
<5


Water
<5









Compound 1 showed low solubility in non-polar solvents such as toluene and 1,4-dioxane, medium solubility in polar aprotic solvents such as acetone, ethyl acetate and acetonitrile and high solubility in polar solvents such as DMSO, DMF and protic solvents such as methanol. In the remainder of cases and where “<” is present, solid was still present after the maximum volume of 2 mL was added. XRPD analysis of the recovered solids from the solvent solubility study returned the same crystalline form of free base in all cases (Form I), however, showing varying degrees of crystallinity and peak intensity (preferred orientation may have an effect on crystallinity of a sample). Insufficient solids were recovered from anisole, 1-butanol, diglyme, 2-ethoxy ethanol, MIBK and N-methylpyrrolidone.


Example 2
Preparation of Crystalline Compound 1 (Form I)

Solid Compound 1 was obtained as follows. A 212 mL of solution containing about 1.04 g of Compound 1 in warm 1,4-dioxane was divided between 26, 20 mL glass vials (approximately 8 mL in each). The solutions were frozen and freeze-dried by lyophilization overnight. The resulting material was then analyzed by XRPD to confirm mostly amorphous material.


The 25 vials each containing approximately 40 mg of amorphous freeze-dried Compound 1 were used. A solvent was added to each vial and Compound 1 was suspended in the solvent. The following 25 solvents were used: acetone, acetonitrile, anisole, 1-butanol, 2-butanone, TBME, cyclohexane, cyclopentylmethyl ether, 1,2-dimethoxyethane, 1,4-dioxane, ethanol, ethyl acetate, 2-ethoxy ethanol, heptane, isobutyl acetate, isopropyl acetate, methanol, methylisobutyl ketone, 2-methyl THF, 2-propanol, 1-propanol, tetrahydrofuran (THF), toluene, TBME:heptane (60:40 v/v), and water. The crystallization conditions consisted of maturation cycles, evaporation, cooling and anti-solvent addition techniques.


Temperature Cycling


Each of the 25 vials was temperature cycled between ambient temperature and 40° C. in 4 hour cycles over 72 h The resulting solids were isolated by centrifugation and analyzed by XRPD and PLM. Solids recovered from temperature cycling and analyzed by XRPD appeared to be the same as the input material (Form I) with varying degrees in crystallinity. No residual solid material was recovered from anisole, 1-butanol, 2-butanone, 2-ethoxy ethanol, 2-methyl THF, 1-propanol and THF.


A filtered saturated solution of Compound 1 in a specified solvent was divided into five vials and used to prepare crystalline forms of the compound according to the procedures described below:


Crash Cool (2° C.)


Saturated solutions of Compound 1 were stored at 2° C. for 24-72 h. At this time any material recovered was analyzed by XRPD. The crash cooling experiments at 2° C. recovered insufficient solids from all solvents for XRPD analysis except from 2-propanol which returned Compound 1 (Form I).


Crash Cool (−18° C.)


Saturated solutions of Compound 1 were stored at −18° C. for 24-72 h. At this time any material recovered was analyzed by XRPD. The crash cooling experiments at −18° C. recovered insufficient solids from all solvents for XRPD analysis except 1-butanol, ethanol, 2-propanol and 1-propanol. From the solids that were analyzed by XRPD analysis, all returned Compound 1 (Form I) with varying degrees of crystallinity.


Anti-Solvent Addition at Ambient Temperature


Approximately 1 mL of anti-solvent (heptane or TBME depending on miscibility) was added dropwise to saturated solutions of Compound 1 free base. Any resulting solid was analyzed by XRPD. The anti-solvent addition at ambient temperature experiments recovered insufficient solids from all solvents for XRPD analysis except acetone, acetonitrile, 2-butanone, 1,2-dimethoxyethane, 1,4-dioxane and ethanol. From the solids that were analyzed by XRPD analysis, all returned Compound 1 (Form I) with varying degrees of crystallinity.


Anti-Solvent Addition at 2° C.


Approximately 1 mL of anti-solvent (heptane or TBME depending on miscibility) was added dropwise to saturated solutions of Compound 1 free base. Any resulting solid was analyzed by XRPD. The anti-solvent addition at 2° C. experiments recovered insufficient solids from all solvents for XRPD analysis except acetone, acetonitrile, 1-butanol, 2-butanone, 1,2-dimethoxyethane, 1,4-dioxane, ethanol, ethyl acetate, MIBK, 1-propanol and THF. From the solids that were analyzed by XRPD, all returned Compound 1 (Form I) with varying degrees of crystallinity.


Evaporation


Saturated solutions of Compound 1 were transferred to 2 mL vials, these vials were then uncapped and allowed to evaporate at ambient temperature to recover material. Any material recovered was analyzed by XRPD. The evaporation experiments recovered insufficient solids from all solvents for XRPD analysis except acetone, acetonitrile, 2-butanone, cyclopropylmethyl ether, 1,2-dimethoxyethane, 1,4-dioxane, ethanol, ethyl acetate, 2-ethoxy ethanol, isobutyl acetate, isopropyl acetate, methanol, MIBK, 2-propanol, 1-propanol and THF. From the solids that were analyzed by)(RFD, all returned Compound 1 (Form I) with varying degrees of crystallinity.


Example 3
Characterization of Crystalline Compound 1 (Form I)

X-Ray Powder Diffraction (XRPD)


Form I of crystalline (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Compound 1 free base) was characterized by XRPD. The XRPD pattern is shown in FIG. 1 and XRPD data is provided in Table 17.











TABLE 17





2-Theta (°)
Height
H %

















7.9
692
4.0


9.1
10133
58.1


11.2
6232
35.7


12.8
695
4.0


13.4
4471
25.6


14.8
2667
15.3


15.2
479
2.8


15.5
144
0.8


16.8
929
5.3


18.3
2049
11.8


18.6
2818
16.2


19.5
792
4.5


20.2
17437
100.0


21.4
1327
7.6


22.7
1668
9.6


23.2
210
1.2


23.6
1908
10.9


24.9
6322
36.3


25.8
783
4.5


26.1
447
2.6


26.5
537
3.1


27.0
1478
8.5


27.7
220
1.3


28.4
259
1.5


28.8
228
1.3


29.4
1795
10.3


30.0
142
0.8


30.3
358
2.1


31.2
197
1.1


32.1
359
2.1


32.3
357
2.1


33.3
248
1.4


34.4
70
0.4









As shown in FIG. 1, according to the XRPD analysis, the material is crystalline. PLM analysis showed birefringence with irregular morphology.


Thermogravimetric/differential Thermal Analysis (TG/DTA)


Form I of crystalline (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Compound 1 free base) was characterized by TGA and DTA. TGA showed a weight loss of approximately 1.1% from outset up to 200° C., while DTA showed an endothermal “melting” event at onset about 315° C. (peak at 317° C.). The TG/DTA thermogram is shown in FIG. 2.


Differential Scanning Calorimetry (DSC)


Form I of crystalline (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one (Compound 1 free base) was characterized by DSC. DSC analysis in the first heat showed a sharp endothermal event at onset 315° C. (peak at 317° C.) which is consistent with TG/DTA. No thermal events were seen in the cooling cycle. The second heating cycle showed a small endothermal event at onset around 118° C. (peak at 124° C.) which is highly likely to be a glass transition (Tg). The DSC thermograms are shown in FIG. 3


In sum, Compound 1 exists as one crystalline form (Form I) with favorable thermal properties with a melting point of 315° C. and low hygroscopicity with a mass uptake of 0.3% at 90% RH and no changes to form or crystallinity after exposure to GVS humidity conditions.


Example 4
Recrystallization of Compound 1 and Characterization of the Recrystallized Material

Compound 1 was recrystallized from 1-propanol as follows. 500 mg of Compound 1 was weighed into a 20 mL vial. To this vial, 20 mL of 1-propanol was added gradually over 3 hours. The sample was placed in a 95° C. heated block to aid dissolution. The sample was slow to dissolve but a clear solution was achieved. The sample was cooled to 10° C. at 5° C./min. Once the cooling cycle had reached 10° C. the sample remained at 10° C. for a further 24 hours to recover material. The solids were then recovered and dried using a vacuum oven at ambient temperature.


XRPD analysis of the recrystallized solid showed no changes in crystalline form, and PLM analysis showed the material to be birefringent with irregular morphology. TGA showed a weight loss of approximately 0.7% from the outset up to 250° C., whilst DTA showed an endothermal ‘melting’ event at onset approximately at 314° C. (peak at about 318° C.).


Purity of recrystallized solid is 99.2% as determined by HPLC-UV. 1H-NMR analysis shows that the spectrum is consistent with the structure and shows little if any obvious residual process solvents. 1H NMR spectrum is shown in FIG. 9.


Differential Scanning Calorimetry (DSC)


DSC analysis in the first heating cycle showed a sharp endothermal event at onset approximately 316° C. (peak at 317° C.). This endothermal event is consistent with TG/DTA. In the first cooling cycle of the DSC analysis, a slow broad recrystallization is observed with a peak at about 284° C. shows the thermogram of the first cooling cycle. DSC analysis in the second heating cycle showed a series of exothermic events which could be potential recrystallizations which were followed by a sharp endothermal event at onset about 313° C. (peak at about 316° C.). shows the thermogram of the second heating cycle of the recrystallized Compound 1 free base.


Infra-Red Analysis (IR)


Recrystallized Form I of crystalline Compound 1 was characterized by IR. FIG. 8 shows IR spectrum and the peaks are listed in Table 18.














TABLE 18







Wave Number
Abs. Intensity
Rel. Intensity
Width





















3344.4
0.8
0.1
27.8



3066.3
0.9
0.0
31.6



3019.9
0.9
0.0
15.3



2962.0
0.8
0.0
18.7



2929.8
0.8
0.0
25.5



2870.4
0.8
0.1
143.0



1649.6
0.6
0.1
2538.9



1625.8
0.5
0.4
43.1



1599.0
0.8
0.0
61.5



1566.9
0.6
0.3
17.6



1537.4
0.6
0.2
13.5



1492.1
0.5
0.2
2106.0



1450.6
0.4
0.5
63.7



1365.4
0.6
0.1
1743.6



1346.7
0.6
0.2
44.5



1281.8
0.7
0.1
8.5



1257.5
0.7
0.1
8.0



1234.4
0.6
0.2
40.6



1219.2
0.6
0.0
5.5



1167.6
0.7
0.1
35.4



1156.0
0.7
0.0
5.6



1114.8
0.8
0.0
9.4



1093.2
0.8
0.0
9.7



1070.8
0.7
0.1
10.7



992.3
0.8
0.0
318.0



964.0
0.7
0.1
13.0



945.2
0.8
0.0
132.0



923.0
0.8
0.1
6.9



903.6
0.7
0.1
7.3



890.8
0.7
0.2
35.4



859.2
0.8
0.0
175.9



796.2
0.6
0.2
11.5



777.7
0.6
0.0
0.1



770.5
0.6
0.3
19.5



740.5
0.8
0.0
147.4



719.7
0.8
0.0
694.4



709.6
0.7
0.1
25.8



686.7
0.8
0.0
170.0



633.0
0.8
0.1
7.9



616.0
0.7
0.1
11.2



552.4
0.6
0.2
43.9



509.5
0.7
0.0
9.3



468.8
0.8
0.1
13.6



442.2
0.7
0.1
16.7



432.1
0.8
0.0
91.7



405.0
0.7
0.1
10.1










In sum, Compound 1 recrystallized from 1-propanol exhibited the same properties as the compound prior to recrystallization, with an increased purity of >99%. As shown in Example 5, the material showed no change to form or purity after exposure to stability stress conditions and no change to form after an aqueous solubility assessment.


Example 5
Stability of Compound 1 (Form I)

Compound 1 (Form I) was subjected to various different environmental conditions to assess stability.


Vapor Sorption—before Recrystallization


Gravimetric vapor sorption (GVS) showed that Compound 1 exhibits slight hygroscopicity with a mass uptake of approximately 0.3% at 90% RH. FIG. 4 shows GVS isotherm plot and FIG. 5 shows GVS kinetic plot. Post-XRPD analysis showed no changes in crystalline form upon exposure to GVS conditions.


Vapor Sorption—Recrystallized Solid


Dynamic vapor sorption (DVS) analysis of the recrystallized compound shows the material to exhibit slight hygroscopicity with a mass uptake of about 0.7% at 90% RH. FIG. 6 shows DVS analysis of the recrystallized compound. FIG. 7 shows DVS kinetic plot of the recrystallized solid. Post-DVS XRPD analysis shows no change in crystalline form upon exposure to DVS humidity conditions.


Humidity, temperature, Ambient Light—Recrystallized Solid


1-week stability tests on recrystallized solid showed no change to form after exposure to 40° C./75% RH, 80° C. and under ambient light. UPLC analysis showed no change in purity of the samples after exposure to stability stress conditions (average purity 99.2 for relative humidity and ambient light tests, and 99.3% for 80° C. test).


Example 6
Single Crystal X-ray Analysis of Compound 1 (Form I)

Crystals of Compound 1 (Form 1) were prepared as follows. Compound 1 (2 mg) was dissolved in methanol (500 μL) in a 1.75 clear glass vial then capped with a pierced lid. The solution was left to stand at ambient for several days without agitation to allow for large rod-like crystals to grow that were suitable for interrogation by single crystal X-ray diffraction.


The highest residual Fourier peak was found to be 0.16 e. Å−3 approx 0.72 Å from C(4), and the deepest Fourier hole was found to be −0.22 e. Å−3 approx 0.75 Å from C(10). Crystal Data for C20H21FN6O (M=380.43 g/mol): orthorhombic, space group P212121 (no. 19), a=6.91792(3) Å, b=13.74742(3) Å, c=19.22580(5) Å, V=1828.442(10) Å3, Z=4, T=207(120) K, μ(CuKα)=0.799 mm-1, Dcalc=1.382 g/cm3, 169333 reflections measured (7.9°≤2Θ≤152.76°), 3833 unique (Rint=0.0639, Rsigma=0.0180) which were used in all calculations. The final R1 was 0.0338 (>2sigma(I)) and wR2 was 0.0908 (all data). Crystallographic parameters and refinement indicators of Compound 1 (Form I) are shown in Table 19.












TABLE 19









Empirical formula
C20H21FN6O



Formula weight
380.43



Temperature/K
120(1)



Crystal system
Orthorhombic



Space group
P212121



a/Å
6.91792(3)



b/Å
13.74742(3)



c/Å
19.22580(5)



α/°
90.00



β/°
90.00



γ/°
90.00



Volume/Å3
1828.442(10)



Z, Z′
4



ρcalc g/cm3
1.382



μ/mm−1
0.799



F(000)
800.0



Crystal size/mm3
0.47 × 0.117 × 0.105



Radiation
CuKα (λ = 1.54178)



2Θ range for data collection/°
7.9 to 152.76



Index ranges
−8 ≤ h ≤ 7, −17 ≤ k ≤




17, −24 ≤ 1 ≤ 24



Reflections collected
169333



Independent reflections
3833 [Rint = 0.0639,




Rsigma = 0.0180]



Data/restraints/parameters
3833/0/258



S
1.060



Final R indexes [F2 > 2σ (F2)]
R1 = 0.0338, wR2 = 0.0907



Final R indexes [all data]
R1 = 0.0340, wR2 = 0.0908



Δρmax , Δρmin/e Å−3
0.16/−0.22



Flack parameter
−0.01(15)











FIG. 10 shows 3-D view of Compound 1 (Form I) with atom labels. FIG. 11 shows ORTEP view of Compound 1 (Form I) with atom labels. All non-hydrogen atoms are shown with thermal ellipsoids set at the 50% probability level.


Example 7
Single Crystal X-ray Analysis of Compound 1, Acetonitrile Solvate

Crystals of Compound 1, acetonitrile solvate were prepared vas follows. Compound 1 (2 mg) was dissolved in acetonitrile (500 μl) in a 1.75 clear glass vial then capped with a pierced lid. This solution was left to stand at ambient for several days without agitation to allow for large rod-like crystals to grow that were suitable for interrogation by single crystal X-ray diffraction.


The highest residual Fourier peak was found to be 0.19 e. Å−3 approx 0.67 Å from C(11), and the deepest Fourier hole was found to be −0.21 e. Å-3 approx 0.81 Å from N(4). Crystal Data for C24H27FN8O (M=462.54 g/mol): orthorhombic, space group P212121 (no. 19), a=6.03307(4) Å, b=16.10794(9) Å, c=23.72624(13) Å, V=2305.73(2) Å3, Z=4, T=294.01(10) K, μ(CuKα)=0.757 mm-1, Dcalc=1.332 g/cm3, 110019 reflections measured (6.64°≤2θ≤152.4°), 4840 unique (Rint=0.0983, Rsigma=0.0211) which were used in all calculations. The final R1 was 0.0339 (>2sigma(I)) and wR2 was 0.0891 (all data). Crystallographic parameters and refinement indicators of Compound 1 (Form I) are shown in Table 20.












TABLE 20









Empirical formula
C24H27FN8O



Formula weight
462.54



Temperature/K
120(1)



Crystal system
orthorhombic



Space group
P212121



a/Å
6.03307(4)



b/Å
16.10794(9)



c/Å
23.72624(13)



α/°
90.00



β/°
90.00



γ/°
90.00



Volume/Å3
2305.73(2)



Z, Z′
4



ρcalc g/cm3
1.332



μ/mm−1
0.757



F(000)
976.0



Crystal size/mm3
0.564 × 0.082 × 0.033



Radiation
CuKα (λ = 1.54184)



2Θ range for data collection/°
6.64 to 152.4



Index ranges
7 ≤ h ≤ 6, −20 ≤ k ≤




20, −29 ≤ 1 ≤ 29



Reflections collected
110019



Independent reflections
4840 [Rint = 0.0983,




Rsigma = 0.0211]



Data/restraints/parameters
4840/0/310



S
1.096



Final R indexes [F2 > 2σ (F2)]
R1 = 0.0339, wR2 = 0.0887



Final R indexes [all data]
R1 = 0.0345, wR2 = 0.0891



Δρmax , Δρmin/e Å−3
0.19/−0.21



Flack parameter
−0.02(14)











FIG. 12 shows 3-D view of Compound 1 bis-acetonitrile solvate with atom labels. FIG. 13 shows ORTEP view of Compound 1 bis-acetonitrile solvate asymmetric unit with atom labels. All non-hydrogen atoms are shown with thermal ellipsoids set at the 50% probability level.


Example 8
Preparation and Characterization of Compound 1 Benzenesulfonic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing benzenesulfonic acid (8.92 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Scale-Up Preparation from Ethanol


About 300 mg of Compound 1 was weighed into a vial and 133 mg of benzenesulfonic acid was weighed into a separate vial. To both vials, 3.75 mL of ethanol was added and the two mixtures combined. The resulting slurry was then temperature cycled for 24 hours (ambient to 40° C. in 4 hours cycles) (1.05 eq. of acid to free base). The resulting slurry was then allowed to evaporate at ambient temperatures to remove excess ethanol.


Observations from the treatment of Compound 1 with benzenesulfonic acid are shown in Table 21 below:











TABLE 21









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Solid
Clear
Gum
Slurry
Slurry
Solid




Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of benzenesulfonic acid experiments recovered 5 crystalline hits, free base (Form I) recovered from acetone and 2-propanol, pattern 1 was recovered from THF and t-BME (FIG. 17) and pattern 2 recovered from ethanol (FIG. 18). Insufficient solids were recovered from ethanol to determine form.


XRPD data for Compound 1 besylate is provided in Table 22.











TABLE 22





2-Theta (°)
Height
H %

















8.1
15179
100.0


9.2
864
5.7


10.0
85
0.6


11.7
591
3.9


12.0
1879
12.4


12.4
394
2.6


13.4
3923
25.9


15.1
548
3.6


16.0
196
1.3


16.7
156
1.0


18.4
302
2.0


19.0
2184
14.4


19.4
1644
10.8


19.9
1220
8.0


20.1
959
6.3


20.6
226
1.5


21.2
3809
25.1


21.7
587
3.9


21.9
362
2.4


22.5
749
4.9


23.3
165
1.1


23.7
114
0.8


24.1
80
0.5


25.5
1263
8.3


25.8
545
3.6


26.0
183
1.2


26.4
159
1.1


26.7
420
2.8


27.0
768
5.1


27.8
126
0.8


28.1
66
0.4


28.5
153
1.0


28.9
39
0.3


29.3
478
3.2


30.3
127
0.8


30.8
50
0.3


32.0
806
5.3


32.7
1080
7.1


33.2
155
1.0


33.4
177
1.2


33.8
153
1.0


34.7
260
1.7










TG/DT Analysis


TGA of besylate pattern 1 from tBME showed a total weight loss of approximately 13% from the outset to about 150° C. DTA showed an endothermal event at onset about 241° C. (peak at about 247° C.). TGA of besylate pattern 1 from ethanol showed a total weight loss of approximately 0.4% from the outset to about 250° C. DTA showed an endothermal event at onset about 244° C. (peak at about 248° C.).


Result of Stability Studies


XRPD analysis of post-stability besylate pattern 1 recovered from THF showed an increase to crystallinity but no changes to form after exposure to stability conditions. XRPD analysis of post-stability besylate pattern 1 recovered from TBME showed preferred orientation but no changes to form after exposure to stability conditions. XRPD analysis of post-stability besylate pattern 1 recovered from ethanol showed a decrease in crystallinity after exposure to stability conditions.


Secondary Salt Scale Up


XRPD analysis of besylate scale up showed successful formation of besylate pattern 2 from ethanol seen in the salt screen, a large amount of preferred orientation is seen in the sample.


TGA (FIG. 37) showed a weight loss of approximately 0.7% from the outset up to around 250° C. whilst DTA showed an endothermal “melting” event at onset around 244° C. (peak at around 248° C.).


DSC analysis (FIG. 38) in the first heating cycle showed a sharp endothermal event at onset around 246° C. (peak at 249° C.). This endothermal event is consistent with TG/DTA and no thermal events were seen in the cooling or second heating cycle. Compound 1 besylate exhibits low hygroscopicity when exposed by DVS conditions with a mass uptake of about 0.7% at 90% RH (FIGS. 39 and 40). Post-DVS XRPD analysis shows no changes in crystalline form after exposure, a large amount of preferred orientation is seen in the sample. The hysteresis observed is most likely caused by a small amount of amorphous content which appears to crystallize at 90% RH.


An IR spectrum of Compound 1 besylate was taken for reference which can be found in FIG. 41 with peak lists in Table 23.









TABLE 23





Wave number















3271


3033


2974


2864


2069


1657


1634


1573


1544


1496


1464


1456


1446


1371


1343


1279


1248


1222


1197


1157


1119


1077


1032


1018


994


968


933


923


902


846


828


791


772


757


734


723


708


692


640


628


609


561


549


526


503


476


459


445


415










1H-NMR spectrum shown in FIG. 42 shows 0.88 eq. benzenesulfonic acid, and 0.028 eq. EtOH. UPLC analysis of Compound 1 besylate gave an average purity of 99.4%.


1 week stability tests at 80° C. and under ambient light showed no change to form after exposure and no change to purity. However, by XRPD analysis, the sample held at 40° C./75% RH appears to be a mixture of besylate salt and something else.


Thermodynamic solubility studies of Compound 1 besylate show the salt is highly soluble in pH 1, moderately soluble in 4.5 and unbuffered water. The sample shows low solubility in pH 6.8. pH and concentration values can be found in Table 24.












TABLE 24







Sample ID
Concentration (mg/mL)



















pH 1
30.8



pH 4.5
12.7



pH 6.8
1.9



Un-buffered Water
17.9










XRPD analysis showed insufficient solids were recovered from pH 1, an unknown form was recovered from pH 4.5 and poorly crystalline free base was recovered from pH 6.8 and unbuffered water. Salt disproportionation studies of Compound 1 besylate showed the recovered material to be poorly crystalline free base by XRPD analysis.


Hydration studies of Compound 1 besylate found insufficient solids were recovered from medium water activity and poorly crystalline besylate salt recovered from low and high water activities, by the poor crystallinity of the recovered material and a peak at around 21 degrees indicate the potential of a hydrate formation.


Example 9
Preparation and Characterization of Compound 1 Citric acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing citric acid (10.67 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Scale-Up Preparation from Acetone


About 300 mg of compound 1 was weighed into a vial and 160 mg of citric acid was weighed into a separate vial. To both vials, 3.75 mL of acetone was added and the two mixtures combined. The resulting slurry was then temperature cycled for 24 hours (ambient to 40° C. in 4 hours cycles). The resulting slurry was then allowed to evaporate at ambient temperature to remove excess acetone (1.05 eq. of acid to free base). Observations from the treatment of Compound 1 with citric acid are shown in Table 25 below:











TABLE 25









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of citric acid experiments recovered 6 crystalline hits, free base (Form I) recovered from ethanol, methanol, 2-propanol, and THF and Form I recovered from acetone and TBME (FIG. 21).


XRPD data for Form I is provided in Table 26.











