Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR PREPARING THE SAME
Document Type and Number:
WIPO Patent Application WO/2018/218100
Kind Code:
A1
Abstract:
The present disclosure relates to solid forms of N-{[2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihydro-1'H-spiro[cyclopropane-1,4'-[2,7]naphthyridin]-6'-yl]methyl}acrylamide, methods of preparation thereof, and intermediates in the preparation thereof, which are useful in the treatment of the FGFR-associated or mediated diseases such as cancer.

Inventors:
PAN YONGCHUN (US)
SHI CHONGSHENG ERIC (US)
TAO MING (US)
HAN WAYNE (US)
XIA MICHAEL (US)
WANG DENGJIN (US)
JIA ZHONGJIANG (US)
ZHOU JIACHENG (US)
LI QUN (US)
Application Number:
PCT/US2018/034559
Publication Date:
November 29, 2018
Filing Date:
May 25, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
INCYTE CORP (US)
International Classes:
C07D471/04; A61K31/438; A61P35/00
Domestic Patent References:
WO2016134314A12016-08-25
Foreign References:
US20120165305A12012-06-28
US20140045814A12014-02-13
US20130338134A12013-12-19
US20140171405A12014-06-19
US20140315902A12014-10-23
US20160115164A12016-04-28
US20160244448A12016-08-25
US20160244449A12016-08-25
US20160244450A12016-08-25
US20170107216A12017-04-20
US20170145025A12017-05-25
US20170174671A12017-06-22
US20170174679A12017-06-22
US20170320875A12017-11-09
US20170342060A12017-11-30
US20170362253A12017-12-21
US20180016260A12018-01-18
Other References:
ESWARAKUMAR ET AL., CYTOKINE & GROWTH FACTOR REVIEWS, 2005
KNIGHTS; COOK, PHARMACOLOGY & THERAPEUTICS, 2010
TURNER; GROSE, NATURE REVIEWS CANCER, 2010
HEINZLE ET AL., CUR. PHARM. DES., vol. 20, 2014, pages 2881
TAYLOR ET AL., JCI, vol. 119, 2009, pages 3395
WUTS; GREENE: "Greene's Protective Groups in Organic Synthesis", 2006, JOHN WILEY & SONS
"Remington: The Science and Practice of Pharmacy", 2005, LIPPINCOTT WILLIAMS & WILKINS
"Handbook of Pharmaceutical Excipients", 2009, THE PHARMACEUTICAL PRESS AND THE AMERICAN PHARMACEUTICAL ASSOCIATION
"Handbook of Pharmaceutical Additives", 2007, GOWER PUBLISHING COMPANY
"Pharmaceutical Preformulation and Formulation", 2009, CRC PRESS LLC
"Physicians' Desk Reference", 1996, MEDICAL ECONOMICS COMPANY
FRENCH ET AL.: "Targeting FGFR4 Inihibits Hepatocellular Carcinoma in Preclinical Mouse Models", PLOS ONE, vol. 7, no. 5, May 2012 (2012-05-01), pages e36713, XP055028245, DOI: doi:10.1371/journal.pone.0036713
Attorney, Agent or Firm:
KING, Annie J. et al. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A solid form of Compound 1 having the formula:

Compound 1 ), or a salt thereof.

2. The solid form of claim 1 wherein the solid form is crystalline.

3. The solid form of claim 2 having Form I.

4. The solid form of claim 3 having one or more characteristic XRPD peaks selected from about 8.1, about 9.0, and about 12.3 degrees 2-theta.

5. The solid form of claim 3 having at least one characteristic XRPD peaks selected from about 8.1, about 9.0, about 12.3, about 16.0, about 18.0, and about 23.3 degrees 2-theta.

6. The solid form of claim 3 having at least two characteristic XRPD peaks selected from about 8.1, about 9.0, about 11.5, about 12.3, about 15.1 , about 16.0, about 18.0, about 19.6, about 20.0, about 20.4, about 21.0, about 23.3, about 24.2, about 24.7, and about 27.1 degrees 2-theta.

7. The solid form of claim 3 having at least three characteristic XRPD peaks selected from about 8.1, about 9.0, about 11.5, about 12.3, about 15.1 , about 16.0, about 18.0, about 19.6, about 20.0, about 20.4, about 21.0, about 23.3, about 24.2, about 24.7, and about 27.1 degrees 2-theta.

8. The solid form of any one of claims 3-7 having an XRPD pattern with characteristic peaks as substantially shown in Figure 1.

9. The solid form of any one of claims 3-8 exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C

10. The solid form of any one of claims 3-8 having a DSC thermogram substantially as depicted in Figure 2.

1 1. The solid form of any one of claims 3-10 having a TGA thermogram substantially as depicted in Figure 3.

12. The solid form of claim 2 having Form II, Form III, Form IV, Form V, Form VI, Form VII, Form VIII, Form IX, Form IXa, Form X, or Form XI.

13. A pharmaceutical composition comprising a solid form of any one of claims 1 -12, and one or more pharmaceutically acceptable carriers or excipients.

14. A method of inhibiting an FGFR4 enzyme comprising contacting said enzyme with a solid form of any one of claims 1 -12, or a composition of claim 13.

15. A method of treating cancer in a patient comprising administering to said patient a therapeutically effective amount of a solid form of any one of claims 1 -12, or a composition of claim 13.

16. A method of treating cancer in a patient comprising administering to said patient a therapeutically effective amount of a solid form of any one of claims 1 -12, or a composition of claim 13, in combination with another therapy or therapeutic agent.

17. The method of claim 15 or 16, wherein said cancer is selected from hepatocellular cancer, bladder cancer, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, gastric cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, prostate cancer, esophageal cancer, gall bladder cancer, pancreatic cancer, thyroid cancer, skin cancer, leukemia, multiple myeloma, chronic lymphocytic lymphoma, adult T cell leukemia, B-cell lymphoma, acute myelogenous leukemia, Hodgkin's or non-Hodgkin' s lymphoma, Waldenstrom's Macroglubulinemia, hairy cell lymphoma, Burkett's lymphoma, glioblastoma, melanoma, and rhabdo sarcoma.

18. The method of claiml5 or 16, wherein said cancer is selected from hepatocellular cancer, breast cancer, bladder cancer, colorectal cancer, melanoma, mesothelioma, lung cancer, prostate cancer, pancreatic cancer, testicular cancer, thyroid cancer, squamous cell carcinoma, glioblastoma, neuroblastoma, uterine cancer, and rhabdosarcoma.

19. A process for preparing Form I of Compound 1 : O (Compound 1), comprising dissolving Compound 1 in a solvent to form a solution; and isolating Form I from the solution.

20. The process of claim 19, wherein the solvent comprises acetone, water, or a mixture thereof.

21. A process for preparing Compound 1 :

O (Compound 1),

or a salt thereof, comprising:

converting Compound 8:

(Compound 8) to Compound 9:

converting Compound 9 to Compound 10: (Compound 10);

converting Compound 10 to Compound 11 :

(Compound 1 1);

converting Compound 1 1 to Compound 12 diacetate:

2 HOAc (Compound 12 diacetate); converting Compound 12 diacetate to Compound 12 hydrochloride:

(Compound 12 hydrochloride); and converting Compound 12 hydrochloride to Compound 1.

22. A process for preparing Compound 1 : (Compound 1), or a salt thereof, comprising converting Compound 12 or a salt thereof to Compound 1. 23. The process of claim 22, wherein the salt of Compound 12 is Compound 12 hydrochloride:

(Compound 12 hydrochloride).

24. The process of claim 21 or 23, wherein the conversion of Compound 12

hydrochloride to Compound 1 comprises reacting Compound 12 hydrochloride with acryloyl chloride in the presence of Bl and S I , wherein Bl is a base and SI is a solvent.

25. The process of claim 24, wherein Bl is an alkali metal hydroxide base.

26. The process of claim 24 or 25, wherein S I comprises a halogenated solvent.

27. The process of any one of claims 21 and 23-26, wherein the conversion of Compound 12 hydrochloride to Compound 1 is carried out at a temperature of about 30 °C or lower.