TABLE 26





2-Theta (°)
Height
H %

















6.5
1116
17.6


8.9
4365
68.8


9.2
1294
20.4


11.1
2946
46.5


13.9
1576
24.9


14.4
2604
41.1


15.4
2495
39.3


15.9
1182
18.6


18.0
755
11.9


19.2
2335
36.8


19.6
1370
21.6


20.7
6342
100.0


21.6
4090
64.5


22.3
274
4.3


22.7
348
5.5


23.3
1387
21.9


23.7
962
15.2


24.2
737
11.6


24.8
4022
63.4


25.6
2421
38.2


26.3
533
8.4


26.5
788
12.4


26.8
581
9.2


27.9
927
14.6


28.9
378
6.0


29.1
350
5.5


30.2
533
8.4


30.6
180
2.9


31.8
205
3.2


32.5
365
5.8


33.1
137
2.2


33.7
347
5.5


34.3
151
2.4


34.5
138
2.2










TG/DT Analysis


TGA of citrate Form A showed a total weight loss of approximately 1% from the outset up to about 175° C. DTA showed several endothermal events; first event at onset about 187° C. (peak at about 194° C.) and the second event at onset about 316° C. (peak at about 318° C.).


Result of Stability studies


XRPD analysis of post-stability citrate Form A recovered from acetone showed a decrease to crystallinity but no change to form after exposure to stability conditions. XRPD analysis of post-stability citrate Form A recovered from TBME showed a decrease to crystallinity but no change to form after exposure to stability conditions.


Secondary Salt Scale Up


XRPD analysis of the scaled up citrate salt shows successful formation of citrate Form A from acetone seen in the salt screen.


TGA (FIG. 43) showed a total weight loss of approximately 3% from the outset up to 175° C. DTA showed several endothermal events, the first event at onset around 188° C. (peak at around 194° C.) and the second event at onset around 316° C. (peak at around 318° C.).


DSC analysis in the first heating cycle (FIG. 44) showed a potential overlap of two endothermal events (peaks at 194 and 205° C.). No thermal events were seen in the cooling or second heating cycle.


Compound 1 citrate exhibits low hygroscopicity by DVS analysis (FIG. 45) with a mass uptake of around 1.8% at 90% RH. Post-DVS XRPD analysis of the material showed no changes in crystalline form upon exposure to DVS conditions.


An IR spectrum of Compound 1 citrate was taken for reference which can be found in FIG. 47 with peak lists in Table 27.









TABLE 27





Wave Number















3430


3066


2967


2518


2033


1929


1718


1626


1568


1497


1456


1373


1338


1281


1178


1141


1109


1074


987


929


899


790


771


747


687


667


623


578


550


532


497


478


459


446


425










1H-NMR spectrum shown in FIG. 48 shows 0.97 eq. citric acid and 0.24 eq. acetone. UPLC analysis of Compound 1 citrate gave an average purity of 99.4%. 1 week stability tests at 40° C./75% RH, 80° C. and under ambient light showed no change to form after exposure and no change to purity. Thermodynamic solubility studies of Compound 1 citrate show the salt is highly soluble in un-buffered water and has high solubility at pH 1 with a lower solubility at 4.5 and 6.8. pH and concentration values can be found in Table 28.












TABLE 28







Sample ID
Concentration (mg/mL)



















pH 1
18.6



pH 4.5
0.3



pH 6.8
0.9



Un-Buffered Water
21.0










XRPD analysis showed poorly crystalline solids were recovered from pH 1, Compound 1 citrate was recovered from pH 4.5 and un-buffered water and poorly crystalline free base was recovered from pH 6.8. Salt disproportionation studies of Compound 1 citrate showed the recovered material to be poorly crystalline citrate salt by XRPD analysis.


Hydration studies of Compound 1 citrate found poorly crystalline citrate salt recovered from high and low water activities and unknown form, referred to here as Form B, recovered from medium water activity. XRPD diffractogram of the Compound I citrate Form B is shown in FIG. 49.


Example 10
Preparation and Characterization of Compound 1 Methanesulfonic Acid Salt

A stock solution of methanesulfonic acid was prepared in water (36 μL of methane sulfuric acid in 964 μL H2O). 400 μL of the appropriate solvent was added to the vial containing the weighed compound 1, 100 μL of the methanesulfonic acid stock solution was then added to the solvent/compound 1 slurry (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Scale-Up Preparation from Acetone


About 300 mg of Compound 1 was weighed into a vial and a stock solution of methanesulfonic acid was prepared in water (538 μL of acid in 10 mL of water). To the weighed compound 1, 6 mL of acetone was added which was then followed by 1.5 mL of the acid stock solution, this slurry was then temperature cycled for 24 hours (ambient to 40° C. in 4 hour cycles) (1.05 eq. of acid to free base). The resulting clear solution was allowed to evaporate to recover solids; to which a crystal/oil mixture was recovered. To this mixture, acetone was added and the vial sonicated to produce solids. These solids were then filtered and dried for 72 hours under vacuum at ambient temperature. Observations from the treatment of Compound 1 with methanesulfonic acid are shown in a Table 29 below:











TABLE 29









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Clear
Slurry
Clear
Slurry
Clear




Solution

Solution

Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of methanesulfonic acid experiments recovered 4 crystalline hits, free base (Form I) recovered from THF and pattern 1 from acetone, methanol and 2-propanol (FIG. 16). Insufficient solids were recovered from ethanol and TBME.


TG/DT Analysis


TGA of crystalline mesylate (FIG. 25) showed a total weight loss of approximately 3% from the outset up to about 200° C. DTA showed an endothermal event at onset about 229° C. (peak at about 232° C.).


Result of Stability Studies


XRPD analysis of post-stability crystalline mesylate recovered from acetone showed no changes to crystallinity or form after exposure to stability conditions. XRPD analysis of post-stability crystalline mesylate recovered from methanol showed a decrease in crystallinity but no changes to form after exposure to stability conditions. XRPD analysis of post-stability crystalline mesylate recovered from isopropanol showed a slight increase in crystallinity but no changes to form after exposure to stability conditions.


Secondary Salt Scale Up


XRPD analysis of the scaled up mesylate from acetone (shown in FIG. 30) showed a different form than seen previously.


TGA showed a series of weight losses with a total of around 9% up to 228° C. (FIG. 31). The weight loss seen at around 120° C. indicates the material to be an acetone solvate. DTA (FIG. 31) showed a small endothermal event at onset around 120° C. (peak at around 125° C.). This event is likely associated with the 6.74% weight loss, which would equate to about 0.59 equivalents of acetone. A larger endothermal “melting” event at onset about 228° C. (peak at about 232° C.). This event is consistent with the earlier collected mesylate TG/DTA.


DSC Analysis in the first heating cycle (FIG. 32) showed a sharp endothermal event at onset around 230° C. (peak at 233° C.). This endothermal event shown is consistent with TG/DTA. At this point, the material had already believed to have been desolvated otherwise there should have been an endothermal event relating to the weight loss.


A broad recrystallization event can be seen in the first cooling cycle with an onset of around 193° C. (peak at around 181° C.) and in the second heating cycle showed an endothermal event at onset around 223° C. (peak at 229° C.).


Compound 1 mesylate salt exhibits high hygroscopicity by upon exposure to GVS humidity conditions (FIGS. 33 and 34); mass uptake of about 32% at 90% RH. Post-GVS XRPD analysis of the mesylate salt shows the material to desolvate and become the mesylate form seen in the salt screen. At 30% RH the material deliquesced and upon drying crystallized to the same form seen in the primary salt screen.


An IR spectrum of Compound 1 mesylate salt was taken for reference which can be found in FIG. 35 and peak listings in Table 30.









TABLE 30





Wave Number















3344


3068


3020


2963


2930


2870


1805


1649


1626


1600


1566


1538


1492


1451


1367


1347


1282


1235


1167


1153


1115


1071


1020


991


964


944


923


904


891


859


796


770


740


720


708


687


633


616


552


528


509


469


442


405










1H NMR Spectrum shown in FIG. 36 shows about 1 eq. of sulfonic acid. It is not possible to accurately quantify any residual acetone from this data due to spectral overlap but the levels, if there are any, are considered low.


UPLC analysis of Compound 1 mesylate gave an average purity of 99.4%.


1 Week stability tests at 40° C./75% RH, 80° C. and under ambient light showed change to form after exposure by XRPD. However, changes to the mesylate form seen previously in the salt screen and no change to purity.


Thermodynamic solubility studies of Compound 1 mesylate show the salt is moderately soluble in pH 1, 4.5 and unbuffered water. The sample shows low solubility in pH 6.8. pH and concentration values can be found in Table 31.












TABLE 31







Sample ID
Concentration (mg/mL)



















pH 1
14.3



pH 4.5
9.3



pH 6.8
1.5



Unbuffered Water
9.6










XRPD analysis showed insufficient solids were recovered from pH 1, mesylate salt was recovered from pH 4.5 and free base was recovered from pH 6.8 and un-buffered water. Salt disproportionation studies of Compound 1 mesylate showed no change to form by XRPD analysis but crystallinity reduced. Hydration studies of Compound 1 mesylate showed mesylate salt recovered from medium water activities, a mixture of free base and salt recovered from low water activities and free base recovered from high water activities.


Example 11
Preparation and Characterization of Compound 1 1,2-Ethane Disulfonic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing 1,2-ethane disulfonic acid (12.94 mg). The solutions/slurries were then added to the solvent/API solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with 1,2-ethane disulfonic acid are shown in Table 32 below:











TABLE 32









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Yellow
Clear
Slurry
Slurry
Slurry
Slurry



solution/
solution



dark



solids









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of 1,2-ethane disulfonic acid experiments recovered 4 crystalline hits, free base (Form I) recovered from acetone, THF and TBME and pattern 1 from 2-propanol (FIG. 14). Insufficient solids were recovered from ethanol and methanol.


Result of Stability studies


XRPD analysis of post-stability edisylate recovered from isopropanol showed the material to become amorphous after exposure to stability conditions.


Example 12
Preparation and Characterization of Compound 1 p-Toluene Sulfonic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing p-toluene sulfonic acid (10.84 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of compound 1 with p-toluene sulfonic acid are shown in Table 33 below:











TABLE 33









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Solid
Clear
Slurry
Slurry
Slurry
Solid




Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of p-toluene sulfonic acid experiments recovered 4 crystalline hits, free base (Form I) recovered from 2-propanol and TBME and pattern 1 from acetone and THF (FIG. 15). Insufficient solids were recovered from ethanol and methanol.


TG/DT Analysis


TGA of p-toluene sulfonate (FIG. 24) showed a total weight loss of approximately 14% from the outset up to about 250° C. DTA showed an endothermal event at onset about 84° C. (peak at about 90° C.).


Result of Stability Studies


XRPD analysis of post-stability p-toluene sulfonate recovered from acetone showed the material to become amorphous after exposure to stability conditions.


Example 13
Preparation and Characterization of Compound 1 Oxalic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing oxalic acid (5.08 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with oxalic acid are shown in Table 34 below:











TABLE 34









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Solid
Slurry
Slurry
Slurry









XRPD analysis of oxalic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from acetone (which was mostly amorphous), 2-propanol, THF and TBME and pattern 1 recovered from ethanol and methanol (FIG. 19).


TG/DT Analysis


TGA of oxalate (FIG. 26) showed a total weight loss of approximately 17% from the outset up to about 300° C. DTA showed a small endothermal event at onset about 314° C. (peak at about 317° C.).


Result of Stability Studies


XRPD analysis of post-stability oxalate recovered from ethanol showed a change in crystallinity and form after exposure to stability conditions. XRPD analysis of post-stability oxalate recovered from methanol showed no change to crystallinity however, changes to form were seen after exposure to stability conditions.


Example 14
Preparation and Characterization of Compound 1 Fumaric Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing fumaric acid (6.48 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with fumaric acid are shown in Table 35 below











TABLE 35









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of fumaric acid experiments recovered 6 crystalline hits, free base (Form I) recovered from ethanol, methanol, 2-propanol, THF and TBME and pattern 1 recovered from acetone (FIG. 20).


TG/DT Analysis


TGA of crystalline fumarate (FIG. 27) showed a total weight loss of approximately 22% from the outset up to about 250° C. DTA showed several endothermal events; first event at onset about 164° C. (peak at about 166° C.), second event at onset about 189° C. (peak at about 191° C.), third event at onset of about 198° C. (peak at about 201° C.) and forth event at onset about 310° C. (peak at about 312° C.).


Result of Stability Studies


XRPD analysis of post-stability crystalline fumarate recovered from acetone showed a slight decrease to crystallinity however, no change to form after exposure to stability conditions.


Example 15
Preparation and Characterization of Compound 1 L-Malic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing L-malic acid (7.49 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with L-malic acid are shown in Table 36 below:











TABLE 36









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Solid









XRPD analysis of L-malic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from acetone (with a large amount of preferred orientation), ethanol, methanol, 2-propanol, and THF and pattern 1 recovered from TBME (FIG. 22).


TG/DT Analysis


TGA of crystalline L-malate (FIG. 28) showed a total weight loss of approximately 26% from the outset up to about 250° C. DTA showed several endothermal events; first event at onset about 158° C. (peak at about 162° C.) and the second event at onset about 310° C. (peak at about 313° C.).


Result of Stability Studies


XRPD analysis of post-stability crystalline L-malate prepared from TBME showed no change to crystallinity and form after exposure to stability conditions.


Example 16
Preparation and Characterization of Compound 1 Succinic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing succinic acid (6.59 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with succinic acid are shown in Table 37 below:











TABLE 37









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of succinic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from ethanol, methanol, 2-propanol, THF and TBME and pattern 1 recovered from acetone (FIG. 23).


TG/DT Analysis


TGA of succinate FIG. 29) showed a total weight loss of approximately 22% from the outset about 210° C. DTA showed several endothermal events; first event at onset about 147° C. (peak at about 151° C.) and the second event at onset about 315° C. (peak at about 315° C.).


Result of Stability Studies


XRPD analysis of post-stability crystalline succinate recovered from acetone showed a decrease in crystallinity but no change to form after exposure to stability conditions.


Example 17
Preparation and Characterization of Compound 1 Hydrochloric Acid Salt

A stock solution of HCl was prepared in water (46 μL of HCl in 954 μL H2O). 400 μL of the appropriate solvent was added to the vial containing the weighed compound 1, 100 μL of the HCl stock solution was then added to the solvent/compound 1 slurry (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with HCl are shown in Table 38 below:











TABLE 38









Solvent













Time-point
Acetone
Ethanol
Methanol
2-Propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Clear
Clear
Clear
Clear
Slurry




Solution
Solution
Solution
Solution









To the samples which were recovered as clear solutions, 2-3 mg of compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. Further solids were recovered from ethanol, methanol, 2-propanol, TBME and THF through anti-solvent additions described in Materials and methods section. XRPD analysis of HCl experiments recovered 6 crystalline hits. Freebase (Form I) was recovered from all solvent systems analyzed.


Example 18
Preparation and Characterization of Compound 1 Sulfuric Acid Salt

A stock solution of sulfuric acid was prepared in water (31 μL of sulfuric acid in 969 μL H2O). 400 μL of the appropriate solvent was added to the vial containing the weighed compound 1, 100 μL of the sulfuric acid stock solution was then added to the solvent/compound 1 slurry (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with sulfuric acid are shown in Table 39 below:











TABLE 39









Solvent

















2-




Time-point
Acetone
Ethanol
Methanol
propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-
Clear
Clear
Clear
Clear
Clear
Clear


Cycling
solution
solution
solution
solution
solution
solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. Further solids were recovered from ethanol, methanol, 2-propanol, TBME and THF through anti-solvent additions described. XRPD analysis of sulfuric acid experiments recovered 6 amorphous hits from all solvent systems analyzed.


Example 19
Preparation and characterization of Compound 1 Naphthalene-2-Sulfonic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing naphthalene-2-sulfonic acid (14.14 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with naphthalene-2-sulfonic acid are shown in Table 40 below.











TABLE 40









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Clear
Clear
Clear
Clear
Slurry
Solid



Solution
Solution
Solution
Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of naphthalene-2-sulfonic acid experiments recovered 3 crystalline hits, free base (Form I) recovered from ethanol, THF and TBME. Insufficient solids were recovered from acetone, methanol and 2-propanol.


Example 20
Preparation and Characterization of Compound 1 2-Hydroxy Ethanesulfonic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing 2-hydroxy ethanesulfonic acid (8.19 mg). The solutions/slurries were then added to the solvent/compound 1 solution. The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with 2-hydroxy-ethanesulfonic acid are shown in Table 41 below:











TABLE 41









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Solid
Slurry
Slurry
Slurry









XRPD analysis of 2-hydroxy ethanesulfonic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 21
Preparation and characterization of Compound 1 L-Aspartic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing L-aspartic acid (7.36 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with L-aspartic acid are shown in Table 42 below:











TABLE 42









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of L-aspartic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 22
Preparation and Characterization of Compound 1 Maleic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing maleic acid (6.48 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with maleic acid are shown in Table 43 below:











TABLE 43









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Solid
Slurry
Gum
Slurry
Slurry
Slurry









XRPD analysis of maleic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 23
Preparation and Characterization of Compound 1 Phosphoric Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing phosphoric acid (5.42 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with phosphoric acid are shown in Table 44 below:











TABLE 44









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Solid
Clear
Clear
Slurry
Slurry
Slurry




Solution
Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of phosphoric acid experiments recovered 3 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 24
Preparation and Characterization of Compound 1 Ethanesulfonic Acid Salt

A stock solution of ethane sulfonic acid was prepared in water (47 μL of sulfuric acid in 953 μL H2O). 400 μL of the appropriate solvent was added to the vial containing the weighed compound 1, 100 μL of the ethane sulfonic acid stock solution was then added to the solvent/compound 1 slurry (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with ethanesulfonic acid are shown Table 45 below:











TABLE 45









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Clear
Clear
Clear
Clear
Clear
Slurry



Solution
Solution
Solution
Solution
Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of ethanesulfonic acid experiments recovered 4 crystalline hits, free base (Form I) recovered from acetone, THF and TBME. Insufficient solids were recovered from methanol, ethanol and 2-propanol.


Example 25
Preparation and characterization of Compound 1 L-Glutamic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing L-glutamic acid (8.13 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with L-glutamic acid are shown in Table 46 below:











TABLE 46









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of L-glutamic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 26
Preparation and Characterization of Compound 1 L-Tartaric Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing L-tartaric acid (8.34 mg). The solutions/slurries were then added to the solvent/compound 1 solution. The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with L-tartaric acid are shown in Table 47 below:











TABLE 47









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of L-tartaric acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 27
Preparation and Characterization of Compound 1 D-Glucuronic Acid salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing D-glucuronic acid (10.73 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with D-glucuronic acid are shown in Table 48 below:











TABLE 48









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of D-glucuronic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 28
Preparation and Characterization of Compound 1 Hippuric Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing hippuric acid (10.1 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with hippuric acid are shown in Table 49 below:











TABLE 49









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of hippuric acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 29
Preparation and Characterization of Compound 1 D-Gluconic Acid Salt

A stock solution of D-gluconic acid was prepared in water (176 μL of D-gluconic acid in 824 μL H2O). 400 μL of the appropriate solvent was added to the vial containing the weighed compound 1, 100 μL of the D-gluconic stock solution was then added to the solvent/compound 1 slurry (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with D-gluconic acid are shown in Table 50 below:











TABLE 50









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Clear
Clear
Clear
Clear
Clear
Slurry



Solution
Solution
Solution
Solution
Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of D-gluconic acid experiments recovered 1 crystalline hit, free base (Form I) recovered from TBME and insufficient solids recovered from acetone, ethanol, methanol, 2-propanol and THF.


Example 30
Preparation and Characterization of Compound 1 DL-Lactic Acid Salt

A stock solution of DL-lactic acid was prepared in water (48 μL of DL-lactic acid in 952 μL H2O). 400 μL of the appropriate solvent was added to the vial containing the weighed compound 1, 100 μL of the DL-lactic acid stock solution was then added to the solvent/compound 1 slurry (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with DL-lactic acid are shown in Table 51 below:











TABLE 51









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Clear
Clear
Clear
Clear
Gum
Slurry



Solution
Solution
Solution
Solution









To the samples which were recovered as clear solutions, 2-3 mg of Compound 1 was added to produce a mobile slurry and the sample temperature cycled for a further 2-3 hours. XRPD analysis of DL-lactic acid experiments recovered 5 crystalline hits, free base (Form I) recovered from acetone, ethanol, methanol, THF and TBME. Insufficient solids recovered from 2-propanol.


Example 31
Preparation and Characterization of Compound 1 L-Ascorbic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of Compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing L-ascorbic acid (9.73 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with L-ascorbic acid are shown in Table 52 below:











TABLE 52









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of L-ascorbic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 32
Preparation and Characterization of Compound 1 Benzoic Acid Salt

250 μL of the appropriate solvent was added to the vials containing 20 mg of compound 1. In a separate vial, 250 μL of the appropriate solvent was added to the vial containing benzoic acid (6.82 mg). The solutions/slurries were then added to the solvent/compound 1 solution (1.05 eq. of acid to free base). The samples were then temperature cycled between ambient and 40° C. in 4 hour cycles over 24 hrs.


Observations from the treatment of Compound 1 with benzoic acid are shown in Table 53 below:











TABLE 53









Solvent













Time-point
Acetone
Ethanol
Methanol
2-propanol
TBME
THF





Pre-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry


Post-Cycling
Slurry
Slurry
Slurry
Slurry
Slurry
Slurry









XRPD analysis of benzoic acid experiments recovered 6 crystalline hits, free base (Form I) recovered from all solvent systems analyzed.


Example 33
Solubility Measurements

General method to measure the thermodynamic aqueous solubility of a crystalline, organic solid.


Preparation of Reagents:


1. Aqueous Phosphate Buffer Solution (PBS): pH 7.4, 30 mM


1 Liter of 0.05 M (50 mM) Phosphate Buffer (25° C.) at pH 7.4 was prepared as follows: 11.2 mL of 1 M potassium phosphate (monobasic) was mixed with 38.8 mL of 1 M potassium phosphate (dibasic) and diluted to one liter with water.


To make a 0.03 M (30 mM) Phosphate Buffer (25° C.) at pH 7.4, the above recipe was adjusted as follows: 6.72 mL of 1 M potassium phosphate (monobasic) were mixed with 23.28 mL of 1 M potassium phosphate (dibasic) and diluted to one liter with water. The pH was adjusted with HCl or NaOH. Alternatively, 0.9144 g (6.72×10−3 mol) of potassium phosphate monobasic were mixed with 4.0908 g (23.28×10−3 mol) of potassium phosphate dibasic and diluted to 1 L with water to get a 30 mM solution of Phosphate Buffer (25° C.).


2. Aqueous phosphate buffer, 30 mM, adjusted to pH 6.5


3. Aqueous 0.1N HCl with 0.2% NaCl, adjusted to pH 1.2


4. If necessary, phosphate buffers of other pHs or other strengths could be made by adjusting the recipe above.


5. Suitable organic solvents (ACN, methanol, etc.) were used for preparation of stock and standards.


Instrumentation Used:


1. Balance


2. Agitator for mixing


3. Pipets


4. Filters or centrifuge


5. HPLC w/UV and MS detection (Waters Acquity UPLC with PDA and ZQ MS)


Standards:


1. ˜0.5 mg of compound were dissolved in 2.5% DMSO/MeOH (or other organic) to a final concentration of 250 μg/mL stock


2. 50 uL of stock were accurately pipetted into 96 Shallow well plate containing 200 uL methanol for high standard concentration of 50 ug/mL.


3. 50 uL of 50 μg/mL high standard were accurately pipetted into adjacent well containing 200 uL methanol for medium standard concentration of 10 ug/mL.


4. 50 uL of 10 μg/mL medium standard were accurately pipetted into adjacent well containing 200 uL methanol for low standard concentration of 2 ug/mL.


Sample Preparation:




  • 1. ≥0.5 mg of compound were accurately weighed into a 4-mL vial for each pH to be tested.

  • 2. The appropriate amount of desired buffer was added to the appropriate vials to get a resulting concentration of 1.02 mg/mL.

  • 3. Vials were capped and shaken at 350 rpm for 24 hours at room temperature.

  • 4. ˜450 μL of sample solutions were pipetted from 4-mL vials into 96 DWP.

  • 5. The plate was centrifuged at 3500 rpm for 10 minutes at 20° C. and 250 μL of supernatant were transferred to catch plate.

  • 6. 125 μL of supernatant were pipetted into 96 shallow well plate containing 125 μL of methanol and mixed to get a 2× dilution of the sample.

  • 7. 50 μL of 2× dilution were pipetted into 96 shallow well plate containing 200 μL of methanol and mixed to obtain a 10× dilution of sample.

  • 8. 50 μL of 10× dilution were pipetted into 96 shallow well plate containing 200 μL of methanol and mixed to obtain a 50× dilution of sample.


    Analysis:


    Data Collection:



1. Each standard (2, 10, 50 μg/mL) and sample (2×, 10×, 50× dilution) was injected in triplicate using a 3 μL injection volume on the UPLC, starting with the lowest concentrations and going to the highest. A standard, linear, rapid gradient method and 220 nm and 254 nm UV detection were used with the appropriate mobile phase and column.


2. The UV peak areas of the analyte were integrated and recorded for each chromatogram. If MS data was available, the mass of the parent peak was confirmed for each sample.


3. The responses for the standards were fitted using a y=mx linear model (through zero).


4. The model was used to quantify the amount of compound in the aqueous solutions. The values of the lowest dilution sample that fit within the standard curve was reported.


Note: Adjustments could be made to the above instructions if appropriate for a given compound.


Example 34
In Vitro Metabolic Stability of the Compound of Formula I (Compound 1) and the Compound for formula I′

Abbreviation Description


° C. Degrees centigrade


CLh Predicted hepatic clearance


CLnit Predicted intrinsic clearance


DMEM Dulbecco's modified Eagle's medium


DMPKCP Drug Metabolism, Pharmacokinetics and Clinical Pharmacology


DMSO Dimethyl sulfoxide


ESI+ Electrospray ionization positive mode


ELN Electronic Laboratory Notebook


ER Extraction ratio


fu Unbound fraction of compound


g Gravity


HCl Hydrochloride


HPLC High performance liquid chromatography


IPA Isopropyl alcohol


IS Internal standard


K2HPO4 Potassium phosphate, dibasic


KH2PO4 Potassium phosphate, monobasic


km Rate of loss of test compound


KPB Potassium phosphate buffer


MgCl2 Magnesium chloride


NADP+ Nicotinamide adenine dinucleotide phosphate


NADPH Nicotinamide adenine dinucleotide phosphate reduce


NaOH Sodium hydroxide


NRS NADPH-regenerating solution


LC-MS/MS Liquid chromatography with tandem mass spectrometric detection


m/z Mass to charge ratio


μL Microliter


μM Micromolar


mM Millimolar


MRM Multiple reaction monitoring


n Number considered for assessment


PAR Peak area ratio


% REM Percent remaining


Abbreviation Description


rpm Revolutions per minute


t1/2 Half-life


In vitro metabolic stabilities were studied at a concentration of 1 μM in the presence of liver microsomes and isolated hepatocytes.


Materials


The following reagents were required for experimentation: acetonitrile (HPLC grade, Burdick & Jackson, Madison, Wis.), potassium phosphate (KH2PO4 and K2HPO4, anhydrous, Sigma-Aldrich, Co., St. Louis, Mo.), magnesium chloride (MgCl2, Sigma-Aldrich), water (HPLC grade, JT Baker, Phillipsburg, N.J.), isopropanol (IPA, reagent grade, EMD Chemicals, Gibbstown, N.J.), formic acid (reagent grade, Sigma-Aldrich), and dimethyl sulfoxide (DMSO; reagent grade, EM Science, Gibbstown, N.J.). Labetalol (Sigma-Aldrich) was used as an internal standard (IS) for analytical purposes. Human liver microsomes were purchased from Corning Life Sciences (Tewksbury Mass.), lot BD38289 (150-donor mixed gender pool, human). Liver microsomes from Sprague Dawley rat were purchased from XenoTech, LLC (Lenexa, Kans.). Lot numbers were XT1110042 and XT1310214. Cryopreserved human hepatocytes were either purchased from Invitrogen/CellzDirect (Pittsboro, N.C.) and lot number HUP50 or purchased from In Vitro ADMET Laboratories, LLC (Malden, Miss.) and lot number PHS9001 was used (10-donor mixed gender pool, human). Rat cryopreserved hepatocytes were purchased from Bioreclamation/In Vitro Technologies (Baltimore, Md.) and were from pooled male donors unless otherwise noted. Lots OGN, PZG and MSO were used for this study. All other reagents, control compounds, and solvents were of the highest analytical grade supplied by Sigma (St. Louis, Mo.).


Methods


Liver Microsomal Incubations


A 100 mM potassium phosphate assay buffer solution (KPB) was prepared as follows. Both KH2PO4 and K2HPO4 were dissolved separately in reagent grade water resulting in final concentrations 100 mM. A 75:25 mixture v/v of K2HPO4:KH2PO4 was prepared and the pH of the solution was adjusted to 7.4 using diluted HCl or diluted NaOH solutions. A stock solution of test compound was prepared at 10 mM (active compound) in DMSO. The stock solution was diluted immediately before use to 2.5 μM using the KPB solution to create the working standard. All test compounds were completely soluble in DMSO by visual inspection at room temperature. The NADPH-regenerating solution (NRS) was prepared on the day of analysis by diluting one volume of 17 mg/mL NADP+ with one volume of 78 mg/mL glucose-6-phosphate (both prepared in KPB, pH 7.4) and 7.9 volumes of 20 mM MgCl2. The final concentrations of NADP+ and glucose-6-phosphate were 1.7 mg/mL and 7.8 mg/mL, respectively. Immediately prior to use, the NRS was activated by the addition of 10 μL of glucose-6-phosphate dehydrogenase (150 Units/mL in KPB, pH 7.4) per mL of NRS stock solution. Liver microsomes were diluted to 2.5 mg protein/mL using KPB.


For the compound of formula I or the compound of formula I′ or each positive control (i.e., dextromethorphan, diazepam, diltiazem, phenacetin, tolbutamide, and verapamil), 20 μL of 2.5 μM working standard solution of test compound and 20 μL of microsomes (2.5 mg protein/mL) were added to each well of a 96-well polypropylene plate (Costar, VWR, West Chester, Pa.) in duplicate. The plates were placed in an incubator at 37° C. for 5 minutes before adding the start solution. A 10-μL aliquot of the NRS solution was added to each original well to initiate metabolism. The concentration of the test compound at the beginning of the incubation was 1 μM. One incubation plate was prepared for each time point (i.e., 0 and 20 minutes). Incubations were conducted at 37° C. and 100% relative humidity. At each time point, the appropriate incubation plate was removed from the incubator and a solution containing internal standard (150 μL, 0.25 μM labetalol in 60% acetonitrile) was added to each well. The plate was immediately spun in a centrifuge at 2,095×g for 7 minutes at room temperature using an Allegra benchtop centrifuge (Beckman Coulter, Fullerton, Calif.). A 200-μL aliquot of the supernatant was transferred from each well to a 96-well shallow plate (Costar). The plates were sealed using disposable plate mats.


Hepatocyte Incubations


A stock solution of the test compound was prepared at 10 mM (active compound) in DMSO. The in vitro stability of the test compound (1 μM) was assessed in the presence of hepatocytes as follows. Cryopreserved hepatocytes were thawed, isolated from shipping media and diluted to a density of 1×106 viable cells/mL, according to the supplier's guidelines, using Dulbecco's Modified Eagle Medium, 1×, high glucose (DMEM, Invitrogen, Carlsbad, Calif.). Viability was determined by trypan blue exclusion using a hemocytometer (3500 Hausser, VWR, West Chester, Pa.). The 10 mM stock solution of test compound was diluted to 2 μM using supplemented DMEM to create the working standard. A 20-μL aliquot of test compound or control (i.e., antipyrine, diazepam, diltiazem, lorazepam, propranolol, verapamil, and 7-ethyl-10-hydroxycamptothecin (SN-38)) was added to each test well of a 96-well polypropylene plate (Costar, VWR, West Chester, Pa.) immediately followed by the addition of 20 μL of the hepatocyte suspension. One incubation plate was prepared for each time point (i.e., 0, 60 and 120 minutes) with samples being prepared in duplicate. Incubations were conducted at 37° C. and 100% relative humidity. At each time point, the appropriate incubation plate was removed from the incubator and a solution containing IS (200 μL,


0.2 μM labetalol in 60% acetonitrile) was added to each well. The plate was mixed at 600 rpm for 2 minute on a plate shaker (IKA MTS 2/4 Digital Microtiter Shaker, VWR) and immediately spun in a centrifuge at 2,095×g for 10 minutes at room temperature using an Allegra benchtop centrifuge (Beckman Coulter, Fullerton, Calif.). A 200-μL aliquot of the supernatant was transferred from each well to a 96-well shallow plate (Costar). The plates were sealed using disposable plate mats.


Analytical Quantitation


The LC-MS/MS system was comprised of an HTS-PAL autosampler (Leap Technologies, Carrboro, N.C.), an HP1200 HPLC (Agilent, Palo Alto, Calif.), and an API4000 triple quadrupole mass spectrometer (PE Sciex, a division of Applied Biosystems, Foster City, Calif.). Chromatographic separation of the analyte and internal standard was achieved at room temperature using a C18 column (Kinetex®, 30×3.0 mm, 2.6 μm particle size, Phenomenex, Torrance, Calif.) in conjunction with gradient conditions using mobile phases A (aqueous 0.1% formic acid with 1% isopropyl alcohol) and B (0.1% formic acid in acetonitrile). The total run time, including re-equilibration, for a single injection was 2 minutes. Mass spectrometric detection of the analytes was accomplished using the ESI+ ionization mode. Ion current was optimized during infusion of a stock solution of test compound. Analyte responses were measured by multiple reaction monitoring (MRM) of transitions unique to each compound.


Data were acquired and peak areas were calculated for test compounds and the internal standard using Analyst 1.6.1 software (Sciex). For the liver microsomal and hepatocyte stability assessments, peak area tables were exported to BioAssay Enterprise (CambridgeSoft, Cambridge, Mass.), where the average analyte-to-internal standard peak area ratios were used to calculate percent remaining (% REM), half-life (t½), predicted hepatic clearance (CLh) and predicted hepatic extraction ratio (ER).


Calculations


All calculations were performed using BioAssay Enterprise. The mean peak area ratios were calculated by averaging the peak area ratios (n=2) of test compound and the internal standard for each sample. Percent remaining was calculated by determining the ratio of the peak area ratio at each time point to the peak area ratio of the time-zero samples. The rate of loss of test compound (km) was determined by linear regression of—1n(f(t)) versus time. The regression used the form “y=mx”, therefore the model forced an intercept of 100% remaining and assumed that the metabolism followed first order kinetics. The t½ was determined dividing 1n(2) by km. The predicted intrinsic clearance (CLint) was calculated by scaling the in vitro half-life for stability of test compound using physical and physiological scaling factors listed in Table 5.1 and employed in the following equation:







C


L

i

n

t



=



ln

2


t

1
/
2





(


D
·
w

C

)







where D is the number of hepatocytes per mass of liver for a particular species. W is the average mass of liver present per weight of animal, and C is the number of hepatocytes present during the incubations per unit volume. The CLh was calculated using the following equation:







CL
h

=


C



L

i

n

t


·
Q




C


L

i

n

t



+
Q







where Q is the species-dependent hepatic blood flow. No adjustment was made for the unbound fraction of the test compound (fu). The ER was determined by calculating the ratio of the CLh to Q:






ER
=


C


L
h


Q






In Vitro Stability in Liver Microsomes


Test compounds were incubated with liver microsomes from Sprague Dawley rats and humans. Control compounds (i.e., dextromethorphan, diazepam, diltiazem, phenacetin, tolbutamide, and verapamil) performed within expected limits with respect to the fraction remaining after incubation in each liver microsomal system. The percentage of the compound of formula I and of the compound of formula I′ remaining after 20 minutes, calculated t½ values, the predicted clearance values, and the predicted hepatic ERs were determined.


In Vitro Stability in Hepatocytes


Test compounds were incubated with hepatocytes from Sprague Dawley rats and humans. Control compounds (i.e., antipyrine, diazepam, diltiazem, lorazepam, propranolol, verapamil, and 7-ethyl-10-hydroxycamptothecin) performed within expected limits with respect to the fraction remaining after incubation in each hepatocyte system. The percentage of the compound of formula I and of the compound of formula I′ remaining after 2 hours, calculated t½ values, the predicted clearance values, and the predicted hepatic extraction ratios as determined in hepatocyte incubations for each species were determined.


Example 35

MDR1 LLC-PK1 Cell Culture and Experimental Conditions


Both LLC-PK1 and MDR1 transfected LLC-PK1 cells were cultured and plated according to manufacturer's recommendations with the exception that the passage media contained only 2% fetal bovine serum so as to extend passage time out to seven days.


Both positive and negative controls were used to assess functionality of P-gp efflux in the assay. Stock solutions for assay controls and the test article were prepared in DMSO for a final test concentrations of 10 μM. Final organic concentration in the assay was 1%. All dosing solutions contained 10 μM lucifer yellow to monitor LLC-PK1 cell monolayer integrity.


For the apical to basolateral determination (A to B), 75 μL of the test article in transport buffer were added to the apical side of the individual transwells and 250 μL of basolateral media, without compound or lucifer yellow, were added to each well. For the basolateral to apical determination (B to A), 250 μL of test article in transport buffer were added to each well and 75 transport buffer, without compound or lucifer yellow, were added to each transwell. All tests were performed in triplicate, and each compound was tested for both apical to basolateral and basolateral to apical transport. The plates were incubated for 2 hours on a Lab-Line Instruments Titer Orbital Shaker (VWR, West Chester, Pa.) at 50 rpm and 37° C. with 5% CO2. All culture plates were removed from the incubator and 50 μL of media were removed from the apical and basolateral portion of each well and added to 150 μL of 1 μM labetalol in 2:1 acetonitrile (ACN): H2O, v/v. The plates were read using a Molecular Devices (Sunnyvale, Calif.) Gemini Fluorometer to evaluate the lucifer yellow concentrations at excitation/emission wavelengths of 425/535 nm. These values were accepted when found to be below 5% for apical to basolateral and basolateral to apical flux across the MDR1 transfected LLC-PK1 cell monolayers. The plates were sealed and the contents of each well analyzed by LC-MS/MS. The compound concentrations were determined from the ratio of the peak areas of the compound to the internal standard (labetalol) in comparison to the dosing solution.


LC-MS Analysis


The LC-MS/MS system was comprised of an HTS-PAL autosampler (Leap Technologies, Carrboro, N.C.), an HP1200 HPLC (Agilent, Palo Alto, Calif.), and a MDS Sciex 4000 Q Trap system (Applied Biosystems, Foster City, Calif.). Chromatographic separation of the analyte and internal standard was achieved at room temperature using a C18 column (Kinetics®, 30×3 mm, 2.6 μm particle size, Phenomenex, Torrance, Calif.) in conjunction with gradient conditions using mobile phases A (water containing 1% isopropyl alcohol and 0.1% formic acid) and B (0.1% formic acid in ACN). The total run time, including re-equilibration time, for a single injection was 1.2 minutes. Mass spectrometric detection of the analytes was accomplished using the ion spray positive mode. Analyte responses were measured by multiple reaction monitoring (MRM) of transitions unique to each compound (the protonated precursor ion and selected product ions for each test article and m/z 329 to m/z 162 for labetalol, the internal standard).


Determination of Apparent Permeability (Papp)


Permeability (Papp) was calculated in BioAssay v. 9.0 (Cambridge Soft, Cambridge, Mass.) using the following equation:








P

a

p

p




(

×

10

-
6








cm
/
s


)


=


(


C
d

·
V
·

(

1
×

10
5
6


)


)


(


t
·
0.12








cm
2

·
C


)







where Cd, V, t and C0 are the detected concentration (μM), the volume on the dosing side (mL), the incubation time (s) and the initial dosing concentration (μM), respectively. The calculations for Papp were made for each replicate and then averaged.


Other Embodiments

It is to be understood that while the present application has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the present application, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims
  • 1. A crystalline form of (6R,15R)-9-fluoro-15-methyl-2,11,16,20,21,24-hexaazapentacyclo[16.5.2.02,6.07,12.021,25]pentacosa-1(24),7,9,11,18(25),19,22-heptaen-17-one having the following structure:
  • 2. The crystalline form of claim 1, wherein the crystalline form is characterized by XRPD peaks, in terms of 2-theta, at 9.1, 20.2, and 24.9.
  • 3. The crystalline form of claim 2, having one or more additional XRPD peaks, at 11.2, 13.4, 14.8, 18.3, 18.6, 23.6, and 29.4.
  • 4. The crystalline form of claim 2, having hygroscopicity characterized by a mass uptake of about 0.3% at 90% relative humidity as determined by dynamic vapor sorption analysis.
  • 5. The crystalline form of claim 2, which is substantially free of an amorphous form.
  • 6. A process for making a pharmaceutical composition comprising mixing a salt or crystalline form according to any one of claim 1 and a pharmaceutically acceptable carrier.
  • 7. A method of treating a cancer mediated by a Trk kinase in a subject in need thereof, comprising administering the crystalline form according to claim 1 to the subject.
  • 8. The method according to claim 7, wherein the cancer is selected from the group consisting of adenocarcinoma, adrenal gland cortical carcinoma, adrenal gland neuroblastoma, anus squamous cell carcinoma, appendix adenocarcinoma, bladder urothelial carcinoma, bile duct adenocarcinoma, bladder carcinoma, bladder urothelial carcinoma, bone chordoma, bone marrow leukemia lymphocytic chronic, bone marrow leukemia non-lymphocytic acute myelocytic, bone marrow lymph proliferative disease, bone marrow multiple myeloma, bone sarcoma, brain astrocytoma, brain glioblastoma, brain medulloblastoma, brain meningioma, brain oligodendroglioma, breast adenoid cystic carcinoma, breast carcinoma, breast ductal carcinoma in situ, breast invasive ductal carcinoma, breast invasive lobular carcinoma, breast metaplastic carcinoma, cervix neuroendocrine carcinoma, cervix squamous cell carcinoma, colon adenocarcinoma, colon carcinoid tumor, duodenum adenocarcinoma, endometrioid tumor, esophagus adenocarcinoma, eye intraocular melanoma, eye intraocular squamous cell carcinoma, eye lacrimal duct carcinoma, fallopian tube serous carcinoma, gallbladder adenocarcinoma, gallbladder glomus tumor, gastroesophageal junction adenocarcinoma, head and neck adenoid cystic carcinoma, head and neck carcinoma, head and neck neuroblastoma, head and neck squamous cell carcinoma, kidney chromophore carcinoma, kidney medullary carcinoma, kidney renal cell carcinoma, kidney renal papillary carcinoma, kidney sarcomatoid carcinoma, kidney urothelial carcinoma, leukemia lymphocytic, liver cholangiocarcinoma, liver hepatocellular carcinoma, lung adenocarcinoma, lung adenosquamous carcinoma, lung atypical carcinoid, lung carcinosarcoma, lung large cell neuroendocrine carcinoma, lung non-small cell lung carcinoma, lung sarcoma, lung sarcomatoid carcinoma, lung small cell carcinoma, lung small cell undifferentiated carcinoma, lung squamous cell carcinoma, lymph node lymphoma diffuse large B cell, lymph node lymphoma follicular lymphoma, lymph node lymphoma mediastinal B-cell, lymph node lymphoma plasmablastic lung adenocarcinoma, lymphoma follicular lymphoma, lymphoma, non-Hodgkin's lymphoma, nasopharynx and paranasal sinuses undifferentiated carcinoma, ovary carcinoma, ovary carcinosarcoma, ovary clear cell carcinoma, ovary epithelial carcinoma, ovary granulosa cell tumor, ovary serous carcinoma, pancreas carcinoma, pancreas ductal adenocarcinoma, pancreas neuroendocrine carcinoma, peritoneum mesothelioma, peritoneum serous carcinoma, placenta choriocarcinoma, pleura mesothelioma, prostate acinar adenocarcinoma, prostate carcinoma, rectum adenocarcinoma, rectum squamous cell carcinoma, skin adnexal carcinoma, skin basal cell carcinoma, skin melanoma, skin Merkel cell carcinoma, skin squamous cell carcinoma, small intestine adenocarcinoma, small intestine gastrointestinal stromal tumors (GISTs), soft tissue angiosarcoma, soft tissue Ewing sarcoma, soft tissue hemangioendothelioma, soft tissue inflammatory myofibroblastic tumor, soft tissue leiomyosarcoma, soft tissue liposarcoma, soft tissue neuroblastoma, soft tissue paraganglioma, soft tissue perivascular epitheliod cell tumor, soft tissue sarcoma, soft tissue synovial sarcoma, stomach adenocarcinoma, stomach adenocarcinoma diffuse-type, stomach adenocarcinoma intestinal type, stomach adenocarcinoma intestinal type, stomach leiomyosarcoma, thymus carcinoma, thymus thymoma lymphocytic, thyroid papillary carcinoma, unknown primary adenocarcinoma, unknown primary carcinoma, unknown primary malignant neoplasm, unknown primary melanoma, unknown primary sarcomatoid carcinoma, unknown primary squamous cell carcinoma, unknown undifferentiated neuroendocrine carcinoma, unknown primary undifferentiated small cell carcinoma, uterus carcinosarcoma, uterus endometrial adenocarcinoma, uterus endometrial adenocarcinoma endometrioid, uterus endometrial adenocarcinoma papillary serous, and uterus leiomyosarcoma.
  • 9. The crystalline form of claim 1, wherein the crystalline form is characterized by a DTA thermogram characterized by an endothermal event at about 317° C.
  • 10. The crystalline form of claim 1, wherein the crystalline form is characterized by a DSC thermogram characterized by an endothermal event at about 317° C.
  • 11. The crystalline form of claim 1, wherein the crystalline form is characterized by a hygroscopicity characterized by a mass uptake of about 0.7% at 90% relative humidity as determined by dynamic vapor sorption analysis.
Parent Case Info