28. The process of any one of claims 21 and 23-27 wherein Compound 12 hydrochloride is prepared by a process comprising converting Compound 12 diacetate:

• 2 HOAc (Compound 12 diacetate)

to Compound 12 hydrochloride.

29. The process of claim 28, wherein the conversion of Compound 12 diacetate to Compound 12 hydrochloride comprises reacting Compound 12 diacetate with B2 and hydrochloric acid in the presence of S2, wherein B2 is a hydroxide base and S2 is a solvent.

30. The process of claim 29, wherein B2 is an alkali metal hydroxide base.

31. The process of claim 29 or 30, wherein S2 comprises a halogenated solvent, protic solvent, or a mixture thereof.

32. The process of any one of claims 21 and 28-31 wherein Compound 12 diacetate is prepared by a process comprising converting Compound 11 :

(Compound 11) to Compound 12 diacetate.

33. The process of claim 32, wherein the conversion of Compound 11 to Compound 12 diacetate comprises reacting Compound 11 with acetic acid and zinc.

34. The process of any one of claims 21, 32 and 33, wherein Compound 11 is prepared by a process comprising converting Compound 10:

(Compound 10) to Compound 11.

35. The process of claim 34 wherein the conversion of Compound 10 to Compound 11 comprises reacting Compound 11 with hydroxylamine hydrochloride in the presence of S3, wherein S3 is a solvent.

36. The process of claim 35, wherein S3 comprises a protic solvent, a basic solvent, or a mixture thereof.

37. The process of any one of claims 21 and 34-36, wherein Compound 10 is prepared by a process comprising converting Compound 9: to Compound 10.

38. The process of claim 37 wherein the conversion of Compound 9 to Compound 10 comprises reacting Compound 10 with sodium periodate and osmium tetroxide in the presence of S4, wherein S4 is a solvent.

39. The process of claim 38, wherein the conversion of Compound 9 to Compound 10 further comprises B4, wherein B4 is a base.

40. The process of claim 39, wherein B4 is an alkali metal bicarbonate base.

41. The process of any one of claims 38-40, wherein S4 comprises an ether solvent, a protic solvent, an aprotic solvent, or a mixture thereof.

42. The process of any one of claims 21 and 37-41 , wherein Compound 9 is prepared by a process comprising converting Compound 8 :

(Compound 8) to Compound 9.

43. The process of claim 42 wherein the conversion of Compound 8 to Compound 9 comprises reacting Compound 8 with 4,4,5, 5-tetramethyl-2-vinyl-l ,3,2-dioxaborolane, PI , and B5 in the presence of S5, wherein PI is a transition metal catalyst, B5 is a base, and S5 is a solvent.

44. The process of claim 43, wherein P I is a palladium catalyst.

45. The process of claim 43, wherein B5 is cesium fluoride.

46. The process of any one of claims 43-45, wherein S5 comprises a protic solvent, an ether solvent, or a mixture thereof.

47. The process of any one of claims 21 and 23-27 wherein Compound 12 hydrochloride is prepared by a process comprising converting Compound 15:

to Compound 12 hydrochloride.

48. The process of claim 47 wherein the conversion of Compound 15 to Compound 12 hydrochloride comprises reacting Compound 15 with hydrochloric acid in the presence of S6, wherein S6 is a solvent.

49. The process of claim 48, wherein S6 comprises an aprotic solvent, an ether solvent, or a mixture thereof.

50. The process of any one of claims 47-49 wherein Compound 15 is prepared by a process comprising converting Compound 8:

(Compound 8)

to Compound 15.

51. The process of claim 50 wherein the conversion of Compound 8 to Compound 15 comprises reacting Compound 8 with potassium N-Boc-aminomethyltrifluoroborate, P2, and B7, wherein P2 is a transitional metal catalyst and B7 is base.

52. The process of claim 51, wherein P2 is a palladium catalyst.

53. The process of claim 51 or 52, wherein B7 is a carbonate base.

54. The process of any one of claims 50-53, wherein the conversion of Compound 8 to Compound 15 is carried out in S7, wherein S7 is a solvent.

55. The process of claim 54, wherein S7 comprises a protic solvent, an ether solvent, or a mixture thereof.

56. The process of any one of claims 47-49 wherein Compound 15 is prepared by a process comprising converting Compound 12 diacetate:

(Compound 12 diacetate) to Compound 15.

57. The process of any one of claims 56 wherein the conversion of Compound 12 diacetate to Compound 15 comprises reacting Compound 12 diacetate with di-tert- butyldi carbonate with B8.

58. The process of claim 57, wherein B8 is an alkali metal hydroxide base.

59. The process of any one of claims 56-58, wherein the conversion of Compound 12 diacetate to Compound 15 is carried out in S8, wherein S8 is a solvent.

60. The process of claim 59, wherein S8 comprises a protic solvent, an ether solvent, or a mixture thereof.

61. A compound which is selected from:

(Compound 10), and (Compound 11),

salt thereof.

A compound which is selected from:

2 HOAc (Compound 12 diacetate) and

(Compound 12 hydrochloride).

Description:
CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR

PREPARING THE SAME

FIELD OF THE INVENTION

This application relates to crystalline forms of a Fibroblast Growth Factor Receptors

(FGFR) inhibitor, including methods of preparation thereof, and intermediates in the preparation thereof, which are useful in the treatment of FGFR mediated disease such as cancer. BACKGROUND OF THE INVENTION

The Fibroblast Growth Factor Receptors (FGFR) are receptor tyrosine kinases that bind to fibroblast growth factor (FGF) ligands. There are four FGFR proteins (FGFR1-4) that are capable of binding ligands and are involved in the regulation of many physiological processes including tissue development, angiogenesis, wound healing, and metabolic regulation. Upon ligand binding, the receptors undergo dimerization and phosphorylation leading to stimulation of the protein kinase activity and recruitment of many intracellular docking proteins. These interactions facilitate the activation of an array of intracellular signaling pathways including Ras-MAPK, AKT-PI3K, and phospholipase C that are important for cellular growth, proliferation and survival (Reviewed in Eswarakumar et al. Cytokine & Growth Factor Reviews, 2005). Aberrant activation of this pathway either through overexpression of FGF ligands or FGFR or activating mutations in the FGFRs can lead to tumor development, progression, and resistance to conventional cancer therapies. In human cancer, genetic alterations including gene amplification, chromosomal translocations and somatic mutations that lead to ligand-independent receptor activation have been described. Large scale DNA sequencing of thousands of tumor samples has revealed that components of the FGFR pathway are among the most frequently mutated in human cancer. Many of these activating mutations are identical to germline mutations that lead to skeletal dysplasia syndromes. Mechanisms that lead to aberrant ligand-dependent signaling in human disease include overexpression of FGFs and changes in FGFR splicing that lead to receptors with more promiscuous ligand binding abilities (Reviewed in Knights and Cook Pharmacology & Therapeutics, 2010; Turner and Grose, Nature Reviews Cancer, 2010). Therefore, development of inhibitors targeting FGFR may be useful in the clinical treatment of diseases that have elevated FGF or FGFR activity. The cancer types in which FGF/FGFRs are implicated include, but are not limited to: carcinomas (e.g., bladder, breast, cervical, colorectal, endometrial, gastric, head and neck, kidney, liver, lung, ovarian, prostate); hematopoietic malignancies (e.g., multiple myeloma, chronic lymphocytic lymphoma, adult T cell leukemia, acute myelogenous leukemia, non- Hodgkin lymphoma, myeloproliferative neoplasms, and Waldenstrom's Macroglubulinemia); and other neoplasms (e.g., glioblastoma, melanoma, and rhabdosarcoma). In addition to a role in oncogenic neoplasms, FGFR activation has also been implicated in skeletal and chondrocyte disorders including, but not limited to, achrondroplasia and craniosynostosis syndromes.