This application is a National Stage application under 35 U.S.C. § 371 of International Application No. PCT/US2017/058518, filed Oct. 26, 2017, which claims priority to U.S. Provisional Application No. 62/524,801, filed Jun. 26, 2017 and International Application No. PCT/US2016/058951, filed Oct. 26, 2016, which are herein incorporated by reference in their entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2017/058518 10/26/2017 WO 00
Publishing Document Publishing Date Country Kind
WO2018/081417 5/3/2018 WO A
US Referenced Citations (191)
Number Name Date Kind
5374659 Gowan Dec 1994 A
5430021 Rudnic et al. Jul 1995 A
5760068 Talley et al. Jun 1998 A
5844092 Presta et al. Dec 1998 A
5877016 Presta et al. Mar 1999 A
5910574 Presta et al. Jun 1999 A
6025166 Presta et al. Feb 2000 A
6027927 Presta et al. Feb 2000 A
6153189 Presta et al. Nov 2000 A
6218375 Raghavan Apr 2001 B1
6534085 Zeligs Mar 2003 B1
7384632 Devaux et al. Jun 2008 B2
7491794 Blatt et al. Feb 2009 B2
7514446 Davis-Ward et al. Apr 2009 B2
7550470 Fraley Jun 2009 B2
7612067 Barbosa et al. Nov 2009 B2
7615383 Devaux et al. Nov 2009 B2
7863289 Spevak et al. Jan 2011 B2
8026247 Bold et al. Sep 2011 B2
8106167 Wild, Jr. et al. Jan 2012 B2
8114989 Wang et al. Feb 2012 B2
8119592 Beigelman et al. Feb 2012 B2
8148107 Macdonald et al. Apr 2012 B2
8299021 Blatt et al. Oct 2012 B2
8299057 Lombardi Borgia et al. Oct 2012 B2
8338417 Li et al. Dec 2012 B2
8399442 Berdini et al. Mar 2013 B2
8450322 Andrews et al. May 2013 B2
8501756 Artman, III et al. Aug 2013 B2
8513263 Haas et al. Aug 2013 B2
8552002 Ding et al. Oct 2013 B2
8568998 Mani Oct 2013 B2
8637256 Ernst Jan 2014 B2
8637516 Fan et al. Jan 2014 B2
8642035 Luehrsen Feb 2014 B2
8673347 Traversa et al. Mar 2014 B2
8691221 Pavone et al. Apr 2014 B2
8791123 Allen et al. Jul 2014 B2
8815901 Furet et al. Aug 2014 B2
8865698 Haas et al. Oct 2014 B2
8911734 Latham et al. Dec 2014 B2
8912194 Ciomei Dec 2014 B2
8912204 Ibrahim et al. Dec 2014 B2
8933084 Andrews Jan 2015 B2
8937071 Eidam et al. Jan 2015 B2
8946226 Ciomei et al. Feb 2015 B2
9006256 Matsui Apr 2015 B2
9035063 Eidam et al. May 2015 B2
9102671 Molteni et al. Aug 2015 B2
9127013 Haas et al. Sep 2015 B2
9187489 Takeda et al. Nov 2015 B2
9227975 Andrews et al. Jan 2016 B2
9242977 Takeuchi et al. Jan 2016 B2
9260437 Ibrahim et al. Feb 2016 B2
9273051 Chen et al. Mar 2016 B2
9346788 Wu et al. May 2016 B2
9447104 Haas et al. Sep 2016 B2
9447135 Rohr et al. Sep 2016 B2
9469876 Kuslich Oct 2016 B2
9493476 Andrews et al. Nov 2016 B2
9511050 Toretsky et al. Dec 2016 B2
9670207 Sasmal et al. Jun 2017 B2
9676783 Haas et al. Jun 2017 B2
9682979 Allen et al. Jun 2017 B2
9701681 Kim et al. Jun 2017 B2
9718822 Andrews et al. Aug 2017 B2
9750744 Andrews et al. Sep 2017 B2
9782400 Yao et al. Oct 2017 B2
9782414 Arrigo et al. Oct 2017 B2
9782415 Allen et al. Oct 2017 B2
9795611 Andrews et al. Oct 2017 B2
9796723 Andrews et al. Oct 2017 B2
9796724 Allen et al. Oct 2017 B2
9840519 Andrews et al. Dec 2017 B2
9902741 Andrews et al. Feb 2018 B2
10005783 Haas et al. Jun 2018 B2
10011604 Andrews et al. Jul 2018 B2
10045991 Cox et al. Aug 2018 B2
10047097 Haas et al. Aug 2018 B2
10137127 Reynolds et al. Nov 2018 B2
10172861 Arrigo et al. Jan 2019 B2
10251889 Shelley et al. Apr 2019 B2
20030118654 Santos Jun 2003 A1
20030229047 Joshi-Hangal et al. Dec 2003 A1
20050209195 Menta et al. Sep 2005 A1
20050239840 Arbuthnot et al. Oct 2005 A1
20060089362 Seno et al. Apr 2006 A1
20060094699 Kampen et al. May 2006 A1
20060128725 Guzi Jun 2006 A1
20060211696 Hibi et al. Sep 2006 A1
20070025540 Travis Feb 2007 A1
20070042941 Hirashima et al. Feb 2007 A1
20070082900 Guzi et al. Apr 2007 A1
20070082902 Pamchet al. Apr 2007 A1
20070184117 Gregory et al. Aug 2007 A1
20070225270 Guzi et al. Sep 2007 A1
20070281951 Guzi et al. Dec 2007 A1
20080226747 Bearss et al. Sep 2008 A1
20090041717 Macdonald et al. Feb 2009 A1
20090099167 Bold et al. Apr 2009 A1
20090130229 Lanzi et al. May 2009 A1
20090227556 Obaishi Sep 2009 A1
20090275622 Linga et al. Nov 2009 A1
20100029633 Allen et al. Feb 2010 A1
20100152219 Block et al. Jun 2010 A1
20100297115 Blaustein Nov 2010 A1
20100324065 Ibrahim et al. Dec 2010 A1
20110053934 Angell et al. Mar 2011 A1
20110166122 Andrews et al. Jul 2011 A1
20110195948 Haas et al. Aug 2011 A1
20110268725 Shelton Nov 2011 A1
20110301157 Bold et al. Dec 2011 A1
20120108568 Allen et al. May 2012 A1
20130029925 Vandier et al. Jan 2013 A1
20130203776 Andrews et al. Aug 2013 A1
20130217662 Andrews et al. Aug 2013 A1
20140121239 Aftab May 2014 A1
20140194403 Haas et al. Jul 2014 A1
20140227287 Kamohara et al. Aug 2014 A1
20140243332 Davare Aug 2014 A1
20140315199 Rhodes et al. Oct 2014 A1
20140336236 Cronin et al. Nov 2014 A1
20150005499 Haas et al. Jan 2015 A1
20150018336 Chen et al. Jan 2015 A1
20150031667 Allen et al. Jan 2015 A1
20150051222 Barbugian et al. Feb 2015 A1
20150073036 Hawryluk et al. Mar 2015 A1
20150166564 Allen et al. Jun 2015 A1
20150218132 Wu Aug 2015 A1
20150218652 Doebele et al. Aug 2015 A1
20150283132 Lim et al. Oct 2015 A1
20150306086 Wilcoxen Oct 2015 A1
20150315657 Rhodes et al. Nov 2015 A1
20150336970 Andrews et al. Nov 2015 A1
20160000783 Takeuchi et al. Jan 2016 A1
20160009785 Lipson et al. Jan 2016 A1
20160010068 Bastian Jan 2016 A1
20160032396 Diehn Feb 2016 A1
20160032402 Jagani et al. Feb 2016 A1
20160032404 Schweighofer et al. Feb 2016 A1
20160108123 Freeman et al. Apr 2016 A1
20160108380 Iavarone et al. Apr 2016 A1
20160137654 Arrigo et al. May 2016 A1
20160145237 Hu et al. May 2016 A1
20160228441 Haas et al. Aug 2016 A1
20160251357 Andrews et al. Sep 2016 A1
20160263086 Toretsky Sep 2016 A1
20160272725 Stransky et al. Sep 2016 A1
20160305943 Takeuchi et al. Oct 2016 A1
20160367547 Yao et al. Dec 2016 A1
20170107232 Andrews et al. Apr 2017 A1
20170112842 Andrews et al. Apr 2017 A1
20170112849 Andrews et al. Apr 2017 A1
20170114059 Andrews et al. Apr 2017 A1
20170114067 Haas et al. Apr 2017 A1
20170114068 Andrews et al. Apr 2017 A1
20170114069 Allen et al. Apr 2017 A1
20170114415 Doebele et al. Apr 2017 A1
20170119770 Allen et al. May 2017 A1
20170165267 Arrigo et al. Jun 2017 A1
20170224662 Motheram et al. Aug 2017 A1
20170260589 Nanda et al. Sep 2017 A1
20170281632 Cox et al. Oct 2017 A1
20170283435 Andrews et al. Oct 2017 A1
20170296544 Reynolds et al. Oct 2017 A1
20180021342 Arrigo et al. Jan 2018 A1
20180030548 Nanda et al. Feb 2018 A1
20180030549 Nanda et al. Feb 2018 A1
20180119228 Nanda et al. May 2018 A1
20180127427 Haas et al. May 2018 A1
20180133222 Cox et al. May 2018 A1
20180140604 Tuch et al. May 2018 A1
20180142306 Nanda et al. May 2018 A1
20180207162 Arrigo et al. Jul 2018 A1
20180263984 Allen et al. Sep 2018 A1
20190031684 Andrews Jan 2019 A1
20190076436 Andrews Mar 2019 A1
20190076437 Andrews Mar 2019 A1
20190151322 Andrews May 2019 A1
20190169193 Andrews et al. Jun 2019 A1
20190211017 Haas et al. Jul 2019 A1
20190216814 Reynolds et al. Jul 2019 A1
20190218222 Reynolds et al. Jul 2019 A1
20190247398 Zhao Aug 2019 A1
20190365763 Allen et al. Dec 2019 A1
20200000807 Arrigo et al. Jan 2020 A1
20200237765 Cox et al. Jul 2020 A1
20200291026 Andrews et al. Sep 2020 A1
20200338079 Reynolds et al. Oct 2020 A1
20210002287 Haas et al. Jan 2021 A1
20210023086 Bilenker et al. Jan 2021 A1
Foreign Referenced Citations (214)
Number Date Country
2015101722 May 2016 AU
1938311 Mar 2007 CN
101119996 Feb 2008 CN
101208093 Jun 2008 CN
009517 Feb 2008 EA
0810217 Dec 1997 EP
1873157 Jan 2008 EP
1948633 Aug 2011 EP
2986736 Feb 2016 EP
2558490 Dec 2016 EP
3266795 Oct 2018 EP
H10120683 May 1998 JP
2004-087707 Mar 2004 JP
2004-277337 Oct 2004 JP
2005-008581 Jan 2005 JP
2006-518364 Aug 2006 JP
2007-504276 Mar 2007 JP
2007-514712 Jun 2007 JP
2008-523034 Jul 2008 JP
2008-285464 Nov 2008 JP
2009-502734 Jan 2009 JP
2009-511487 Mar 2009 JP
2009-221199 Oct 2009 JP
2009-541242 Nov 2009 JP
2010-508315 Mar 2010 JP
2011-520887 Jul 2011 JP
2012-506446 Mar 2012 JP
2012-507569 Mar 2012 JP
2013-226108 Nov 2013 JP
2014-082984 May 2014 JP
WO 199849167 Nov 1998 WO
0059929 Oct 2000 WO
0241920 May 2002 WO
WO 2003080064 Oct 2003 WO
WO 2004022561 Mar 2004 WO
WO 2004052286 Jun 2004 WO
WO 2004052315 Jun 2004 WO
WO 2004074290 Sep 2004 WO
WO 2004082458 Sep 2004 WO
WO 2004087707 Oct 2004 WO
WO 2004089415 Oct 2004 WO
WO 2004089471 Oct 2004 WO
WO 2005044835 May 2005 WO
WO 2005049033 Jun 2005 WO
WO 2005051366 Jun 2005 WO
WO 2005077954 Aug 2005 WO
WO 2006052913 May 2006 WO
2006061417 Jun 2006 WO
WO 2006087538 Aug 2006 WO
WO 2006115452 Nov 2006 WO
WO 2006123113 Nov 2006 WO
WO 2006131051 Dec 2006 WO
WO 2006131952 Dec 2006 WO
WO 2007002325 Jan 2007 WO
WO 2007002433 Jan 2007 WO
WO 2007013673 Feb 2007 WO
WO 2007015824 Feb 2007 WO
WO 2007022999 Mar 2007 WO
WO 2007024680 Mar 2007 WO
WO 2007025090 Mar 2007 WO
WO 2007025540 Mar 2007 WO
WO 2007038314 Apr 2007 WO
WO 2007044410 Apr 2007 WO
WO 2007044449 Apr 2007 WO
WO 2007044893 Apr 2007 WO
WO 2007048066 Apr 2007 WO
WO 2007057399 May 2007 WO
2007070504 Jun 2007 WO
WO 2007062805 Jun 2007 WO
WO 2007084815 Jul 2007 WO
WO 2007087245 Aug 2007 WO
WO 2007102679 Sep 2007 WO
WO 2007103308 Sep 2007 WO
WO 2007110344 Oct 2007 WO
WO 2007113000 Oct 2007 WO
WO 2007129161 Nov 2007 WO
WO 2007136103 Nov 2007 WO
2007147647 Dec 2007 WO
2008016192 Feb 2008 WO
WO 2008016131 Feb 2008 WO
WO 2008021924 Feb 2008 WO
WO 2008030579 Mar 2008 WO
WO 2008031551 Mar 2008 WO
WO 2008037477 Apr 2008 WO
WO 2008052734 May 2008 WO
WO 2008058126 May 2008 WO
WO 2008079903 Jul 2008 WO
WO 2008079906 Jul 2008 WO
WO 2008079909 Jul 2008 WO
WO 2008080001 Jul 2008 WO
WO 2008080015 Jul 2008 WO
WO 2008115974 Sep 2008 WO
WO 2008116898 Oct 2008 WO
WO 2008155421 Dec 2008 WO
WO 2009007748 Jan 2009 WO
WO 2009012283 Jan 2009 WO
WO 2009013126 Jan 2009 WO
WO 2009014637 Jan 2009 WO
WO 2009017838 Feb 2009 WO
WO 2009052145 Apr 2009 WO
WO 2009053442 Apr 2009 WO
WO 2009060197 May 2009 WO
WO 2009070567 Jun 2009 WO
WO 2009071480 Jun 2009 WO
WO 2009092049 Jul 2009 WO
WO 2009118411 Oct 2009 WO
WO 2009140128 Nov 2009 WO
WO 2009143018 Nov 2009 WO
WO 2009143024 Nov 2009 WO
WO 2009152083 Dec 2009 WO
WO 2010012733 Feb 2010 WO
WO 2010031816 Mar 2010 WO
WO 2010033941 Apr 2010 WO
WO 2010048314 Apr 2010 WO
WO 2010051549 May 2010 WO
WO 2010058006 May 2010 WO
WO 2010093928 Aug 2010 WO
WO 2010111527 Sep 2010 WO
WO 2010145998 Dec 2010 WO
WO 2011006074 Jan 2011 WO
WO 2011092120 Aug 2011 WO
WO 2011130340 Oct 2011 WO
WO 2011133637 Oct 2011 WO
WO 2011146336 Nov 2011 WO
2011156588 Dec 2011 WO
WO 2012024650 Feb 2012 WO
WO 2012034091 Mar 2012 WO
WO 2012034095 Mar 2012 WO
WO 2012053606 Apr 2012 WO
WO 2012101029 Aug 2012 WO
WO 2012101032 Aug 2012 WO
WO 2012109075 Aug 2012 WO
WO 2012113774 Aug 2012 WO
WO 2012116217 Aug 2012 WO
WO 2012139930 Oct 2012 WO
WO 2012143248 Oct 2012 WO
WO 2012152763 Nov 2012 WO
WO 2012158413 Nov 2012 WO
WO 2013014039 Jan 2013 WO
WO 2013050446 Apr 2013 WO
WO 2013050448 Apr 2013 WO
WO 2013059740 Apr 2013 WO
WO 2013074518 May 2013 WO
WO 2013102059 Jul 2013 WO
WO 2013174876 Nov 2013 WO
WO 2013183578 Dec 2013 WO
2014005021 Jan 2014 WO
2014016433 Jan 2014 WO
2014018567 Jan 2014 WO
WO 2014011900 Jan 2014 WO
WO 2014019908 Feb 2014 WO
WO 2014036387 Mar 2014 WO
WO 2014047572 Mar 2014 WO
WO 2014071358 May 2014 WO
WO 2014072220 May 2014 WO
WO 2014078322 May 2014 WO
WO 2014078323 May 2014 WO
WO 2014078325 May 2014 WO
WO 2014078328 May 2014 WO
WO 2014078331 May 2014 WO
WO 2014078372 May 2014 WO
WO 2014078378 May 2014 WO
WO 2014078408 May 2014 WO
WO 2014078417 May 2014 WO
WO 2014078454 May 2014 WO
WO 2014083567 Jun 2014 WO
WO 2014130975 Aug 2014 WO
WO 2014134096 Sep 2014 WO
WO 2014152777 Sep 2014 WO
WO 2014160521 Oct 2014 WO
WO 2014184069 Nov 2014 WO
WO 2014194127 Dec 2014 WO
WO 2015017528 Feb 2015 WO
WO 2015017533 Feb 2015 WO
WO 2015039006 Mar 2015 WO
WO 2015057873 Apr 2015 WO
WO 2015058129 Apr 2015 WO
WO 2015061572 Apr 2015 WO
WO 2015064621 May 2015 WO
WO 2015108992 Jul 2015 WO
WO 2015112806 Jul 2015 WO
WO 2015124697 Aug 2015 WO
WO 2015161274 Oct 2015 WO
WO 2015161277 Oct 2015 WO
WO 2015175788 Nov 2015 WO
WO 2015183836 Dec 2015 WO
WO 2015183837 Dec 2015 WO
WO 2015184443 Dec 2015 WO
WO 2015191666 Dec 2015 WO
WO 2015191667 Dec 2015 WO
WO 2016011141 Jan 2016 WO
WO 2016011144 Jan 2016 WO
WO 2016011147 Jan 2016 WO
WO 2016022569 Feb 2016 WO
WO 2016027754 Feb 2016 WO
WO 2016075224 May 2016 WO
WO 2016077841 May 2016 WO
WO 2016081450 May 2016 WO
WO 2016097869 Jun 2016 WO
WO 2016187508 Nov 2016 WO
WO 2016196141 Dec 2016 WO
WO 2016196671 Dec 2016 WO
WO 2017001491 Jan 2017 WO
WO 2017004342 Jan 2017 WO
WO 2017075107 May 2017 WO
WO 2017155018 Sep 2017 WO
2017176744 Oct 2017 WO
2017176751 Oct 2017 WO
WO 2017184597 Oct 2017 WO
WO 2017201156 Nov 2017 WO
WO 2017201241 Nov 2017 WO
WO 2018081417 May 2018 WO
WO 2018170381 Sep 2018 WO
2019191659 Oct 2019 WO
Non-Patent Literature Citations (611)
Entry
U.S. Appl. No. 16/044,653, filed Jul. 25, 2018, Allowed.
U.S. Appl. No. 14/943,014, filed Nov. 16, 2015, Published.
U.S. Appl. No. 16/366,368, filed Mar. 27, 2019, Pending.
U.S. Appl. No. 15/622,388, filed Apr. 4, 2017, Issued.
U.S. Appl. No. 15/861,017, filed Jan. 3, 2018, Allowed.
U.S. Appl. No. 16/199,739, filed Nov. 26, 2018, Published.
U.S. Appl. No. 15/900,019, filed Feb. 20, 2018, Pending.
U.S. Appl. No. 16/025,281, filed Jul. 2, 2018, Published.
U.S. Appl. No. 15/335,378, filed Oct. 26, 2016, Published.
U.S. Appl. No. 15/785,218, filed Oct. 16, 2017, Published.
U.S. Appl. No. 15/860,789, filed Jan. 3, 2018, Published.
U.S. Appl. No. 16/199,818, filed Nov. 26, 2018, Pending.
U.S. Appl. No. 16/199,875, filed Nov. 26, 2018, Published.
U.S. Appl. No. 16/377,514, filed Apr. 8, 2019, Pending.
Bayer. “A Study to Test the Effect of the Drug Larotrectinib in Adults and Children With NTRK-fusion Positive Solid Tumors (NAVIGATE).” https://clinicaltrials.gov/ct2/show/NCT02576431. First Posted Oct. 15, 2015. Updated Aug. 20, 2020. 19 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2018/039502, dated Jan. 9, 2020, 8 pages.
U.S. Appl. No. 16/366,368, filed Mar. 27, 2019, Published.
U.S. Appl. No. 15/922,388, filed Apr. 4, 2017, Issued.
U.S. Appl. No. 16/739,845, filed Jan. 10, 2020, Pending.
U.S. Appl. No. 16/199,739, filed Nov. 26, 2018, Allowed.
U.S. Appl. No. 15/900,019, filed Feb. 20, 2018, Allowed.
U.S. Appl. No. 16/025,281, filed Jul. 2, 2018, Allowed.
U.S. Appl. No. 15/785,218, filed Oct. 16, 2017, Allowed.
U.S. Appl. No. 15/785,28, filed Oct. 16, 2017, Issued.
U.S. Appl. No. 16/377,514, filed Apr. 8, 2019, Published.
U.S. Appl. No. 16/345,571, filed Oct. 26, 2017, Pending.
Adriaenssens et al., “Nerve Growth Factor Is a Potential Therapeutic Target in Breast Cancer,” Cancer Res., 2008, 68(2):346-351.
Agaram et al., “Recurrent NTRK1 gene fusions define a novel subset oflocally aggressive lipofibromatosis-like neural tumors,” Am. J. Surg. Pathol, Oct. 2016, 40(10): 1407-1416.
Agaram, et al., “Abstract 33: NTRK1 Associated Gene Fusions in Pediatric Fibroblastic Myofibroglastic Neoplasms: A Molecular Study of 58 Cases,” 105th Annual Meeting of the United States and Canadian Academy of Pathology, 2016, 12A.
Aisner et al., “ROSI and ALK fusions in colorectal cancer, with evidence of intra-tumoral heterogeneity for molecular drivers.”, Mal. Cancer Res., 12(1): 111-8, 2014.
Alassiri et al., “ETV6-NTRK3 Is Expressed in a Subset of ALK-Negative Inflammatory Myofibroblastic Tumors,” Am J Surg Pathol., Aug. 2016, 40(8): 1051-1061.
Albaugh et al., “Discovery of GNF-5837, a Selective TRK Inhibitor with Efficacy in Rodent Cancer Tumor Models,” ACS Medicinal Chemistry Letters, 2012, 3(2):140-145.
Ali et al., “Comprehensive Genomic Profiling Identifies a Subset of Crizotinib-Responsive ALK-Rearranged Non-Small Cell Lung Cancer Not Detected by Fluorescence In Situ Hybridization.”, Oncologist, 21(6): 762-70, 2016.
Alvarez-Breckenridge et al., “Clinical and radiographic response following targeting ofBCAN-NTRK1 fusion in glioneuronal tumor,” NPJ Precision Oncology, Mar. 2017, 5 pages.
Amatu et al., “NTRK gene fusions as novel targets of cancer therapy across multiple tumour types”, ESMD Open, Jan. 9, 2016.
American Association for Cancer Research, “TRK Inhibitor Shows Early Promise,” Cancer Discovery, 6(1), Jan 1, 2016, XP009194480.
American Cancer Society,“Sarcoma: Adult Soft Tissue Cancer,” Jun. 2014, retrieved on Apr. 27, 2015, http://www. cancer.org/cancer/sarcoma-adultsofttissuecancer/detailedguide/sarcoma-adult-soft-tissue-cancer-key-statistics, 45 pages.
Andreason et al., “ETV6 Gene Rearrangements Characterize a Morphologically Distinct Subset of Sinonasal Low-grade Non-intestinal-type Adenocarcinoma,” Am. J. Surg. Pathol, Nov. 2017, 41(11):1552-1560.
Arce et al., “Secretory carcinoma of the breast containing the ETV6-NTRK3 fusion gene in a male: case report and review of the literature,” World J. Sug. Oncol, Jun. 2005, 3:35.
Ardini et al., “The TPM3-NTRK1 rearrangement is a recurring event in colorectal carcinoma and is associated with tumor sensitivity to TRKA kinase inhibition,” Mol. Oncol. 8(8): 1495-1507, 2014.
Asaumi et al., “Expression of neurotrophins and their receptors (TRK) during fracture healing,” Bone, 2000, 26(6):625-633.
Awad et al., “Acquired resistance to crizotinib from a mutation in CD74-ROS1. ”, N Engl. J Med, 368(25): 2395-401, 2013.
Bailey, Justin J., et al. “Tropomyosin receptor kinase inhibitors: an updated patent review for 2010-2016—Part II.” Expert opinion on therapeutic patents 27.7 (2017): 831-849.
Bardelli et al., “Mutational Analysis of the Tyrosine Kinome in Colorectal Cancers,” Science, May 2003, 300(5621):949.
Bartenstein et al., “Lipofibromatosis-like neural tumor: Case report of a unique infantile presentation,” JAAD Case Reports, 4(2):185-188, 2018.
Baughn et al., “Abstract 5115: Whole-Genome Mate Pair Sequencing Reflex Test to Characterize Chromosome Rearrangements in Hematologic Neoplasia,” Blood, 2017, 130: 5115.
Bavle et al., “Abstract GENE-04: Pediatric Malignant Epithelioid Glioneuronal Tumor: Pathological, Clinical, and Molecular Characterization of a Rare and Deadly Malignancy,” Neuro-Oncology, Jun. 2017, iv18-iv19.
Behrens et al., “Go 6976 is a potent inhibitor of neurotrophin-receptor intrinsic tyrosine kinase,” J Neurochem., Mar. 1999, 72(3):919-924.
Beimfohr et al., “NTRK.1 re-arrangement in papillary thyroid carcinomas of children after the Chernobyl reactor accident,” Int. J Cancer, Mar. 15, 1999;80(6):842-847.
Bender et al., Abstract H-024: Multiple Novel Fusion Genes with the RTK-RAS-PBK Signalling Axis Highlight its Central Role in the Turmorigenesis of Pediatric Gioblastoma, Neuro-oncology, Jun. 2014, 145.
Bensinger et al., “Transplantation of allogeneic peripheral blood stem cells mobilized by recombinant human granulocyte colony stimulating factor,” Stem Cells, Jan. 1996;14(1):90-105.
Bensinger et al., “Transplantation of allogeneic peripheral blood stem cells mobilized by recombinant human granulocyte colony-stimulating factor [see comments].,” Blood, Mar. 15, 1995;85(6):1655-8.
Bertrand et al., “The crystal structures of TrkA and TrkB suggest key regions for achieving selective inhibition,” Journal of molecular biology, Oct. 26, 2012;423(3):439-53.
Birch et al., “Chromosome 3 anomalies investigated by genome wide SNP analysis of benign, low malignant potential and low grade ovarian serous tumours.”, PLoS One, 6(12): e28250, 2011.
Bonanno et al., Journal of Thoracic Oncology, vol. 11, No. 4, Supp. Suppl. 1, pp. S67. Abstract No. 28P; 6th European Lung Cancer Conference, ELCC 2016, Geneva, Switzerland.
Bongarzone et al., “Age-related activation of the tyrosine kinase receptor protooncogenes RET and NTRK.1 in papillary thyroid carcinoma,” J Clin. Endocrinol. Metab., May 1996, 81(5):2006-2009.
Bouhana et al., “Abstract #1798: Identification of Pan-Trk Inhibitors for the Treatment of Trk-Driven Cancers,” Poster, Presented at Proceedings of the AACR 103rd Annual Meeting, Apr. 15, 2012.
Bourgeois et al., “Molecular Detection of the ETV6-NTRK3 Gene Fusion Differentiates Congenital Fibrosarcoma From Other Childhood Spindle Cell Tumors,” Am. J Surg. Pathol., Jul. 2000, 24(7):937-946.
Braga, Dario, et al. “Crystal polymorphism and multiple crystal forms.” Struct Bond (2009) 132:25-50. Springer-Verlag Berlin Heidelberg.
Branford, S., et al. “High frequency of point mutations clustered within the adenosine triphosphate-binding region of BCR/ABL in patients with chronic myeloid leukemia or Ph-positive acute lymphoblastic leukemia who develop imatinib (STI571) resistance,” Blood, May 2002, 99, 3472-3475.
Brastianos et al., “Abstract OS06.4: Identification of Novel NTRK Fusion in Glioneuronal Tumors and Radiographic Response Following Therapy with an NTRK Inhibitor,” Neuro-Oncology, May 2017, iii1 1, 1 page, Meeting Info: 5th Quadrennial Meeting of the World Federation of Neuro-Oncology Societies, WFNOS. Zurich, Switzerland, 2017.
Brenca et al., “Transcriptome sequencing identifies ETV6-NTRK3 as a gene fusion involved in GISTt,” J. Pathol. 238(4):543-549, 2016.
Brinner et al., “A rapid and general method for asymmetric synthesis of 2-substituted pyrrolidines using ter-butanesulfinamide,” Organic & Biomolecular Chemistry, Jan. 2005, 3(11): 2019.
Brodeur, “Neuroblastoma: biological insights into a clinical enigma,” Nat. Rev. Cancer, 2003, 3:203-216.
Bruse et al., “Improvements to Bead Based Oligonucleotide Ligation SNP Genotyping Assays,” Biotechniques, Nov. 2008, 45:559-571.