The FGFR4-FGF19 signaling axis, specifically, has been implicated in the pathogenesis of a number of cancers including hepatocellular carcinoma (Heinzle et al., Cur. Pharm. Des. 2014, 20:2881). Ectopic expression of FGF19 in transgenic mice was shown to lead to tumor formation in the liver and a neutralizing antibody to FGF19 was found to inhibit tumor growth in mice. In addition, overexpression of FGFR4 has been observed in a multiple tumor types including hepatocellular carcinoma, colorectal, breast, pancreatic, prostate, lung, and thyroid cancers. Furthermore, activating mutations in FGFR4 have been reported in rhabdomyosarcoma (Taylor et al. JCI 2009,119:3395).

Inhibitors of FGFR are currently being developed for the treatment of cancer. For example, the molecule N-{[2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihyd ro- H- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-6'-yl]methyl}acry lamide and other small molecule inhibitors of FGFR are reported in e.g., US Publication Nos. : 2012/0165305; 2014- 0045814; 2013-0338134; 2014/0171405; 2014/0315902; 2016/0115164; 2016/0244448; 2016/0244449; and 2016-0244450. Accordingly, there is a need for new forms of FGFR- inhibiting molecules for preparing pharmaceutically useful formulations and dosage forms with suitable properties related to, for example, facilitating the manufacture of safe, effective, and high quality drug products.

SUMMARY OF THE INVENTION

Provided herein are solid forms of N-{[2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo- 2',3'-dihydro-lΉ-spiro[cyclopropane-l,4'-[2,7]naphthyridin] -6'-yl]methyl} acrylamide, or salts thereof.

Provided herein are also pharmaceutical compositions, which include the solid forms (e.g., crystalline forms) as described herein, and one or more pharmaceutically acceptable carriers or excipients. The present disclosure also provides methods of inhibiting FGFR4 enzyme using the solid forms (e.g., crystalline forms) as described herein.

The present disclosure also provides therapeutic methods of using the solid forms (e.g., crystalline forms) as described herein.

Provided herein are also processes for preparing N-{[2'-(2,6-difluoro-3,5- dimethoxyphenyl)-3'-oxo-2',3'-dihydro- H-spiro[cyclopropane-l,4'-[2,7]naphthyridin]-6'- yl]methyl} acrylamide and its solid forms (e.g., crystalline forms) as described herein.

Provided herein are also intermediates useful for the preparation of N-{[2'-(2,6- difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihydro- H-spiro[cyclopropane-l,4'- [2,7]naphthyridin]-6'-yl]methyl}acrylamide and its solid forms (e.g., crystalline forms) described herein.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 shows an X-ray powder diffraction (XRPD) pattern of Compound 1, Form I. FIG. 2 shows a differential scanning calorimetry (DSC) thermogram of Compound 1,

Form I.

FIG. 3 shows a thermogravimetric analysis (TGA) thermogram of Compound 1, Form

I.

FIG. 4 shows an XRPD pattern of Compound 1 , Form II.

FIG. 5 shows a DSC thermogram of Compound 1, Form II.

FIG. 6 shows an XRPD pattern of Compound 1 , Form III.

FIG. 7 shows a DSC thermogram of Compound 1, Form III.

FIG. 8 shows an XRPD pattern of Compound 1 , Form IV.

FIG. 9 shows a DSC thermogram of Compound 1, Form IV.

FIG. 10 shows an XRPD pattern of Compound 1, Form V.

FIG. 11 shows a DSC thermogram of Compound 1, Form V.

FIG. 12 shows a TGA thermogram of Compound 1, Form V.

FIG. 13 shows an XRPD partem of Compound 1, Form VI.

FIG. 14 shows an XRPD pattern of Compound 1, Form VII.

FIG. 15 shows a DSC thermogram of Compound 1, Form VII.

FIG. 16 shows a TGA thermogram of Compound 1, Form VII.

FIG. 17 shows an XRPD pattern of Compound 1, Form VIII.

FIG. 18 shows a DSC thermogram of Compound 1, Form VIII.