Brzezianska et al., “Rearrangements of NTRK.1 oncogene in papillary thyroid carcinoma,” Neuroendocrinology Letters, 2007, 28(3):221-229.
Burris et al., “Pharmacokinetics (PK) of LOXO-101 During the First-in-Human Phase I Study in Patients with Advanced Solid Tumors,” Interim Update AACR Annual Meeting, Mar. 2015, Philadelphia, PA., 1 page.
Butti et al., “A sequence analysis of the genomic regions involved in the rearrangements between TPM3 and NTRK1 genes producing TRK oncogenes in papillary thyroid carcinomas,” Genomics. 28(1):15-24, 1995.
Caira, “Crystalline Polymorphism of Organic Compounds,” Topics in Current Chemistry, Feb. 1999, 198: 163-208.
Cajaiba et al., “Expanding the spectrum of ALK-rearranged renal cell carcinomas in children: Identification of a novel HOOK1-ALK fusion transcript.”, Genes Chromosomes Cancer, 55(10): 814-7, 2016.
Calabresi and Chabner, Goodman & Gilnnan's The Pharmacological Basis of Therapeutics, 10th ed., 2001, ne: 1388, para 2, lines 4-5.
Calero et al., “Sunitinib suppress neuroblastoma growth through degradation of MYCN and inhibition of angiogenesis,” PLoS One, Apr. 23, 2014;9(4):e95628. doi: 10.1371/iournal.pone.0095628. eCollection 2014.
Camidge, D. Ross, William Pao, and Lecia V. Sequist. “Acquired resistance to TKIs in solid tumours: learning from lung cancer.” Nature reviews Clinical oncology 11.8 (2014): 473.
Camoratto et al., “CEP-751 inhibits TRK receptor tyrosine kinase activity in vitro exhibits anti-tumor activity,” Int. J Cancer, Aug. 1997, 72:673-679.
Campos et al., “Enantioselective, palladium-catalyzed alpha-arylation ofN-Boc-pyrrolidine,” J. Am. Chem Soc., 2006, 128:3538-3539.
Cancer.gov [online]. “National Cancer Institute: Oral TRK Inhibitor LOXO-101 (Larotrectinib) for Treatment of Advanced Pediatric Solid or Primary Central Nervous System Tumors,” ClinicalTrials.gov Identifier: NCT02637687, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<https://www.cancer.gov/about-cancer/treatment/clinical-trials/search/view?cdrid=781 000>, 5 pages.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.1 p. V321M / c.961G>A,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<cancer.sanger.ac.uk/cosmic/mutation/ overview?id=1259646>, 1 page.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.1 p.D679N / c.2035G>A,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<cancer. sanger.ac.uk/cosmic/mutation/overview?id=897427>, 1 page.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.3 p.D537Y / c.1609G>T,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<cancer.sanger.ac.uk/cosmic/mutation/ overview?id=966118>, 1 page.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.3 p.D609V / c.1826A>T,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL:<cancer.sanger.ac.uk/cosmic/mutation/ overview?id=124878>, 1 page.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.3 p.G608S / c.1822G>A,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<cancer.sanger.ac.uk/cosmic/mutation/ overview?id=88799>, 1 page.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.3 p.L282M / c.844C>A,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<cancer.sanger.ac.uk/cosmic/mutation/ overview?id=401588>, 1 page.
Cancer.sanger.ac.uk [online]. “COSMIC, Catalog of Somatic Mutations in Cancer: Cosmic» Mutation» Overview» NTRK.3 p.V539M I c.1615G>A,” Catalog of Somatic Mutations in Cancer (COSMIC) database, [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<cancer. sanger.ac.uk/cosmic/mutation/ overview?id=1708512>, 1 page.
Capparelli et al., “Stromal neuregulin-1 modulates the response to MEK inhibitors in WT BRAF/WT NRAS (WT/WT) melanomas”, Pigment Cell Melanoma Res. vol. 30, No. 5, pp. e61, 2017.
Caria et al., “Cytogenetic and molecular events in adenoma and well-differentiated thyroid follicular-cell neoplasia,” Cancer Genet. Cytogenet., 2010, 203:21-29.
Carpinelli et al., “PHA-739358, a potent inhibitor of Aurora kinases with a selective target inhibition profile relevant to cancer,” Mol Cancer Ther, Dec. 2007;6(12 Pt 1):3158-3168.
Carvalho et al., Neuro-Oncology 1 7:iii1-iii40, 2015, Abstract No. HG-09, 1 page.
Catalog of Somatic Mutations in Cancer (COSMIC) database, cancer.sanger.ac.uk/cosmic/mutation/overview?id=1517968, downloaded on May 31, 2016, 2 pages.
Catalog of Somatic Mutations in Cancer (COSMIC) database, cancer.sanger.ac.uk/cosmic/mutation/overview?id=1636266, downloaded on May 31, 2016, 2 pages.
Catalog of Somatic Mutations in Cancer (COSMIC) database, cancer.sanger.ac.uk/cosmic/mutation/overview?id=1688778, downloaded on May 31, 2016, 2 pages.
Catalog of Somatic Mutations in Cancer (COSMIC) database, cancer.sanger.ac.uk/cosmic/mutation/overview?id=3711772, downloaded on May 31, 2016, 2 pages.
Catalog of Somatic Mutations in Cancer (COSMIC) database, cancer.sanger.ac.uk/cosmic/mutation/overview?id=471203, downloaded on May 31, 2016, 2 pages.
Catalog of Somatic Mutations in Cancer (COSMIC) database, cancer.sanger.ac.uk/cosmic/mutation/overview?id=48622, downloaded on May 31, 2016, 2 pages.
Catic et al., “A novel cytogenetic and molecular characterization of renal metanephric adenoma, identification of partner genes involved in translocation t(9;15)(p24;q24),” Cancer Genet. 214-215:9-15, doi: 10.1016/j.cancergen.2017.03.001, 2017.
Catic et al., “Abstract 1537: The frequency of a novel KANK1 and NTRK3translocation and BRAFV600E mutation in patients diagnosed with metanephric adenoma utilizing molecular mechanisms,” 2017 Annual Meeting of the American Society of Clinical Oncology, 2017, 1 page.
Center for Drug Evaluation and Research: www.accessdata.fda.gov/drugsatfda_docs/nds/2018/210861Origls000_211710Origls000ChemR.pdf , 2017, 64 pages.
Chang-Qi et al., “Brain derived neurotrophic factor (BDNF) contributes to the pain hypersensitivity following surgical incision in the rats,” Molecular Pain, 2008, 4:27.
Chaudhuri et al., “Early Detection of Molecular Residual Disease in Localized Lung Cancer by Circulating Tumor DNA Profiling,” Cancer Discov, Dec. 2017, 7(12):1394-1403.
Chen et al.,“40: The landscape of kinase fusions in 445 Chinese NSCLC patients,” Annals of Oncology, Oct. 2017, 28(7): vii16, 1 page.
Cherry et al., “Recent kinase and kinase inhibitor X-ray structures: mechanisms of inhibition and selectivity insights,” Curr Med Chem. Mar. 2004;11(6):663-73.
Chiang et al., “NTRK Fusions Define a Novel Uterine Sarcoma Subtype with Features of Fibrosarcoma,” Am. J. Surg. Pathol. doi: 10.1097IPAS.0000000000001055, 2018.
Chintakuntlawar et al., “High-grade transformation of acinic cell carcinoma: an inadequately treated entity?,” Oral Surg Oral Med Oral Pathol Oral Radiol, May 2016, 121(5):542-549.
Chmielecki et al., “Abstract LB-178: Genomic profiling of 1239 diverse pediatric cancers identifies novel discoveries across tumors”, Cancer Research, vol. 76, No. 14, Supp. Supplement. Abstract No. LB-178. 107th Annual meeting of the American Association for Cancer Research, AACR. New Orleans, LA Apr. 16-20, 2016.
Chmielecki et al., “Genomic Profiling of a Large Set of Diverse Pediatric Cancers Identifies Known and Novel Mutations across Tumor Spectra.”, Cancer Research, 77(2): 509-519, 2017.
Cho et al., “Expression of mRNA for brain-derived neurotrophic factor in the dorsal root ganglion following peripheral inflammation,” Brain Research, 1997, 749:358-362.
Choi et al., “(R)-2-Phenylpyrrolidine Substituted Irnidazopy ridazines. A New Class of Potent and Selective Pan-TRK Inhibitors,” ACS medicinal chemistry letters, Mar. 2015 I 9;6(5):562-7.
Chung et al., “Infantile fibrosarcoma,” Cancer, Aug. 1976, 38(2):729-739.
Church et al., “Abstract ST16: A Novel EML4-NTRK3 Translocation in Infantile Fibrosarcoma and Congenital Mesoblastic Nephroma Requires a New Approach to Conventional Diagnostic Algorithms,” J Molecular Diag, 2015, 816.
Church et al., “Recurrent EML4-NTRK3 fusions in infantile fibrosarcoma and congenital mesoblastic nephroma suggest a revised testing strategy,” Mod. Pathol. 31(3), 463-473, 2018.
Cocce et al., “Identification of ZCCHC8 as fusion partner of ROS1 in a case of congenital glioblastoma multiforme with a t(6;12)(q21;q24.3)”, Genes Chromosomes Cancer, 55(9): 677-87, 2016.
Coebergh et al., “Abstract 490: Identification of oncogenic gene fusions in primary colon cancers,” Cancer Research, Jul. 2017, DOI: 10.1158/1538-7445.AM2017-490, 2 pages.
Comina-Mendez and Turner, “Predicting Relapse with Circulating Tumor DNA Analysis in Lung Cancer,” CancerDiscov, Dec. 2017, 7(12): 1368-1370.
Cook et al., “Somatic chromosomal engineering identifies BCAN-NTRK1 as a potent glioma driver and therapeutic target,” Nat. Comm. 8(15987). DOI 10.1038/ncomms15987, 2017.
Creancier et al., “Chromosomal rearrangements involving the NTRK.1 gene in colorectal carcinoma,” Cancer Lett., Awmst 2015, 365(1):107-111.
Crescenzo et al., “Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma.”, Cancer Cell., 27(4): 516-32, 2015.
Croucher et al., “TrkB inhibition by GNF-4256 slows growth and enhances chemotherapeutic efficacy in neuroblastoma xenografts,” Cancer Chemother Pharmacol. Jan. 2015;75(1):131-41. doi: 10.1007/s00280-014-2627-1. Epub Nov. 14, 2014.
Cruz, “Lung cancer: epidemiology, etiology and prevention,” Clinics in Chest Medicine, 2011, 32(4): 1-61.
Cui et al., “Abstract #MA 07.09: ALK/ROS1/inhibitor TPX-0005 Effectively Overcomes Clinical Resistance Solvent Front Mutations,” Abstracts, Nov. 2017, p. S1829.
Cui et al., “Use of capture-based next-generation sequencing to detect ALK fusion in plasma cell-free DNA of patients with non-small-cell lung cancer”, Oncotarget, 2771-2780, 2016.
Dacie et al., “ALK FISH patterns and the detection of ALK fusions by next generation sequencing in lung adenocarcinoma”, Oncotarget, vol. 7, No. 50, pp. 82943-82952, 2016.
Dang et al., “Expression of nerve growth factor receptors is correlated with progression and prognosis of human pancreatic cancer,” J. Gastroenterology and Hepatology, 2006, 21(5): 850-858.
Das et al., “Synergistic Effects of Crizotinib and Temozolomide in Experimental FIG-ROS1 Fusion-Positive Glioblastoma.”, Cancer Growth Metastasis, 8:51-60, 2015.
Davare et al., “Foretinib is a potent inhibitor of oncogenic ROS1 fusion proteins.”, Proc. Natl. Acad Sci. USA., 110(48): 19519-24, 2013.
Davare et al., “Structural insight into selectivity and resistance profiles of ROSI tyrosine kinase inhibitors.”, Proc. Natl. Acad Sci. USA., 112(39): E5381-90, 2015.
Davidson et al., “Expression and activation of the nerve growth factor receptor TrkA in serous ovarian carcinoma,” Clin. Cancer Res., 2003, 9(6):2248-2259.
Davies and Dobele, “Molecular pathways: ROS1 fusion proteins in cancer.”, Clin. Cancer Res, 19(15): 4040-4045, 2013.
Davies et al., “Identifying and targeting ROS1 gene fusions in non-small cell lung cancer.”, Clin Cancer Res 18: 4570-4579, 2012.
Davies et al., “Resistance to ROSI inhibition mediated by EGFR pathway activation in non-small cell lung cell,” PLoS One, 2013, 8(12):e82236, 13 pages.
Davis et al., “Infantile NTRK-associated Mesenchymal Tumors,” Pediatr. Dev. Pathol. 21(1):68-78, 2018.
De Smith et al., “Clonal and microclonal mutational heterogeneity in high hyperdiploid acute lymphoblastic leukemia”, Oneatarget., 7(45) 72733-72745, 2016.
Deihimi et al., “BRCA2, EGFR, and NTRK mutations in mismatch repair-deficient colorectal cancers with MSH2 or MLH1 mutations,” Oncotarget. Jun. 20;8(25):39945-39962, 2017.
Delafoy et al., “Role of nerve growth factor in the trinitrobenzene sulfonic acid-induced colonic hypersensitivity,” Pain, 2003, 105:489-497.
Demaria et al., “Development of tumor-infiltrating lymphocytes in breast cancer after neoadjuvant paclitaxel chemotherapy,” Clin Cancer Res, Oct. 2001;7(10):3025-30.
Di Mola et al., “Nerve growth factor and Trk high affinity receptor (TrkA) gene expression in inflammatory bowel disease,” Gut, 2000, 46(5):670-678.
Diner et al., “Preparation of 3-substituted-1-isopropyl-1H-pyrazolo[3,4-d]pyrimidin-4-amines as RET kinase inhibitors,” J. Med. Chem., May 2012, 55 (10), 4872-4876.
Dionne et al., “Cell cycle-independent death of prostate adenocarcinoma is induced by the trk tyrosine kinase inhibitor CEP-751 (KT6587),” Clin. Cancer Research, 1998, 4(8):1887-1898.
Doebele et al., “Abstract 8023: NTRK1 gene fusions as a novel oncogene target in lung cancer,” 2013 Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, 2013, 1 page.
Doebele et al., “An oncogenic NTRK fusion in a soft tissue sarcoma patient with response to the tropomysin-related kinase (TRK) inhibitor LOXO-101,” Cancer Discovery, Jul. 2015, 5(10):1049-1057.
Doebele et al., “Phase II Trial of Stereotactic Body Radiation Therapy Combined with Erlotinib for Patients with Limited but Progressive Metastatic Non-Small-Cell Lung Cancer,” J. Clin. Oncol., 2014, 32:9 pages.
Dolle et al., “Nerve growth factor-induced migration of endothelial cells,” J. Pharmacol Exp Ther, 2005, 315(3):1220-1227.
Dolomanov et al., “OLEX2: a complete structure solution, refinement and analysis program,” J Annl. Cryst. 2009, 42, 339-341.
Dou et al., “Increased nerve growth factor and its receptors in atopic dermatitis: an immunohistochemical study,” Archives of Dermatological Research, 2006, 298(1):31-37.
Drexler et al., “Pathobiology of NPM-ALK and variant fusion genes in anaplastic large cell lymphoma and other lymphomas,” Leukemia, Sep. 2000, 14:1533-1559.
Drilon et al., “A Novel Crizotinib-Resistant Solvent-Front Mutation Responsive to Cabozantinib Therapy in a Patient with ROSI-Rearranged Lung Cancer.”, Clin. Cancer Res., 22(10): 2351-8, 2016.
Drilon et al., “A phase 1 study of oral LOXO 292 in adult patients with advanced solid tumors, including RET-fusion non-small cell lung cancer, medullary thyroid cancer and other tumors with increased RET activity,” Annals of oncology Developmental Therapeutics, Sep. 2017, 28(5):138.
Drilon et al., “Abstract CT007: Entrectinib, an oral pan-Trk, ROSI, and ALK inhibitor in TKI-naive patients with advanced solid tumors harboring gene rearrangements: Updated phase I results,” Cancer research, 76(14), AACR 107th Annual Meeting, Apr. 2016, URL <http://cancerres.aacrjournals.org/content/76/14 Supplement/CT007.short>, 5 pages.
Drilon et al., “Entrectinib, an oral pan-Trk, ROSI, and ALK inhibitor in TK1-naive patients with advanced solid tumors harboring gene rearrangements,” Cancer research, vol. 76, No. 14, Supp. Supplement., Abstract No. 15 CT007; Presented at the 107th Annual Meeting of the American Association for Cancer Research, Aacr 2016. New Orleans, LA; Apr. 16-20, 2016, 35 pages.
Drilon et al., “What hides behind the MASC: clinical response and acquired resistance to entrectinib after ETV6-NTRK3 identification in a mammary analogue secretory carcinoma (MASC),” Annals of Oncology., Feb. 15, 2016, 27(5):920-926.
Du et al., “Expression ofNGF family and their receptors in gastric carcinoma: a cDNA microarray study,” World Journal of Gastroenterology, http://www.wjgnet.com/1007-9327/full/v9/i7/1431.htm, Jul. 2003, 9(7):1431-1434.
Duranti et al., “Homologation of Mexiletine alkyl chain and stereoselective blockade of skeletal muscle sodium channels,” Euro. J. Med. Chem., 2000, 35:147-156.
Durham et al. “Diverse and Targetable Kinase Alterations Drive Histiocytic Neoplasms,” Blood. 126(23):481, 2015.
Edgren et al., Cancer Res. 75(15 Supplement): 4793, 2015; Abstract only, 3 pages.
Eguchi et al., “Absence of t(12;15) associated ETV6-NTRK3 fusion transcripts in pediatric acute leukemias,” Med Pediatr. Oncol., Oct. 2001, 37:417.
Eguchi et al., “Fusion of ETV6 to neurotrophin-3 receptor TRKC in acute myeloid leukemia with t(12;15)(p13;q25),” Blood, 1999, 93(4):1355-1363.
Eisenhauer et al., “New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1),” Eur J Cancer, Jan. 2009, 45(2):228-247.
Ellison et al., “Abstract 013: Genetic alterations in uncommon low-grade neural tumors—BRAF, FGFR1, and MYB/MYBL1 mutations occur frequently and align with morphology,” Neuropathology and Applied Neurobiology, 2016, 42(S1): 18.
Elvin et al., “319: Genomic profiling of uterine leiomyosarcomas reveal frequent alterations in Akt/mammalian target of rapamycin (mTOR) pathway genes and other actionable genomic abnormalities linked to targeted therapies,” Poster Session—Molecular Targeted Agents I, Nov. 2014, 1 page.
Endometrial Cancer Gene Database, ecgene.bioinfominzhao.org/gene_mutation.cgi?gene=4915, downloaded on May 31, 2016, 13 pages.
Engman et al., “Syngeneic transplant in mantle cell lymphoma: a rare event and review of the literature,” Clin Adv Hematol Oncol. May 2009;7(5):321-3.
Esmo, “TRK Cancer-Causing Mutation Discovered in 1982 Finally Target of Clinical Trials: Matching drugs to long-overlooked oncogene,” European Society of Medical Oncology, Jan. 2015, 2 pages.
Essand et al., “Genetically engineered T cells for the treatment of cancer,” J Intern Med. Feb. 2013;273(2):166-81. doi: 10.1111/joim.12020.
Estrada-Bernal et al., “Abstract#: C65: TRK kinase domain mutations that induce resistance to a pan-TRK inhibitor,” Poster, Presented at Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics, Boston MA, Nov. 5-9, 2015; Mol Cancer Ther, Dec. 2015, 14(12)(Suppl. 2): 1 page.
Estrada-Bernal et al., “Abstract#: LB-118: Identification of TRKA and TRKB kinase domain mutations that induce resistance to a pan-TRK inhibitor,” Poster, Presented at Proceedings of the 107th Annual Meeting of the American Association for Cancer Research, New Orleans LA, Apr. 16-20, 2016; Cancer Res, Jul. 2016, 76(14): 1 page.
Euthus et al., “ETV6-NTRK3—Trk-ing the primary event in human secretory breast cancer,” Cancer Cell, 2002, 2(5):347-348.
Evans et al., “Antitumor activity of CEP-751 (KT-6587) on human neuroblastoma and medulloblastomaxenografts,” Clin. Cancer Res., Nov. 1999, 5(11):3594-3602.
Extended European Search Report in European Application No. 13197815.7, dated Apr. 1, 2014, 5 pages.
Extended European Search Report in European Application No. 16166461.0, dated Sep. 28, 2016, 5 pages.
Extended European Search Report in European Application No. 17163978.4, dated Jul. 17, 2017, 5 pages.
Extended European Search Report in European Application No. 17199899.0, dated Feb. 26, 2018, 7 pages.
Extended European Search Report in European Application No. 18151233.6, dated Jun. 26, 2018, 6 pages.
Extended European Search Report in European Application No. 18208279.2, dated Jun. 27, 2019, 10 pages.
Facchinetti et al., “Crizotinib-Resistant ROSI Mutations Reveal a Predictive Kinase Inhibitor Sensitivity Model for ROS1- and ALK-Rearranged Lung Cancers.”, Clin. Cancer Res., 22(24): 5983-5991, 2016.
Farago et al., “Abstract MINB0.09: Clinical Response to Entrectinib in a Patient with NTRK1-Rearranged Non-small cell Lung Cancer,” J Thoracic Oncol, Sep. 2015, 10(9-S2): S374-S375.
Farago et al., “Durable clinical response to entrectinib in NTRK1-rearranged non-small cell lung cancer,” J. Thorac Oncol. 10(12):1670-1674, 2015.
Farhat et al., “Primary benign and malignant thyroid neoplasms with signet ring cells: cytologic, histologic, and molecular features,” Am. J. Clin. Pathol., 148(3):251-258, 2017.
Fernandez-Cuesta et al., “Abstract 1531: Cross-entity mutation analysis of lung neuroendocrine tumors sheds light into their molecular origin and identifies new therapeutic targets,” AACR Annual Meeting 2014, Apr. 2014, URL <http://cancerres.aacrjournals.org/content/7 4/19 Supplement/1531.short>, 5 pages.
Flannery et al., “Immunomodulation: NK cells activated by interferon-conjugated monoclonal antibody against human osteosarcoma,” Eur J Cancer Clin Oncol. Jun. 1984;20(6):791-8.
Forghieri et al., Abstract P137: Chronic Eosinophilic Leukemia with ETV6-NTRK3 Fusion Transcript in an Elderly Patient Affected with Pancreatic Carcinoma, Haemologica, 2010, 95(s3):S125-S126.
Frattini et al., “The integrated landscape of driver genomic alterations in glioblastoma,” Nature Genet., 2013, 45:1141-1149.
Freund-Michel and Frossard, “The nerve growth factor and its receptors in airway inflammatory diseases,” Pharmacology & Therapeutics, 2008, 117(1):52-76.
Frey et al., “7-Aminopyrazolo[1,5-a]pyrimidines as potent multitargeted receptor tyrosine kinase inhibitors,” J. Med. Chem, Jul. 2008, 51(13):3777-3787.
Fu et al., “The Frequency and Clinical Implication of ROSI and RET Rearrangements in Resected Stage IIIA-N2 Non-Small Cell Lung Cancer Patients.”, PLoS One, 10(4):e0124354, 2015.
Fuse et al., “Mechanisms of Resistance to NTRK Inhibitors and Therapeutic Strategies in NTRK1-Rearranged Cancers,” Mol. Cancer Ther., Oct. 2017; 16(10); 2130-43.
Gainor et al., “Patterns of Metastatic Spread and Mechanisms of Resistance to Crizotinib in ROS1-Positive Non-Small-Cell Lung Cancer”, JCO Precis Oneal. 10.1200/PO. 1 7.00063, 2017.
Gang et al., “The landscape of fusion transcripts in spitzoid melanoma and biologically indeterminate spitzoid tumors by RNA sequencing.”, Mod Pathol., 29(4): 359-69, 2016.
Gao et al., “Driver fusions and their implications in the development and treatment of human cancers,” Cell Rep. 23(1):227-238.e3, 2018.
Gatalica et al., “Abstract A047: Molecular characterization of the malignancies with targetable NTRK gene fusions,” American Association for Cancer Research, Jan. 2018, 2 pages.
Gaudet et al., “Allele-specific PCR in SNP genotyping,” Methods Mol Biol. 2009;578:415-24. doi: 10.1007/978-1-60327-411-126.
Gavrin et al., “Synthesis of Pyrazolo[1,5-[alpha]]pyrimidoinone Regioisomers,” J Org Chem, Feb. 2007, 72(3): 1043-1046.
Geiger et al., “Functional Characterization of Human Cancer-Derived TRKB Mutations,” PLoS ONE, Feb. 17, 2011, 6(2):e16871.
Geiger et al., “The neurotrophic receptor TrkB in anoikis resistance and metastasis: a perspective,” J Cancer Res., Aug. 2005, 65(16):7033-7036.
GenBank Accession No. AAB33109.1, “trkB [Homo sapiens],” Jul. 27, 1995, 1 page.
GenBank Accession No. AAB33111.1 “trkC [Homo sapiens],” Jul. 27, 1995, 1 page.
GenBank Accession No. NM_ 002529, “high affinity nerve growth factor receptor isoform 2 precursor [Homo sapiens],” May 11, 2014, 4 pages.
GenBank Accession No. NM_001007792 “Homo sapiens neurotrophic tyrosine kinase, receptor, type 1 (NTRK1), transcript variant 3, mRNA,” May 10, 2014, 5 pages.
GenBank Accession No. NM_001012338, “Homo sapiens neurotrophic tyrosine kinase, receptor, type 3 (NTRK3), transcript variant 1, mRNA,” May 10, 2014, 6 pages.
GenBank Accession No. NM_006180, “Homo sapiens neurotrophic tyrosine kinase, receptor, type 2 (NTRK2), transcript variant a, mRNA,” May 12, 2014, 9 pages.
GenBank Accession No. NP 001007793, “high affinity nerve growth factor receptor isoform 3 [Homo sapiens],” May 10, 2014, 3 pages.
GenBank Accession No. NP_ 002520 “high affinity nerve growth factor receptor isoform 2 precursor [Homo sapiens],” May 11, 2014, 4 pages.
GenBank Accession No. NP_001007157, “NT-3 growth factor receptor isoform c precursor [Homo sapiens],” May 10, 2014, 3 pages.
GenBank Accession No. NP_001012331.1, “high affinity nerve growth factor receptor isoform 1 precursor [Homo sapiens],” May 10, 2014, 4 pages.
GenBank Accession No. NP_001012338, “NT-3 growth factor receptor isoform a precursor [Homo sapiens],” May 10, 2014, 3 pages.
GenBank Accession No. NP_006171, “BDNF/NT-3 growth factors receptor isoform a precursor [Homo sapiens],” May 12, 2014, 4 pages.
GenBank Accession No. S76473.1, “trkB [human, brain, mRNA, 3194 nt],” Jul. 27, 1995, 2 pages.
GenBank Accession No. S76475.1, “trkC [human, brain, mRNA, 2715 nt],” Jul. 27, 1995, 2 pages.
Genevois et al., “Dependence receptor TrkC is a putative colon cancer tumor suppressor,” Proc. Nat. Acad. Sci. U.S.A. Feb. 19, 2013, 110(8):3017-3022.
Giacomini et al., “Breakpoint Analysis of Transcriptional and Genomic Profiles Uncovers Novel Gene Fusions Spanning Multiple Human Cancer Types”, PLoS Gene.t, 9(4): e1003464, 2013.
Gimm et al., “Mutation analysis ofNTRK.2 and NTRK.3, encoding 2 tyrosine kinase receptors, in sporadic human medullary thyroid carcinoma reveals novel sequence variants,” International Journal of Cancer, Apr. 1, 2001, 92(1):70-74.
Greco et al., “Chromosome I rearrangements involving the genes TPR and NTRK1 produce structurally different thyroid-specific TRK oncogenes,” Genes Chromosomes Cancer. 19(2):112-23, 1997.
Greco et al., “Rearrangements of NTRK1 gene in papillary thyroid carcinoma,” Molecular and Cellular Endocrinology, 2010, 321(1):44-49.
Greco et al., “The DNA rearrangement that generates the TRK-T3 oncogene involves a novel gene on chromosome 3 whose product has a potential coiled-coil domain,” Mol. Cell. Biol. 15(11):6118-6127, 1995.
Greco et al., “TRK-T1 is a novel oncogene formed by the fusion ofTPR and TRK genes in human papillary thyroid carcinomas,” Oncogene. 7(2):237-42, 1992.
Green & Wuts, eds, “Protective Groups in Organic Synthesis,” John Wiley & Sons Inc, May 8, 1999.
Groisberg et al., “Clinical next-generation sequencing in sarcomas”, Journal of Clinical Oncology, vol. 34, Supp. Supplement 15; Abstract No. 11046; 2016 Annual Meeting of the American Society of Clinical Oncology, ASCO 2016, Chicago, IL. Jun. 3-7, 2016.
Gruber-Olipitz et al., “Neurotrophin 3/TrkC-regulated proteins in the human medulloblastoma cell line DAOY,” J. Proteome Research, 2008, 7(5):1932-1944.
Gu et al., “Lung adenocarcinoma harboring concomitant SPTBN1-ALK fusion, c-Met overexpression, and HER-2 amplification with inherent resistance to crizotinib, chemotherapy, and radiotherapy.”, J Hematol Oneal, 9(1): 66, 2016.
Gwak et al., “Attenuation of mechanical hyperalgesia following spinal cord injury by administration of antibodies to nerve growth factor in the rat.” Neurosci. Lett., 2003, 336:117-120.
Hainsworth et al., “Lung Adenocarcinoma with Anaplastic Lymphoma Kinase (ALK) Rearrangement Presenting as Carcinoma of Unknown Primary Site: Recognition and Treatment Implications.”, Drugs Real World Outcomes, 3:115-120, 2016.
Hakimi et al., “Minimally invasive approaches to prostate cancer: a review of the current literature.”, Urol. J., 4: 130-137, 2007.
Hallberg and Palmer, “The role of the ALK receptor in cancer biology.”, Ann. Oncology, 27 (Suppl 3):iii4-iii15. doi: 10.1093/annonc/mdw301, 2016.
Haller et al., “Paediatric and adult soft tissue sarcomas with NTRK.1 gene fusions: a subset of spindle cell sarcomas unified by a prominent myopericytic/haemangiopericytic pattern,” J Pathol, Apr. 2016, 238(5):700-710.
Hamdouchi et al “Imidazo[1,2-b]pyridazines, novel nucleus with potent and broad spectrum activity against human picornavimses: design, synthesis, and biological evaluation” J Med Chem., Sep. 25, 2003;46(20):4333-4341.
Hansen et al., “Autophagic cell death induced by TrkA receptor activation in human glioblastoma cells,” J. of Neurochemistry, 2007, 103:259-275.
Harada et al., “Role and Relevance of TrkB Mutations and Expression in Non-Small Cell Lung Cancer,” Clinical Cancer Research, Jan. 17, 2011, 17(9):2638-2645.
Harris et al., “Multicenter Feasibility Study of Tumor Molecular Profiling to Inform Therapeutic Decisions in Advanced Pediatric Solid Tumors: The Individualized Cancer Therapy (iCat) Study,” JAMA Oncol, Jan. 2016; 10.1001/jamaoncol.2015.5689, 8 pages.
Harwood et al., “Experimental organic chemistry—Principles and practice,” Experimental Chemistry—Organic Chemistry and Reaction, Jan. 1, 1989, 127-132.
Hayashi et al., “Crizotinib treatment for refractory pediatric acute myeloid leukemia with RAN-binding protein 2-anaplastic lymphoma kinase fusion gene.”, Blood Cancer J, 6(8): e456, 2016.
Hechtman et al., “Identification of targetable kinase alterations in patients with colorectal carcinoma that are preferentially associated with wild-type RAS/RAF,” Mol. Cancer Res. 14(3):296-301, 2016.
Hechtman et al., Abstract 1837: Pan-TRK IHC Is an Efficient and Reliable Screening Assay for Targetable NTRK Fusions, Annual Meeting Abstracts, 2017, 457A.
Herzberg et al., “NGF involvement in pain induced by chronic constriction injury of the rat sciatic nerve,” Neuroreport, 1997, 8:1613-1618.
Hilfiker, Rolf, Fritz Blatter, and Markus von Raumer. “Relevance of solid-state properties for pharmaceutical products.” Polymorphism in the pharmaceutical industry (2006): 1-19.
Hinrichs et al., “Exploiting the curative potential of adoptive T-cell therapy for cancer,” Immunol Rev. Jan. 2014;257(1):56-71. doi: 10.1111/imr.12132.
Hobbs et al., “Effects of T-Cell Depletion on Allogeneic Hematopoietic Stem Cell Transplantation Outcomes in AML Patients,” J Clin Med. Mar. 19, 2015;4(3):488-503. doi: 10.3390/jcm4030488.
Hotta et al., “ALK: a tyrosine kinase target for cancer therapy”, Cold Spring Harb Mol Case Study, 3(1):a001115. doi: 10.1101/mcs.a001115, 20 pages, 2017.
Hong et al., “Clinical Safety and activity from a Phase 1 study ofLOXO-101, a selective TRKA/B/C inhibitor, in solid-tumor patients with NTRK gene fusions,” 2016 AAACR Annual Meeting, Apr. 17, 2016, 32 pages.
Hong et al., Abstract PR13: Clinical safety and activity from a phase 1 study ofLOXO-101, a selective TRKA/B/C inhibitor, in solid-tumor patients with NTRK gene fusions, Molecular Cancer Therapeutics 2015:14(12 Supplement 2):PR13.; Abstract only, 4 pages.
Hornick et al., “Expression of ROSI predicts ROSI gene rearrangement in inflammatory myofibroblastic tumors.”, Mod Pathol., 28(5): 732-9, 2015.
Hover et al., “Abstract TMOD-07: NTRK3 Gene Fusions Drive Tumorigenesis in Novel Models of Pediatric HighGrade Glioma,” Neuro-Oncology, Jun. 2017, iv49.
Howell et al., “Dynamic allele-specific hybridization. A new method for scoring single nucleotide polymorphisms,” Nat Biotechnol. Jan. 1999;17(1):87-8.
Hu et al., “Decrease in bladder overactivity with REN1820 in rats with cyclophosphamide induced cystitis,” J. Urology, 2005, 173(3):1016-1021.
Hu et al., “Identification of brain-derived neurotrophic factor as a novel angiogenic protein in multiple myeloma” Cancer Genetics and Cytogenetics, 2007, 178:1-10.
Huehls et al., “Bispecific T-cell engagers for cancer immunotherapy,” Immunol Cell Biol. Mar. 2015;93(3):290-6. doi: 10.1038/icb.2014.93. Epub Nov. 4, 2014.
Hyrcza et al., “Abstract OFP-06-007: Comparison of ultrastructural features between pediatric Mammary Analogue Secretory Carcinoma (MASC) of the salivary glands and Pediatric Secretory Breast Carcinoma (SBC) reveals similar pathological features,” Virchows Arch, Sep. 2016, 469(S1): S17.
Hyrcza et al., vol. 469, Supp. Supplement 1, pp. S17. Abstract No. OFP-1997-7; 31st International Congress of the International Academy of Pathology and the 28th Congress of the European Society of Pathology, Cologne, Germany. Sep. 25-29, 2016.
Igaz et al., “Biological and clinical significance of the JAK-STAT pathway; lessons from knockout mice,” Inflamm Res., 2001, 50:435-441.
Ihle et al., “The Roles of Jaks and Stats in Cytokine Signaling,” Cane. J. Sci. Am., 1998, 4(1):84-91.
Ihuegbu et al., “Non-invasive detection of crizotinib resistance in ALK-rearranged lung adenocarcinoma directs treatment with next-generation ALK inhibitors”, J Clin. Oncology, vol. 34, Supp. Supplement 15, Abstract No. e20643, 2016 Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, 2016.
Ikeda et al., “Basic Science”, Annals of Oncology. vol. 28 (suppl_1O): xl x6.10.1093/annonc/mdx652, 2017.
Imamura et al., “Allogeneic hematopoietic stem cell transplantation in adult acute lymphoblastic leukemia: potential benefit of medium-dose etoposide conditioning,” Exp Hematol Oncol, Jul. 16, 2015;4:20. doi: 10.1186/s40164-015-0015-0. eCollection 2015.
Iniguez-Ariza et al., “Abstract 6087: NTRK.1-3-point mutations in poor prognosis thyroid cancers,” J Clinical Oncology, May 2017, 35(15): 6087.
Isdori et al., “Advancement in high dose therapy and autologous stem cell rescue in lymphoma,” World J Stem Cells, Aug. 2015, 7(7):1039-1046.
Iyama et al., “Identification of Three Novel Fusion Oncogenes, SQSTM1/NTRK3, AFAP1L2/RET, and PPFIBP2/RET, in Thyroid Cancers of Young Patients in Fukushima,” Thyroid. 27(6):811-818, 2017.
Iyer et al., “AZ64 inhibits TrkB and enhances the efficacy of chemotherapy and local radiation in neuroblastoma xenografts,” Cancer Chemother Pharmacol. Sep. 2012;70(3):477-86. doi: 10.1007/s00280-012-1879-x. Epub May 24, 2012.
Iyer, R., “Entrectinib is a potent inhibitor of Trk-driven neuroblastomas in a xenograft mouse model.” Cancer letters 372.2 (2016): 179-186. (Year: 2016).
Jaggar et al., “Inflammation of the rat urinary bladder is associated with a referred thermal hyperalgesia which is nerve growth factor dependent,” Br. J. Anaesth, 1999, 83:442-448.
Jencks and Regenstein, “Ionization Constants of Acids and Bases,” Handbook of Biochemistry and Molecular Biology, 3rd ed., G.D. Fassman, CRC Press, 1976, 1: 305-347.
Jin et al., “TrkC plays an essential role in breast tumor growth and metastasis,” Carcinogenesis, 2010, 31(11):1939-1947.
Johnson et al., “Comprehensive Genomic Profiling of 282 Pediatric Low- and High-Grade Gliomas Reveals Genomic Drivers, Tumor Mutational Burden, and Hypermutation Signatures.”, Oncologist. 22(12): 1478-1490, 2017.
Jones et al., “Recurrent somatic alterations of FGFR1 and NTRK.2 in pilocytic astrocytoma,” Nature Genetics, 2013, 45:927-932.
Kao et al., “Recurrent BRAF Gene Fusions in a Subset of Pediatric Spindle Cell Sarcomas,” Am. J. Surg. Pathol. 42(1):28-38, 2018.
Karachialiou et al., “Real-time liquid biopsies become a reality in cancer treatment”, Ann. Transl. Med, 3(3):36, 2016.
Katayama et al., “Cabozantinib Overcomes Crizotinib Resistance in ROS1 Fusion-Positive Cancer”, Clin. Cancer Res., 21 (I): 166-7 4, 2015.
Katayama et al., “Mechanisms of Acquired Crizotinib Resistance in ALK Rearranged Lung Cancers,” Sci Transl Med, Feb. 2012, 4(120): 120ra17.
Katayama et al., “Therapeutic targeting of anaplastic lymphoma kinase in lung cancer: a paradigm for precision cancer medicine.”, Clin Cancer Res, 21(10): 2227-35, 2015.
Keysar et al., “A patient tumor transplant model of Squamous cell cancer identifies PBK inhibitors as candidate therapeutics in defined molecular bins,” Molecular Oncology, 2013, 7(4):776-790.
Kim et al., “Mammaglobin-A is a target for breast cancer vaccination”, OncoImmunology 5(2): e1069940, 2016.
Kim et al., “NTRK.1 fusion in glioblastoma multiforme,” PloS ONE, 2014, 9(3): e91940.
Kim et al., “SEC31A-ALK Fusion Gene in Lung Adenocarcinoma”, Cancer Res Treat, 48(1): 398-402,2016.
Klijn et al., “A comprehensive transcriptional portrait of human cancer cell lines,” Nat Biotechnol., 2015, 33(3):306-312.
Knezevich et al., “A novel ETV6-NTRK.3 gene fusion in congenital fibrosarcoma,” Nat Genet, Feb. 1998:18(2):184-187.
Knezevich et al., “ETV6-NTRK3 gene fusions and trisomy 11 establish a histogenetic link between mesoblastic nephroma and congenital fibrosarcoma,” Cancer Res, Nov. 1998:58(22):5046-5048.
Koboldt et al., “The next-generation sequencing revolution and its impact on genomics,” Cell, Sep. 26, 2013;155(1):27-38. doi: 10.1016/j.cell.2013.09.006.
Kohsaka et al., Pediatric soft tissue tumor of the upper arm with LMNA-NTRK1 fusion, Hum. Pathol. 72:167-173, 2017.
Kolokythas et al., “Nerve growth factor and tyrosine kinase A receptor in oral squamous cell carcinoma: is there an association with perineural invasion?” J. Oral Maxillofacial Surgery, 2010, 68(6):1290-1295.
Konicek et al., Cancer research, vol. 76, No. 14, Supp. Supplement. Abstract No. 2647; 107th Annual Meeting of the American Association for Cancer Research, AACR 2016. New Orleans, LA; Apr. 16-20, 2016; Abstract only, 3 pages.
Kralik et al., “Characterization of a newly identified ETV6-NTRK3 fusion transcript in acute myeloid leukemia,” Diagn. Pathol. 6:19, 2011.
Kremer et al., “The safety and efficacy of a JAK inhibitor in patients with active rheumatoid arthritis: Results of a double-blind, placebo-controlled phase Ila trial of three dosage levels of CP-690,550 versus placebo,” Arth. & Rheum., 2009, 60:1895-1905.
Kruttgen et al., “The dark side of the NGF family: neurotrophins in neoplasias,” Brain Pathology, 2006, 16:304-310.
Kubler et al., “Self-adjuvanted mRNA vaccination in advanced prostate cancer patients: a first-in-man phase I/Ila study.”, J. Immunother Cancer 3 :26, 2015.
Kusano et al., “Two Cases of Renal Cell Carcinoma Harboring a Novel STRN-ALK Fusion Gene.”, Am J SurgPathol. 40(6): 761-9, 2016.
Lamb et al., “Nerve growth factor and gastric hyperalgesia in the rat,” Neurogastrenterol. Motil., 2003, 15:355-361.
Lannon et al., “ETV6-NTRK3: a chimeric protein tyrosine kinase with transformation activity in multiple cell lineages,” Semin Cancer Biol, Jun. 2005:15(3):215-223.
Lansky et al., “The measurement of performance in childhood cancer patients,” Cancer, 1987, 60(7):1651-1651.
Lecht et al., “Angiostatic effects ofK252a, a TRK inhibitor, in murine brain capillary endothelial cells,” Mol Cell Biochem, Jun. 2010;339(1-2):201-13. doi: 10.1007/s11010-010-0386-9. Epub Feb. 11, 2010.
Lee et al., “Identification of ROS1 rearrangement in gastric adenocarcinoma.”, Cancer, 119(9): 1627-1635, 2013.
Leeman-Neill et al., “ETV6-NTRK3 is a common chromosomal rearrangement in radiation-associated thyroid cancer,” Cancer, 2014, 120(6):799-807.
Leukemia, Wikipedia the Free Encyclopedia, Dec. 8, 2001, https://en.wikipedia.org/wiki/Leukemia, 15 pages.
Leyvraz et al., Abstract No. 897. Meeting Info: 33. Deutscher Krebskongress, DKK. Berlin, Germany, 2018.
Lezcano et al., “Regular transfusion lowers plasma free hemoglobin in children with sickle-cell disease at risk for stroke,” Am. J. Surg. Pathol. doi: 10.1097/P AS.0000000000001070, 2018.
Li et al., “Brain derived neurotrophic factor (BDNF) contributes to the pain hypersensitivity following surgical incision in the rats,” Molecular Pain, 2008, 4(28):1-11.
Li et al., “Combinational Analysis of FISH and Immunohistochemistry Reveals Rare Genomic Events in ALK Fusion Patterns in NSCLC that Responds to Crizotinib Treatment”, J Thorac. Oneal., 12(1):94-101. doi: 10.1016/i .itho.2016.08.145, 2017.
Li et al., “Correlation of expressions of GFAP, NT-3, TRK and NCAM with neurotropic molecular mechanism and clinical factors in adenoid cystic carcinoma of salivary gland,” Chinese Journal of Cancer Prevention and Treatment, 2009, 16(6): 428-430 (with English abstract).
Li et al., “In vivo sensitized and in vitro activated B cells mediate tumor regression in cancer adoptive immunotherapy,” J Immunol, Sep. 1, 2009;183(5):3195-203. doi: 10.4049/jimmunol.0803773. Epub Aug. 10, 2009.
Li et al., “Lumbar 5 ventral root transection-induced upregulation of nerve growth factor in sensory neurons and their target tissues: a mechanism in neuropathic pain,” Mol. Cell. Neurosci., 2003, 23:232-250.
Li et al., “Trk inhibitor attenuates the BDNF/TrkB-induced protection of neuroblastoma cells from etoposide in vitro and in vivo,” Cancer Biol. Ther., Feb. 2015, 16(3):477-483.
Lin et al., “HG-48. Integrated sequencing of pediatric pilocytic Astrocytoma with anaplasia reveals molecular features of both Lowand high-grade glial tumors”, Neuro-Oneol, vol. 18, Supp. Supplement 3, pp. iii58, Abstract No. HG-48; 17th International Symposium on Pediatric Neuro-Oncology, ISPNO 2016. Liverpool, UK, Jun. 12, 2016-Jun. 15, 2016.
Lin et al., Neuro-Oncol, vol. 18, Supp. Supplement 3, pp. iii58, Abstract No. HG-48; 17th International Symposium on Pediatric Neuro-Oncology, ISPNO 2016. Liverpool, UK, Jun. 12, 2016-Jun. 15, 2016.
Linch et al., “Systemic treatment of soft-tissue sarcoma [mdash] gold standard and novel therapies,” Nature Reviews Clinical Oncology, 2014, 11(4):187-202.
Loh et al., “Treatment of infantile fibrosarcoma with chemotherapy and surgery: results from the Dana-Farber Cancer Institute and Children's Hospital, Boston,” J Pediatr Hematol Oncol, Dec. 2002:24(9):722-726.
Lorigan et al., “Phase III trial of two investigational schedules of ifosfamide compared with standard-dose doxombicin in advanced or metastatic soft tissue sarcoma: a European Organisation for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group Study,” J. Clin Oncol., 2007, 25(21):3144-3150.
Lovly et al., “Inflammatory myofibroblastic tumors harbor multiple potentially actionable kinase fusions,” Cancer Discov., 2014, 4(8):889-895.
Lu et al., “Targeted next generation sequencing identifies somatic mutations and gene fusions in papillary thyroid carcinoma,” Oncotarget. 8(28):45784-45792, 2017.
Ma and Woolf, “The progressive tactile hyperalgesia induced by peripheral inflammation is nerve growth factor dependent,” Neuroreport, 1997, 8:807-810.
Ma et al., “Responses to crizotinib in patients with ALK-positive lung adenocarcinoma who tested immunohistochemistry (IHC)-positive and fluorescence in situ hybridization (FISH)-negative”, Oncotarget, 7(39), 64410-64420, 2016.
Macleod, et al., “Abstract 0294: Gene Targets ofETV6-NTRK3 Fusion,” Haematologica, 14th Congress of the European Hematology Association,2009, 94(s2): 116.
Majweskaetal., CancerResearch, vol. 76, No. 14, Supp. Supplement. Abstract No. 3190. 107th Annual meeting ofthe American Association for Cancer Research, AACR. New Orleans, LA Apr. 16-20, 2016.
Makretsov et al., “A fluorescence in situ hybridization study ofETV6-NTRK3 fusion gene in secretory breast carcinoma,” Genes, Chromosomes and Cancer, Jun. 2004:40(2):152-157.
Marchetti et al., “Frequent mutations in the neurotrophic tyrosine receptor kinase gene family in large cell neuroendocrine carcinoma of the lung,” Human Mutation, 2008, 29(5):609-616.
Marras et al., “Genotyping SNPs with molecular beacons,” Methods Mol Biol, 2003;212:111-28.
Marras et al., Single Nucleotide Polymorphism: Methods and Protocols. Methods in Molecular Biology, Kwok, P.-Y., Ed., Totowa, NJ, Humana Press, vol. 212, pp. 111-128, 2003.
Martin-Zanca et al., “A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences,” Nature, 1986, 319:743-748.
Matayoshi, “Actions of brain-derived neurotrophic factor on spinal nociceptive transmission during inflammation in the rat,” J. Physiol., 2005, 569:685-695.
McCahon et al., “Non-Resectable Congenital Tumors with the ETV6-NTRK3 Gene Fusion Are Highly Responsive to Chemotherapy,” Med. Pediatr. Oncol., May 2003, 40(5):288-292.
McCarthy et al., “Tropomyosin receptor kinase inhibitors: a patent update 2009-2013,” Expert Opin TherPat. Jul. 2014;24(7):731-44. doi: 10.1517/13543776.2014.910195. Epub May 8, 2014.
McMahon et al., “The biological effects of endogenous nerve growth factor on adult sensory neurons revealed by a trkA-lgG fusion molecule,” Nat. Med., 1995, 1:774-780.
McMahon., “VEGF Receptor Signaling in Tumor Angiogenesis,” The Oncologist, 2000, 5(1): 3-10.
Mekinist, Highlights of Prescribing Information, Initial Approval 2013, revised Nov. 2015, Novartis Pharmaceuticals Com., 27 pages.
Melo-Jorge et al., The Chagas' disease parasite Trypanosoma cmzi exploits nerve growth factor receptor TrkA to infect mammalian hosts Cell Host & Microbe, 2007, 1(4):251-261.
Meyer et al., “Remarkable leukemogenic potency and quality of a constitutively active neurotrophin receptor, delta TrkA,” Leukemia, 2007, 21:2171-2180.
Milione et al., “Identification and characterization of a novel SCYL3-NTRK1 rearrangement in a colorectal cancer patient,” Oncotarget, 8(33):55353-55360, 2017.
Miranda et al., “Functional characterization of NTRK1 mutations Identified in melanoma,” Genes Chromosomes & Cancer, Jun. 26, 2014, 53(10):875-880.
Montagnoli et al., “Anti-proliferative effects of GW441756, a novel inhibitor of NGF receptor tyrosine kinase a (TRKA), in human sarcoma,” Italian Journal of Anatomy and Embryology, Nov. 11, 2010, 115:117.
Montalli et al., “Mammaglobin and DOG-1 expression in polymorphous low-grade adenocarcinoma: an appraisal of its origin and morphology,” J Oral Pathol Med., Mar. 2017, 46(3):182-187.
Morissette et al., “High-throughput crystallization: polymorphs, salts, co-crystals and solvates of pharmaceutical solids,” Adv. Drug Deliv Rev, 2004, 56: 375-300.
Mulligan, “RET revisited: expanding the oncogenic portfolio.”, Nature Reviews Cancer, 14, 173-186,2014.
Murakami et al., “Integrated molecular profiling of juvenile myelomonocytic leukemia”, Blood, blood-2017-07-798157; DOI: 10.1182/blood-2017-07-798157, 2018.
Myers, “Synthesis of Chiral Amines by Asymmetric Additions to tert-Butylsulfinimines (Ellman Auxiliary),” Chem 115, retrieved on May 18, 2016, retrieved from the Internet. URL: <faculty.chemistry.harvard.edu/files/myers/files/15-ellman auxiliarv.pelf>, 6 pages.
Nagasubmamanian et al., “Brief Report: Infantile Fibrosarcoma With NTRK3-ETV6 Fusion Successfully Treated with the Tropomyosin-Related Kinase Inhibitor LOXO-101,” Pediatric Blood & Cancer, 2016, DOI 10.1002, 3 pages.
Nakagawara, “Trk receptor tyrosine kinases: a bridge between cancer and neural development,” Cancer Letters, 2001, 169(2):107-114.
Nakano et al., “Novel Oncogenic KLC1-ROS1 Fusion in Pediatric Low Grade Glioma”, Pediatr Blood Cancer. vol. 64, S54-S55 Suppe. 4. 013-1-7, 2017.
Narayanan et al., “Discovery and preclinical characterization of novel small molecule TRK and ROS1 tyrosine kinase inhibitors for the treatment of cancer and inflammation,” PLoS One, Dec. 26, 2013;8(12):e83380. doi: 10.1371/iournal.pone.0083380. eCollection 2013.
National Cancer Institute at the National Institutes of Health, posted on or before Jan. 5, 2000,n retrieved on Jan. 13, 2015, http://www.cancer.gov/, 2 pages.
National Comprehensive Cancer Network, posted on or before Dec. 3, 1998, retrieved on Jan. 13, 2015, http://www.nccn.org/, 1 page.
NCT02050919, “Sorafenib Tosylate, Combination Chemotherapy, Radiation Therapy, and Surgery in Treating Patients with High-Risk Stage IIB-IV Soft Tissue Sarcoma,” ClinicalTrials.gov, Last Updated Dec. 16, 2015, https://www.clinicaltrials.gov/ct2/show/NCT02050919, 5 pages.
NCT02122913, “Oral TRL Inhibitor LOXO-101 for Treatment of Advanced Adult Solid Tumors,” ClinicalTrials.gov, Last Updated Dec. 7, 2015, https://clinicaltrials.gov/ct2/show/NCT02122913.
Ni et al., “siRNA interference with a proliferation-inducing ligand gene in the Sgr-7901 gastric carcinoma cell line,” Asian Pacific Journal of Cancer Prevention, 2012, 13:1511-1514.
Ni et al., “Tyrosine receptor kinase Bis a drug target in astrocytomas,” Neuro Oncol., Jan. 2017, 19(1):22-30.
NIH National Cancer Institute [online], “progression (pm-GREH-shun),” NCI Dictionary of Cancer Terms, retrieved on Sep. 17, 2018, URL: <https://www.cancer.gov/publications/dictionaries/cancer-terms/def/progression>, 1 page.
NIH National Cancer Institute [online], “recurrence (ree-KER-ents),” NCI Dictionary of Cancer Terms, retrieved on Sep. 21, 2018, URL: <https://www.cancer.gov/publications/dictionaries/cancer-terms/def/recurrence>, 1 page.
NIH National Cancer Institute [online], “relapse (REE-laps),” NCI Dictionary of Cancer Terms, retrieved on Sep. 17, 2018, URL: <https://www.cancer.gov/publications/dictionaries/cancer-terms/def/relapse>, 1 page.
NIH, “List of Cancer-causing Agents Grows,” National Institute of Environmental Health Sciences, https://www.niehs.nih.gov/news/newsroom/releases/2005/january31/index.cfm, 4 pages.
Nikiforova et al., Abstract No. 5. Meeting Info: 84th Annual Meeting of the American Thyroid Association. Coronado, CA, United States, 2014.
Nollau et al., “Methods for detection of point mutations: performance and quality assessment. IFCC Scientific Division, Committee on Molecular Biology Techniques,” Clin Chem. Jul. 1997;43(7):1114-28.
Obianyo et al., “Novel small molecule activators of the Trk family of receptor tyrosine kinases. BiochimBiophys Acta, 1834:2214-2218,” BiochimBiophys Acta, Oct. 2013, 1834(10):2213-2218.
Ocgene.bioinfo-minzhao org [online]. “Ovarian Cancer Gene Database, Gene ID: 4914,” [retrieved on Jul. 17, 2017] Retrieved from the Internet: URL<ocgene.bioinfominzhao.org/gene mutation/cgi?gene=4914>, 13 pages.
Oken et al., “Toxicity and response criteria of the Eastern Cooperative Oncology Group,” Am J Clin Oncol, 1982, 5:649-655.
Olivier, “The Invader assay for SNP genotyping,” Mutat Res, Jun. 3, 2005;573(1-2): 103-10.
Orbach et al., “Conservative strategy in infantile fibrosarcoma is possible: The European paediatric Soft tissue sarcoma Study Group experience,” Eur J Cancer, Apr. 2016, 57:1-9.
Orbach et al., “Infantile fibrosarcoma: management based on the European experience,” J Clin Oncol, Jan. 2010, 28(2):318-323.
O'Shea, “Jaks, STATs, cytokine signal transduction, and immunoregulation: are we there yet?” Immunity, 1997, 7:1-11.
Otsubo et al., “Sporadic pediatric papillary thyroid carcinoma harboring the ETV6/NTRK3 fusion in oncogene in a 7-year-old Japanese girl: a case report and review of literature,” J. Pediatr. Endocrinol. Metab. 28;31(4):461-467, 201.
Ott et al., “An immunogenic personal neoantigen vaccine for patients with melanoma.”, Nature 547: 217-221, 2017.
Ou et al., “Emergence of novel and dominant acquired EGFR solvent-front mutations at Gly796 (G796S/R) together with C797S/R and L792F/H mutations in one EGFR (L858R/T790M) NSCLC patient who progressed on osimertinib,” Lung Cancer, 2017, 108: 228-231.
Ou et al., “Identification of a novel TMEM106B-ROS1 fusion variant in lung adenocarcinoma by comprehensive genomic profiling.”, Lung Cancer, 88(3):352-4, 2015.
Ou et al., “Next-Generation Sequencing Reveals a Novel NSCLC ALK Fl 174V Mutation and Confirms ALK G1202R Mutation Confers High-Level Resistance to Alectinib (CH5424802/RO5424802) in ALK-Rearranged NSCLC Patients Who Progressed on Crizotinib,” Journal of Thoracic Oncology, Apr. 2014, 9: 549-553.
Ovarian Cancer Gene Database, ocgene.bioinfo-minzhao.org/gene_mutation.cgi?gene=49 1 4, downloaded on May 31, 2016, 14 pages.
Ovarian Cancer Gene Database, ocgene.bioinfo-minzhao.org/gene_mutation.cgi?gene=4916, downloaded on May 31, 2016, 21 pages.
Pan et al., Laboratory Investigation, vol. 96, Supp. Suppl. 1, pp. 367A, Abstract No. 1450, 105th Annual Meeting of the United States and Canadian Academy of Pathology, Seattle, WA, 2016.
Panagopoulos et al., “Recurrent fusion of the genes FN1 and ALK in gastrointestinal leiomyomas”, Modem Pathology 29: 1415-1423, 2016.
Pao, W., et al. “Acquired resistance of lung adenocarcinomas to gefitinib or erlotinib is associated with a second mutation in the EGFR kinase domain,” PLoS Med, Feb. 2005, 2(3), e73.
Papatsoris et al., “Manipulation of the nerve growth factor network in prostate cancer,” Exper Opin Invest Drugs, 2007, 16(3):303-309.
Park et al., “Genomic alterations in BCL2L1 and DLC1 contribute to drug sensitivity in gastric cancer,” Proc. Natl. Acad. Sci. U.S.A., Oct. 2015, 112(40):12492-12497.
Park et al., “NTRK1 fusions for the therapeutic intervention of Korean patients with colon cancer,” Oncotarget. 7(7):8399-412, 2016.
Patani et al., “Bioisosterism: A rational approach in Drug Design,” Chem Rev., Dec. 1996, 96(8):3147-3176.
Patapoutian et al., “Trk receptors: mediators of neurotrophin action,” Current Opinion in Neurobiology, 2001, 11:272-280.
Pavlick et al., “Identification of NTRK fusions in pediatric mesenchymal tumors,” Pediatr Blood Cancer, Aug. 2017, 64(8). doi: 10.1002/pbc.26433. Epub Jan. 18, 2017.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2009/057729, dated Mar. 22, 2011, 7 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2009/061519, dated Apr. 26, 2011, 6 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2010/041538, dated Jan. 10, 2012, 7 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US201 1/036452, dated Nov. 29, 2012, 6 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2015/060953, dated May 16, 2017, 7 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2016/035327, dated Dec. 14, 2017, 9 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2016/058951, dated May 11, 2018, 11 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2017/033257, dated Nov. 20, 2018, 8 pages.
PCT International Preliminary Report on Patentability in International Application. No. PCT/US2017/058518, dated Apr. 30, 2019, 8 pages.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2018/022833, dated Sep. 26, 2019, 8 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2009/0161519, dated Feb. 2, 2010, 8 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2009/057729, dated Feb. 4, 2010, 10 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2010/041538, dated Oct. 1, 2010, 10 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2011/036452, dated Aug. 18, 2011, 9 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2015/060953, dated Feb. 8, 2016, 12 pages.
PCT International Search Report and Written Opinion for International Application No. PCT/US2016/035327, dated Aug. 18, 2016, 15 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2016/058951, dated Feb. 7, 2017, 20 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2017/025932, dated May 31, 2017, 16 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2017/025939, dated May 31, 2017, 16 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2017/033257, dated Jul. 24, 2017, 13 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2017/058518, dated May 2, 2018, 17 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2018/022833, dated Aug. 13, 2018.
PCT International Search Report and Written Opinion in International Application No. PCT/US2018/039502, dated Apr. 16, 2018, 16 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2018/057542, dated Mar. 6, 2019, 19 pages.
PCT International Search Report and Written Opinion in International Application No. PCT/US2019/024961, dated Jul. 23, 2019, 13 pages.
Pediatric Cancer Gene Database, pedican.bioinfominzhao.org/gene_mutation.cgi?gene=4914, downloaded on May 31, 2016, 6 pages.
Pediatric Cancer Gene Database, pedican.bioinfominzhao org/gene_mutation.cgi?gene=4915, downloaded on May 31, 2016, 5 pages.
Pediatric Cancer Gene Database, pedican.bioinfominzhao.org/gene_mutation.cgi?gene=4916, downloaded on May 31, 2016, 9 pages.
Perales et al., “Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies,” Biol Blood Marrow Transplant, Jan. 2016;22(1):17-22. doi: 10.1016/j.bbmt.2015.10.014. Epub Oct. 17, 2015.
Perez-Pinera et al., “The Trk tyrosine kinase inhibitor K252a regulates growth of lung adenocarcinomas,” Molecular and Cellular Biochemistry, 2007, 295(1&2):19-26.
Perrault et al., “The Synthesis ofN-Aryl-5(S)-aminomethy1-2-oxazolidinone Antibacterials and Derivatives in One Step from Aryl Carbamates,” Org. Process Res. Dev., 2003, 7:533-546.
Peus et al., “Appraisal of the Kamofsky Performance Status and proposal of simple algorithmic system for its evaluation,” BMC Med Inform and Decision Making, 2013, 13:72.
Picarsic et al., “Molecular characterization of sporadic pediatric thyroid carcinoma with the DNA/RNA ThyroSeq v2 next-generation sequencing assay,” Pediatr. Dev. Pathol, Mar. 2016, 19(2):115-122.
Pierottia and Greco, “Oncogenic rearrangements of the NTRK1/NGF receptor,” Cancer Letters, 2006, 232:90-98.
Pinedo et al., “Translational Research: The Role of VEGF in Tumor Angiogenesis,” The Oncologist, 2000, 5(1): 1-2.
Pinski et al., “Trk receptor inhibition induces apoptosis of proliferating but not quiescent human osteoblasts,” Cancer Res, 2002, 62:986-989.
Plosker, “Sipuleucel-T: in metastatic castration-resistant prostate cancer.”, Drugs 71(1): 101-108, 2011.
Ponsaerts et al., “Cancer immunotherapy using RNA-loaded dendritic cells,” Clin. Exp. Immunol., Dec. 2003, 134:378-384.
Prabhakaran et al., “Novel TLE4-NTRK2 fusion in a ganglioglioma identified by array-CGH and confirmed by NGS: Potential for a gene targeted therapy,” Neuropathology, Mar. 2018, doi:10.1111/neup.12458.
Prasad et al., “NTRK fusion oncogenes in pediatric papillary thyroid carcinoma in northeast United States,” Cancer, Apr. 2016, 122(7):1097-1107.
PubChem, “Larotrectinib,” https://pubchem.ncbi.nlm.nih.gov/compound/46188928, retrived on Apr. 29, 2019, 20 pages.
Pulciani et al., “Oncogenes in solid human tumours,” Nature, 1982, 300(5892):539-542.
Qaddoumi et al., “Genetic alterations in uncommon low-grade neuroepithelial tumors: BRAF, FGFR1, and MYB mutations occur at high frequency and align with morphology,” Acta Neuropathol, Jun. 2016, 131(6):833-845.
Qiu et al., “Next generation sequencing (NGS) in wild type GISTs”, J Clin. Oneal. 35: 15 _suppl, e22507-e22507,2017.
Ramer and Bisby, “Adrenergic innervation of rat sensory ganglia following proximal or distal painful sciatic neuropathy: distinct mechanisms revealed by anti-NFD treatment,” Eur. J. Neurosci., 1999, 11:837-846.
Rausch et al., “mRNA vaccine CV9103 and CV9104 for the treatment of prostate cancer.”, Human Vaccinimmunother 10(11): 3146-52, 2014.
Raychaudhuri et al., K252a, a high-affinity nerve growth factor receptor blocker, improves psoriasis: an in vivo study using the severe combined immunodeficient mouse-human skin model, J. Investigative Dermatology, 2004, 122(3):812-819.
Reshmi et al., “Abstract 477: Genomic and Outcome Analyses of Philadelphia Chromosome like (Ph-like) NCI Standard Risk B-Acute Lymphoblastic Leukemia (SR B-ALL) Patients Treated on Children's Oncology Group (COF) AALL0331,” Blood, 2017, 130(SI): 477.
Reungwetwattana et al., “Targeted therapies in development for non-small cell lung cancer,” J Carcinog, Dec. 2013, 12:22, doi: 10.4103/1477-3163.123972. eCollection 2013.
Reuther et al., “Identification and characterization of an activating TrkA deletion mutation in acute myeloid leukemia,” Mol. Cell. Biol. 2000, 20:8655-8666.
Ricarte-Filho et al., “Identification of kinase fusion oncogenes in post-Chernobyl radiation-induced thyroid cancers,” J. Clin. Invest, Nov. 2013, 123(11): 4935-4944.
Ricci et al., Neurotrophins and neurotrophin receptors in human lung cancer, Am. J. Respiratory Cell and Molecular Biology, Oct. 2001, 25(4): 439-446.
Richard et al., “Syngeneic stem cell transplant for spent-phase polycythaemia vera: eradication of myelofibrosis and restoration of normal haematopoiesis,” Br. J Haematol., Apr. 2002, 117(1):245-246.
Rimkunas et al., “Analysis of receptor tyrosine kinase ROSI-positive tumors in non-small cell lung cancer: identification of a FIG-ROSI fusion.”, Clin. Cancer Res., 18: 4449-58, 2012.
Ritterhouse et al., “ROSI Rearrangement in Thyroid Cancer.”, Thyroid, 26(6): 794-7, 2016.
Ro et al., “Effect of NGF and anti-NGF on neuropathic pain in rats following chronic constriction iniury of the sciatic nerve,” Pain, 1999, 79:265-274.
Roberts et al., “Targetable kinase-activating lesions in Ph-like acute lymphoblastic leukemia,” N Engl J Med, Sep. 2014, 371(11):1005-1015.
Roberts et al., Blood, vol. 128, No. 22. Abstract No. 278, 58th Annual Meeting of the American Society of Hematology, ASH 2016. San Diego, CA, United States. Dec. 3, 2016-Dec. 6, 2016, 2 pages.
Roblin et al., “Topical TrkA Kinase Inhibitor CT327 is an Effective, Novel Therapy for the Treatment of Pmritus due to Psoriasis: Results from Experimental Studies, and Efficacy and Safety of CT327 in a Phase 2b Clinical Trial in Patients with Psoriasis,” Acta Denn. Venereal., 2015, 95:542-548.
Rosenbaum et al., “Next Generation Sequencing Reveals Genomic Heterogenity of ALK Fusion Breakpoints in Non-Small Cell Lung Cancer”, Laboratory Investigation, vol. 96, Supp. Suppl. 1, pp. 481A-482A, Abstract No. 1914, 105th Annual Meeting of the United States and Canadian Academy of Pathology, Seattle, WA, 2016.
Roskoski, Jr. et al., “Classification of small molecule protein kinase inhibitors based upon the structures of their drug-enzyme complexes,” Pharmacological Research, 2016, 103: 26-48.
Ross et al., “New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing,” Oncologist, 2014, 19:235-242.
Rossi et al., “Abstract 84: RNA-Sequencing Identifies ETV6-NTRAK3 as a Gene Fusion Involved in Gastrointestinal Stromal Tumors,” Meeting Info: 105th Annual Meeting of the United States and Canadian Academy of Pathology, Seattle, WA, Annual Meeting Abstracts, 24A.
Rubin et al., “Congenital mesoblastic nephroma t(12;15) is associated with ETV6-NTRK3 gene fusion: cytogenetic and molecular relationship to congenital (infantile) fibrosarcoma,” Am. J. Pathol, Nov. 1998, 153(5):1451-1458.
Rubin et al., “Growth, survival and migration: the Trk to cancer,” Cancer Treat Res, 2003, 115:1-18.
Russo et al., “Acquired Resistance to the Trk Inhibitor Entrectinib in Colorectal Cancer,” Cancer Discovery, Jan. 1, 2016, 6(1):36-44.
Rutkowski et al., “Treatment of advanced dermatofibrosarcoma protuberans with imatinib mesylate with or without surgical resection,” J. Eur. Acad. Dermatol. Venereol., 2011, 25:264-270.
Sahin et al., “Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer.”, Nature 547: 222-226, 2017.
Saborowski et al., “Mouse model of intrahepatic cholangiocarcinoma validates FIG-ROS as a potent fusion oncogene and therapeutic target.”, Proc. Natl. Acad Sci. USA, 110(48): 19513-19518, 2013.
Santoro et al., “Doxombicin versus CYVADIC versus doxombicin plus ifosfamide in first-line treatment of advanced soft tissue sarcomas: a randomized study of the European Organization for Research and Treatment of Cancer Soft Tissue and Bone Sarcoma Group,” J. Clin Oncol., 1995, 13(7):1537-1545.
Saragovi et al., “A TrkA-selective, fast internalizing nerve growth factor-antibody complex induces trophic but not neuritogenic signals,” J Biol Chem, Dec. 25, 1998 25;273(52):34933-34940.
Sartore-Bianchi et al., “Sensitivity to Entrectinib Associated With a Novel LMNA-NTRK1 Gene Fusion in Metastatic Colorectal Cancer,” J. Natl. Cancer Inst, Nov. 2015, 108(1). doi: 10.1093/inci/div306.
Sassolas et al., “Oncogenic alterations in papillary thyroid cancers of young patients,” Thyroid Jan. 2012, 22(1):17-26.
Scaruffi et al., “Detection of DNA polymorphisms and point mutations of high-affinity nerve growth factor receptor (TrkA) in human neuroblastoma,” Int. J. Oneal., May 1999, 14:935-938.
Schmidt et al., “Heilmittelchemische untersuchungen in der Heterocyclischen Rihe. Pyrazolo-(3,4-D)-Pyrimidine (Medicinal chemical studies in the heterocyclic series. Pyrazolo-(3,4-D)-Pyrimidine),” Helvetica Chimica, Verlag Helvetica Chimica Acta, Jan. 1956, 39: 986-991 (with English Abstract).
Schmidt, Charles. “Combinations on trial.” Nature 552.7685 (Dec. 21, 2017): S67-S69.
Schram et al., “Abstract LB-302: Potential role of larotrectinib (LOXO-101), a selective pan-TRK inhibitor, in NTRX fusion-positive recurrent glioblastoma, ”Cancer Research, Jul. 2017, DOI: 10.1158/1538-7445.AM2017-LB-302, 2 pages.
Schrock et al., “Gastrointestinal tumours, non-colorectal”, Annals of Oncology. vol. 27, Suppl 6, 6130, 2016.
Shah et al., “Cardiac metastasis and hypertrophic osteoarthropathy in recurrent infantile fibrosarcoma,” Pediatr. Blood Cancer, Jul. 2012, 59(1):179-181.
Shaver et al., “Diverse, Biologically Relevant, and Targetable Gene Rearrangements in Triple-Negative Breast Cancer and Other Malignancies.”, Cancer Res, 76(16): 4850-60, 2016.
Shaw et al., “Ceritinib in ALK-rearranged non-small-cell lung cancer,” N Engl J Med, Mar. 27, 2014;370(13):1189-97. doi: 10.1056/NEJMoal311 107.
Shaw et al., “Crizotinib in ROSI-rearranged non-small-cell lung cancer,” N Engl J Med, Nov. 20, 2014;371(21):1963-71. doi: 10.1056/NEJMoal406766. Epub Sep. 27, 2014.
Shaw et al., “Tyrosine kinase gene rearrangements in epithelial malignancies,” Nat Rev Cancer, Nov. 2013, 13(11):772-787.
Sheldrick, “A short history of SHELX,” Acta Crystallogr A, Jan. 2008, 64(Pt1): 112-122.
Shelton et al., “Nerve growth factor mediates hyperalgesia and cachexia in auto-immune arthritis,” Pain, 2005, 116:8-16.
Sheng et al., “Congenital-infantile fibrosarcoma. A clinicopathologic study of 10 cases and molecular detection of the ETV6-NTRK3 fusion transcripts using paraffin-embedded tissues,” Am. J Clin. Pathol., Mar. 2001, 115:348-355.
Sigal, et al., “Activity of Entrectinib in a Patient with the First Reported NTRK Fusion in Neuroendocrine Cancer,” J. Natl. Compr. Cane. Netw, Nov. 2017, 15(11): 1317-1322.
Silverman, The Organic Chemistry of Drug Design and Drug Action, Second Edition, 2007, 20-21.
Sims et al., Abstract P280: Profiling abscopal regression in a pediatric fibrosarcoma with a novel EML4-NTRK3 fusion using immunogenomics and high-dimensional histopathology, J mmunotherapy of Cancer, Nov. 2016, 4(S1): 73.
Skalova et al., “Mammary Analogue Secretory Carcinoma of Salivary Glands: Molecular Analysis of 25 ETV6 Gene Rearranged Tumors with Lack of Detection of Classical ETV6-NTRK3 Fusion Transcript by Standard RT-PCR: Report of 4 Cases Harboring ETV6-X Gene Fusion,” Am. J. Surg. Pathol, Jan. 2016, 40(1):3-13.
Skalova et al., “Molecular Profiling of Mammary Analog Secretory Carcinoma Revealed a Subset of Tumors Harboring a Novel ETV6-RET Translocation: Report of 10 Cases,” Am. J. Surg. Pathol, Feb. 2018, 42(2):234-246.
Skalova et al., “Newly described salivary gland tumors,” Modem Pathology, Jan. 2017, 30, S27-S43.
Sleijfer et al., “Prognastic and predictive factors for outcome to firs-line ifosfamide-containing chemotherapy for adult patients with advanced soft tissue sarcomas: an exploratory, retrospective analysis on large series from the European Organization for Research and Treatment of Cancer-Soft Tissue and Bone Sarcoma Group,” Eur J. Cancer, 2010, 46(1):72-83.
Sleijfer et al., “Using single-agent therapy in adult patients with advanced soft tissue sarcoma can still be considered standard care,” Oncologist, 2005, 10(10):833-841.
Smith et al., “Annotation of human cancers with EGFR signaling-associated protein complexes using proximity ligation assays,” Sci Signal, 2015, 8(359):ra4, 12 pages.
Sohrabji et al., “Estrogen-BDNF interactions: implications for neurodegenerative diseases,” Frontiers in Neuroendocrinology, 2006, 27(4):404-414.
Song et al., “Molecular Changes Associated with Acquired Resistance to Crizotinib in ROS1-Rearranged Non-Small Cell Lung Cancer.”, Clin. Cancer Res., 21(10): 2379-87, 2015.
Stephens et al., “Trk receptors use redundant signal transduction pathways involving SHC and PLC-gamma 1 to mediate NGF responses,” Neuron, Mar. 1994, 12(3):691-705.
Stransky et al., “The landscape of kinase fusions in cancer,” Nature comm., 2014, 5:4846.
Subramaniam et al., Abstract 2019: RNA-Seq analysis of glioma tumors to reveal targetable gene fusions, 2017 Annual Meeting of the American Society of Clinical Oncology,2017, 1 page.
Sun et al., “P-loop conformation governed crizotinib resistance in G2032R-mutated ROSI tyrosine kinase: clues from free energy landscape,” PLoS computational biology, Jul. 17, 2014, 10(7): e1003729.
Tacconelli et al., “TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma,” Cancer Cell, 2004, 6:347-360.
Tafinlar, Highlights of Prescribing Information, GlaxoSmithKline, Jan. 2014, 41 pages.
Tahira et al., “dbQSNP: a database of SNPs in human promoter regions with allele frequency information determined by single-strand conformation polymorphism-based methods,” Hum Mutat, Aug. 2005;26(2):69-77.
Taipale et al., “Chaperones as thermodynamic sensors of drug-target interactions reveal kinase inhibitor specificities in living cells,” Nat Biotech, 2013, 31(7):630-637.
Tan et al., “Genetic landscape of ALK+ non-small cell lung cancer (NSCLC) patients (pts) and response to ceritinib in ASCEND-I”, J. Clin. Oncology, vol. 34, Supp. Supplement 15, Abstract No. 9064, 2015 Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, 2016.
Tanaka et al., “Brain-derived neurotrophic factor (BDNF)-induced tropomyosin-related kinase B (Trk B) signaling is a potential threapeutic target for peritoneal carcinomatosis arising from colorectal cancer,” PLoS One May 6, 2014, 9(5):e96410.
Tannenbaum, et al., “Abstract 749: Characterization of a Novel Fusion Gene, EML4-NTRK3, in Infantile Fivrosarcoma,” Pediatr Blood Cancer, DOI 10.1002/pbc, 1 page.
Tannenbaum-Dvir et al., “Characterizatio of a novel fusion gene EML4-NTRK3 in a case of recurrent congenital fibrosarcoma,” Cold Spring Harb. Mol. Case Stud., Oct. 1, 2015(1):a00471.
Tarate et al., “Oral Solid Self-Emulsifying Formulatins: A Patent Review,” Recent Patents on Drug Delivery & Formulation, 2014, 8(2):126-143.
Taylor et al., “Abstract 794: Characterization of NTRK fusions and Therpeutic Response to NTRK Inhibition in Hematologic Malignancies,” Blood, 2017, 130: 794.
The Cancer Genome Atlas Netowrk, “Comprehensive Molecular Characterizaito of Human colon and Rectal Cancer,” Nature, Jan. 2013, 487(7407): 330-337.
Theodosiou et al., “Hyperalgesia due to nerve damage: role of Nerve growth factor,” Pain, 1999, 81:245-255.
Thiele, “On Trk—the TrkB signal transduction pathway is an increasingly importang target in cancer biology,” Clinical Cancer Research, 2009, 105(19):5962-5967.
Thompson et al., “Brain-derived neurotrophic factor is an endogenous modulator of nociceptive responses iin the spinal cord,” Proc. Natl. Acad. Sci. USA, 1999, 96:7714-7718.
Thress et al., “Identification and preclinical characterization of AZ-23, a novel, selective, and orally bioavailable inhibitor of the Trk kinase pathway,” Mol Cancer Ther, Jul. 2009;8(7):1818-27. doi: 10.1158/1535-7163.MCT-09-0036. Epub Jun. 9, 2009.
Truzzi et al., “Neurotrophins and their receptors stimulate malanoma cell proliferation and migration,” J. Investigative Dermatology, 2008, 128(3):2031-2040.
Truzzi et al., “Neurotrophins in healthy and diseased skin,” Dermato-Endrocrinology, 2008, 3(1):32-36.
Tognon et al., “Expression of the ETV6-NTRK3 gene fusion as a primary event in human secretory breast carcinoma,” Cancer Cell, Nov. 2002, 2(5):367-376.
Turtle et al., “Artificial antigen-presenting cells for use in adoptive immunotherapy,” Cancer J, Jul.-Aug. 2010;16(4):374-51. doi: 10.1097/PPO.0b013e3181eb33a6.
UniProtKB/Swiss-Prot: P04629.4, “RecName: Full=High affinity nerve growth factor receptor; AltName: Full=Neurotrophic tyrosine kinase receptor type 1; AltName: Full=TRK.1-transforming tyrosine kinase protein; AltName: Full=Tropomyosin-related kinase A; AltName: Full=Tyrosine kinase receptor; AltName: Full=Tyrosine kinase receptor A; Short=Trk-A; AltName: Full=gp140trk; AltName: Full=p140-TrkA; Flags: Precursor,” May 14, 2014, 28 pages, available at URL<https://www.ncbi.nlm.nih.gov/protein/94730402?sat=18&satkey=12407077>.
UniProtKB/Swiss-Prot: Q16288.2, “RecName: Full=NT-3 growth factor receptor; AltName: Full=GP145-TrkC; Short=Trk-C; AltName: Full=Neurotrophic tyrosine kinase receptor type 3; AltName: Full=TrkC tyrosine kinase; Flags: Precursor,” May 14, 2014, 13 pages, available at URL<www.ncbi.nlm.nih.gov/protein/134035335?report=genbank&log$=protalign&blast_rank=O&RID=0>.
UniProtKB/Swiss-Prot: Q16620.1, “RecName: Full=BDNF/NT-3 growth factors receptor; AltName: Full=GP145-TrkB; Short=Trk-B; AltName: Full=Neurotrophic tyrosine kinase receptor type 2; AltName: Full=TrkB tyrosine kinase; AltName: Full=Tropomyosin-related kinase B; Flags: Precursor,” May 14, 2014, 17 pages, available at URL<www.ncbi.nlm.nih.gov/protein/2497560?report=genbank&log$=protalign&blast_rank=O&RID=0>.
Vaishnavi et al., “Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer.”, Nature Med 19: 1469-1472, 2013.
Vaishnavi et al., “TRK.ing Down an Old Oncogene in a New Era of Targeted Therapy,” Cancer Discovery, Jan. 2015, 5(1):25-34.
Van Gurp et al., “Phase 1 dose-escalation study of CP-690 550 in stable renal allograft recipients: preliminary findings of safety, tolerability, effects on lymphocyte subsets and pharmacokinetics,” Am. J. Transpl., 2008, 8:1711-1718.
Van Noesel et al., “Pediatric neuroblastomas: genetic and epigenetic ‘danse macabre’,” Gene, 2004, 325:1-15.
Vanden et al., “endocrine and neuroendocrine tumours”, Annals of Oncology, vol. 27, Supp. Supplement 6. Abstract No. 427PD 4pt European Society for Medical Oncology Congress, ESMO 2016; Copenhagen, Denmark; Oct. 7-11, 2016.
Vippagunta et al., “Crystalline Solids” Advanced Drug Delivery Rev., 2001, 48(1): 3-26.
Vogelstein and Kinzler, The Genetic Basis of Human Cancer, 2nd ed., 2002, ng 3, col. 1, para 2.
Wadhwa et al., “Expression of the neurotrophin receptors Trk A and Trk Bin adult human astrocytoma and glioblastoma,” Journal of Biosciences, 2003, 28(2):181-188.
Walch et al., “Role of neurotrophins and neurotrophins receptors in the in vitro invasion and heparanase production of human prostate cancer cells,” Clin. Exp. Metastasis, 1999, 17:307-314.
Walther et al., “Cytogenetic and single nucleotide polymorphism array findings in soft tissue tumors in infants,” Cancer Genet, Jul.-Aug. 2013, 206(7-8): 299-303.
Wang et al., “Design, synthesis and biological evaluation of novel 4-arylaminopyrimidine derivatives possessing a hydrazone moiety as dual inhibitors of L1196M ALK and ROS1.”, Eur. J Med Chem., 123, 80-99, 2016.
Wang et al., “Identification of 4-aminopyrazolylpyrimidines as potent inhibitors of Trk kinases,” J Med Chem, Aug. 14, 2008;51(15):4672-84. doi: 10.1021/jm800343j. Epub Jul. 23, 2008.
Wang et al., “Identification ofNTRK3 fusions in childhood melanocytic neoplasms,” J. Mol. Diagn, May 2017, 19(3):387-396.
Wang et al., “T cells sensitized with breast tumor progenitor cell vaccine have therapeutic activity against spontaneous HER2/neu tumors,” Breast Cancer Res Treat, Jul. 2012;134(1):61-70. doi: 10.1007/s10549-011-1912-5. Epub Dec. 16, 2011.
Wang et al., “Trk kinase inhibitors as new treatments for cancer and pain,” Expert Opin. Ther Patents, Mar. 2009, 19(3):305-319.
Wang, “Pan-cancer analysis of ROSI genomic aberrations”, University of Hong Kong, Pokfulam, Hong Kong SAR (Thesis), 44 pages, 2015.
Watanbe et al., “Cryptic t(12;15)(p13;q26) producing the ETV6-NTRK3 fusion gene and no loss of IGF2 imprinting in congenital mesoblastic nephroma with trisomy 11: fluorescence in situ hybridization and IGF2 allelic expression analysis,” Cancer Genet. Cytogenet, Jul. 2002, 136(1):10-16.
Wei et al., “Abstract #2136: Entrectinib is Effective Against the Gatekeeper and Other Emerging Resistance Mutations in NTRK-, ROSI- and Alk-Rearranged Cancers,” Poster, Presented at Proceedings of the 107th Annual Meeting of the American Association for Cancer Research, New Orleans LA, Apr. 16-20, 2016; Cancer Res, Jul. 2016, 76(14): 1 page.
Wei et al., “Abstract 78: Entrectinib, a highly potent pan-Trk, and ALK inhibitor, has broad-spectrum, histology-agnostic anti-tumor activity in molecularly defined cancers,” 28thEORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics, Munich, Germany, 2016, 1 page.
Weinstein, “Cancer. Addiction to oncogenes—the Achilles heal of cancer,” Science, Jul. 2002, 297(5578):63-64.
Wen et al, “Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group,” J Clin Oncol, Apr. 2010, 28(11): 1963-1972.
Wiesner et al., “Kinase fusions are frequent in Spitz tumours and spitzoid melanomas,” Nature Comm., 2014, 5:3116.
Winski et al., “LOXO-101, a pan-TRJ inhibitor, for the treatment of TRK-driven cancers,” 26th EORTC-NCI-AACR Symposium on Molecular Targets and Cancer Therapeutics, Barcelona, Spain, 2014, 175.
Wittwer et al., “High-resolution genotyping by amplicon melting analysis using LCGreen,” Clin Chem, Jun. 2003;49(6 Pt 1):853-60.
Wlodarska et al., “ALK-Positive Anaplastic Large Cell Lymphoma with the Variant EEF1G-, RNF213- and Atic-ALK Fusions Is Featured by Copy Number Gain of the Rearranged ALK Gene”, Blood, vol. 126(23): 3654, 57th Annual Meeting of the American Society of Hematology, San Diego, CA, 2015.
Won et al., “Post-crizotinib management of effective ceritinib therapy in a patient with ALK-positive non-small cell lung cancer”, BMC Cancer, 16: 568, 2016.
Wong et al., “Evaluation of a Congenital Infantile Fibrosarcoma by Comprehensive Genomic Profiling Reveals an LMNA-NTRK.1 Gene Fusion Responsive to Crizotinib,” J Natl Cancer Inst, Nov. 2016, 108(1) pii: div307.
Woodward, “Bi-allelic SNP genotyping using the TaqMan® assay,” Methods Mol Biol., 2014;1145:67-74. doi: 10.1007/978-1-4939-0446-4 6.
Woolf et al., “Letter to Neuroscience: Nerve Growth Factor Contributes to the Generation of Inflammatory Sensory Hypersensitivity,” Neuroscience, 1994, 62:327-331.
Wu et al., “The genomic landscape of diffuse intrinsic pontine glioma and pediatric non-brainstem high-grade glioma,” Nature Genetics, 2014, 444-450.
Wu et al., “The landscape of fusion transcripts in spitzoid melanoma and biologically indeterminate spitzoid tumors by RNA sequencing,” Modern Pathol., Apr. 2016, 29(4):359-369.
Xalkori, Highlights of Prescribing Information, Pfizer Labs, Initial approval 2011, revised Mar. 2016, 20 pages.
Yakirevich et al., “Colorectal Adenocarcinoma with ALK Rearrangement: Clinicopathologic and Molecular Characteristics”, Laboratory Investigation, vol. 96, Supp. Suppl. 1, pp. 209A, Abstract No. 827, 105th Annual Meeting of the United States and Canadian Academy of Pathology, Seattle, WA, 2016.
Yakirevich et al., “Oncogenic ALK Fusion in Rare and Aggressive Subtype of Colorectal Adenocarcinoma as a Potential Therapeutic Target.”, Clin Cancer Res, 22(15): 3831-40, 2016.
Yamamoto et al., “ALK, ROS1 and NTRK3 gene rearrangements in inflammatory myofibroblastic tumours.”, Histopathology, 69(1): 72-83, 2016.
Yamamoto et al., “Anaplastic lymphoma kinase-positive squamous cell carcinoma of the lung: A case report.”, Mal Clin. Oneal. 5(1): 61-63, 2016.
Yanai et al., “A rare case of bilateral stage IV adrenal neuroblastoma with multiple skin metastases in a neonate: diagnosis, management, and outcome,” J Pediatr. Surg., Dec. 2004, 39(12):1782-1783.
Yeh et al., “NTRK.3 kinase fusions in Spitz tumours,” J Pathol., Nov. 2016, 240(3): 282-290.
Yilmaz et al., “Theraputic targeting of Trk supresses tumor proliferation and enhances cisplatin activity inHNSCC,” Cancer Biology and Therapy, 2010, 10(6):644-653.
Ying et al., “Atypical negative ALK FISH accompanied by immunohistochemistry positivity harbored various ALK rearrangements in NSCLC patients and respond to targeted therapy.”, J Clin. Oncology, vol. 34, Supp. Supplement 15, Abstract No. e20506, 2016 Annual Meeting of the American Society of Clinical Oncology, Chicago, IL, 2016.
Yu et al., “Denaturing high performance liquid chromatography: high throughput mutation screening in familial hypertrophic cardiomyopathy and SNP genotyping in motor neurone disease,” J Clin Pathol, May 2005;58(5):479-85.
Yu et al., “Detection of ALK rearrangements in lung cancer patients using a homebrew PCR assay”, Oncotarget, 8(5): 7722-7728, 2016.
Yuzugullu et al., “NTRK.2 activation cooperates with PTEN deficiency in T-ALL through activation of both the PBK-AKT and JAK-STAT3 pathways,” Cell Discov., Sep. 2016, 2: 16030.
Zage et al., “The selective Trk inhibitor AZ623 inhibits brain-derived neurotrophic factor-mediated neuroblastoma cell proliferation and signaling and is synergistic with topotecan,” Cancer, Mar. 2011, 117(6): 1321-1391.
Zahn et al., “Effect of blockade of nerve growth factor and tumor necrosis factor on pain behaviors after plantar incision,” J. Pain, 2004, 5:157-163.
Zehir et al., “Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients,” Nat. Med, Jun. 2017, 23(6):703-713.
Zelboraf, Highlights of Prescribing Information, Genentech USA, Initial Approval 2011, revised Aug. 2015, 18 pages.
Zhang et al., “A novel multiplex tetra-primer ARMS-PCR for the simultaneous genotyping of six single nucleotide polymorphisms associated with female cancers,” PLoS One, Apr. 17, 2013;8(4):e62126. doi: 10.1371/iournal.pone.0062126. Print 2013.
Zhang et al., “Expression of nerve growth factor receptors and their prognostic value in human pancreatic cancer,” Oncology Reports, 2005, 14:161-171.
Zhang et al., “Novel Phenotypic and Genetic Analysis ofT-Cell Prolymphocytic Leukemia (T-PLL),” Blood, 2014, 124(21):1682.
Zhang et al., “Whole-genome sequencing identifies genetic alterations in pediatric low-grade gliomas,” Nat. Genet., Jun. 2013, 45(6): 602-612.
Zheng et al., “Anchored multiplex PCR for targeted next-generation sequencing,” Nature Med., Dec. 2014, 20(12):1479-1486.
Zhu et al., “TPD52L1-ROS1, a new ROSI fusion variant in lung adenosquamous cell carcinoma.identified by comprehensive genomic profiling”, Lung Cancer, 97:48-50, doi: 10.1016/j.lungcan.2016.04.013, 2012.
Ziemiecki et al., “Oncogenic activation of the human trk proto-oncogene by recombination with the ribosomal large subunit protein L7a,” EMBO J, Jan. 1990, 9(1):191-196.
Zou et al., “PF-06463922 is a potent and selective next-generation ROS1/ALK inhibitor capable of blocking crizotinib-resistant ROS1 mutations.”, Proc. Natl. Acad Sci. USA., 112(11): 3493-8, 2015.
U.S. Appl. No. 14/575,633, filed Dec. 18, 2014, Issued.
U.S. Appl. No. 15/41,839, filed Jan. 9, 2017, Issued.
Byrn, Stephen, et al. “Pharmaceutical solids: a strategic approach to regulatory considerations.” Pharmaceutical research 12.7 (1995): 945-954.
Ghilardi, Joseph R., et al. “Administration of a tropomyosin receptor kinase inhibitor attenuates sarcoma-induced nerve sprouting, neuroma formation and bone cancer pain.” Molecular pain 6 (2010). doi: 10.1186/1744-8069-6-87. 12 pages.
Lipska, Beata S., et al. “c. 1810C> T Polymorphism of NTRK1 Gene is associated with reduced Survival in Neuroblastoma Patients.” BMC cancer 9.1 (2009): 436.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2018/057542, dated May 7, 2020. 12 pages.
Perrigo Compounding Information for ORA-Blend® SF (Year: 2015). “ORA-BLEND® SF Flavoured Sugar-Free Oral Suspending Vehicle.” 4 pages.
Spectrum Pharmacy Products (2015) “Suggested Formula.” 3 pages.
Wood, Laura D., et al. “Somatic mutations of GUCY2F, EPHA3, and NTRK3 in human cancers.” Human mutation 27.10 (2006): 1060-1061.
Loftsson, Thorsteinn, and Marcus E. Brewster. “Pharmaceutical applications of cyclodextrins. 1. Drug solubilization and stabilization.” Journal of pharmaceutical sciences 85.10 (1996): 1017-1025.
Nagasubramanian et al., “Infantile Fibrosarcoma With NTRK3-ETV6 Fusion Successfully Treated With the Tropomyosin-Related Kinase Inhibitor LOXO-101,” Pediatr Blood Cancer., Aug. 2016, 63(8):1468-70.
PCT International Preliminary Report on Patentability in International Application No. PCT/US2019/024961, dated Oct. 8, 2020. 8 pages.
U.S. Appl. No. 13/125,263, filed Oct. 21, 2009, Issued.
U.S. Appl. No. 13/943,590, filed Jul. 16, 2013, Issued.
U.S. Appl. No. 14/490,460, filed Sep. 18, 2014, Issued.
U.S. Appl. No. 14/596,611, filed Jan. 14, 2015, Issued.
U.S. Appl. No. 14/846,166, filed Sep. 4, 2015, Issued.
U.S. Appl. No. 15/399,389, filed Jan. 5, 2017, Issued.
U.S. Appl. No. 15/860,948, filed Jan. 3, 2018, Issued.
U.S. Appl. No. 16/044,653, filed Jul. 25, 2018, Issued.
U.S. Appl. No. 17/020,461, filed Sep. 14, 2020, Published.
U.S. Appl. No. 14/943,014, filed Nov. 16, 2015, Issued.
U.S. Appl. No. 15/399,207, filed Jan. 5, 2017, Issued.
U.S. Appl. No. 15/706,062, filed Sep. 15, 2017, Issued.
U.S. Appl. No. 15/872,769, filed Jan. 16, 2018, Issued.
U.S. Appl. No. 16/366,368, filed Mar. 27, 2019, Issued.
U.S. Appl. No. 16/302,312, filed May 18, 2017, Published.
U.S. Appl. No. 15/579,007, filed Jun. 1, 2016, Published.
U.S. Appl. No. 15/6922,388, filed Apr. 4, 2017, Issued.
U.S. Appl. No. 15/861,017, filed Jan. 3, 2018, Issued.
U.S. Appl. No. 16/739,845, filed Jan. 10, 2020, Published.
U.S. Appl. No. 15/622,544, filed Apr. 4, 2017, Issued.
U.S. Appl. No. 16/199,739, filed Nov. 26, 2018, Issued.
U.S. Appl. No. 16/859,275, filed Apr. 27, 2020, Published.
U.S. Appl. No. 17/043,134, filed Sep. 29, 2020, Published.
U.S. Appl. No. 13/698,922, filed May 13, 2011, Issued.
U.S. Appl. No. 14/575,663, filed Dec. 18, 2014, Issued.
U.S. Appl. No. 15/350,888, filed Nov. 14, 2016, Issued.
U.S. Appl. No. 15/401,839, filed Jan. 9, 2017, Issued.
U.S. Appl. No. 15/632,187, filed Jun. 23, 2017, Issued.
U.S. Appl. No. 15/900,019, filed Feb. 20, 2018, Issued.
U.S. Appl. No. 15/401,952, filed Jan. 9, 2017, Issued.
U.S. Appl. No. 16/818,125, filed Mar. 13, 2020, Published.
U.S. Appl. No. 13/063,894, filed Sep. 21, 2009, Issued.
U.S. Appl. No. 13/614,968, filed Sep. 13, 2012, Issued.
U.S. Appl. No. 14/984,353, filed Dec. 30, 2015, Issued.
U.S. Appl. No. 15/401,792, filed Jan. 9, 2017, Issued.
U.S. Appl. No. 15/401,969, filed Jan. 9, 2017, Issued.
U.S. Appl. No. 16/025,281, filed Jul. 2, 2018, Issued.
U.S. Appl. No. 16/170,976, filed Oct. 25, 2018, Published.
U.S. Appl. No. 17/078,508, filed Oct. 23, 2020, Pending.
U.S. Appl. No. 15/335,378, filed Oct. 26, 2016, Issued.
U.S. Appl. No. 15/785,174, filed Oct. 16, 2017, Issued.
U.S. Appl. No. 15/785,218, filed Oct. 16, 2017, Issued.
U.S. Appl. No. 15/860,789, filed Jan. 3, 2018, Allowed.
U.S. Appl. No. 15/785,228, filed Oct. 16, 2017, Issued.
U.S. Appl. No. 17/163,930, filed Feb. 1, 2021, Pending.
U.S. Appl. No. 16/199,818, filed Nov. 26, 2018, Allowed.
U.S. Appl. No. 16/199,867, filed Nov. 26, 2018, Published.
U.S. Appl. No. 16/199,875, filed Nov. 26, 2018, Issued.
U.S. Appl. No. 17/074,746, filed Oct. 20, 2020, Pending.
U.S. Appl. No. 13/382,858, filed Jul. 9, 2010, Issued.
U.S. Appl. No. 14/321,246, filed Jul. 1, 2014, Issued.
U.S. Appl. No. 15/401,895, filed Jan. 9, 2017, Issued.
U.S. Appl. No. 15/401,913, filed Jan. 9, 2017, Issued.
U.S. Appl. No. 15/724,601, filed Oct. 4, 2017, Issued.
U.S. Appl. No. 16/377,514, filed Apr. 8, 2019, Issued.
Related Publications (1)
Number Date Country
20200216451 A1 Jul 2020 US
Provisional Applications (1)
Number Date Country
62524801 Jun 2017 US
Continuation in Parts (1)
Number Date Country
Parent PCT/US2016/058951 Oct 2016 US
Child 16345571 US