FIG. 19 shows a TGA thermogram of Compound 1, Form VIII. FIG. 20 shows an XRPD pattern of Compound 1, Form IX.

FIG. 21 shows a DSC thermogram of Compound 1, Form IX.

FIG. 22 shows a TGA thermogram of Compound 1, Form IX.

FIG. 23 shows an XRPD pattern of Compound 1, Form IXa.

FIG. 24 shows a DSC thermogram of Compound 1, Form IXa.

FIG. 25 shows a TGA thermogram of Compound 1, Form IXa.

FIG. 26 shows an XRPD pattern of Compound 1, Form X.

FIG. 27 shows an XRPD pattern of Compound 1, Form XI.

FIG. 28 shows a DSC thermogram of Compound 1, Form XI.

FIG. 29 shows a TGA thermogram of Compound 1, Form XI.

DETAILED DESCRIPTION

The present disclosure is directed to, inter alia, solid forms of N-((2'-(2,6-difluoro- 3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihydro- H-spiro[cyclopropane-l ,4'-[2,7]naphthyridine]- 6'-yl)methyl)acrylamide (Compound 1), or a salt thereof, the structure of which is shown below.

Compound 1

Compound 1 is described in US 2016/0244448, the entirety of which is incorporated herein by reference.

Compound 1 can be isolated as one or more solid forms. The solid forms (e.g., crystalline forms) described herein have many advantages, for example they have desirable properties, such as ease of handling, ease of processing, storage stability, and ease of purification. Moreover, the crystalline forms can be useful for improving the performance characteristics of a pharmaceutical product such as dissolution profile, shelf-life and bioavailability

As used herein, and unless otherwise specified, the term "about", when used in connection with a numeric value or range of values which is provided to describe a particular solid form (e.g., a specific temperature or temperature range, such as describing a melting, dehydration, or glass transition; a mass change, such as a mass change as a function of temperature or humidity; a solvent or water content, in terms of, for example, mass or a percentage; or a peak position, such as in analysis by, for example, 13 C NMR, DSC, TGA and XRPD), indicate that the value or range of values may deviate to an extent deemed reasonable to one of ordinary skill in the art while still describing the particular solid form. Specifically, the term "about", when used in this context, indicates that the numeric value or range of values may vary by 5%, 4%, 3%, 2%, 1 %, 0.9%, 0.8%, 0.7%, 0.6%, 0.5%, 0.4%, 0.3%, 0.2% or 0.1% of the recited value or range of values while still describing the particular solid form. The term "about", when used in reference to a degree 2-theta value refers to +/-0.3 degrees 2-theta or +/-0.2 degrees 2-theta.

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

As used herein, the term "peak" or "characteristic peak" refers to a reflection having a relative height/intensity of at least about 3% of the maximum peak height/intensity.

As used herein, the term "crystalline" or "crystalline form" refers to a crystalline solid form of a chemical compound, including, but not limited to, a single-component or multiple- component crystal form, e.g., including solvates, hydrates, clathrates, and a co-crystal. For example, crystalline means having a regularly repeating and/or ordered arrangement of molecules, and possessing a distinguishable crystal lattice. The term "crystalline form" is meant to refer to a certain lattice configuration of a crystalline substance. Different crystalline forms of the same substance typically have different crystalline lattices (e.g., unit cells), typically have different physical properties attributed to their different crystalline lattices, and in some instances, have different water or solvent content. The different crystalline lattices can be identified by solid state characterization methods such as by X-ray powder diffraction (XRPD). Other characterization methods such as differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), dynamic vapor sorption (DVS), and the like further help identify the crystalline form as well as help determine stability and solvent/ water content.

Different crystalline forms of a particular substance, such as Compound 1 as described herein, can include both anhydrous forms of that substance and solvated/hydrated forms of that substance, where each of the anhydrous forms and solvated/hydrated forms are distinguished from each other by different XRPD patterns, or other solid state

characterization methods, thereby signifying different crystalline lattices. In some instances, a single crystalline form (e.g., identified by a unique XRPD pattern) can have variable water or solvent content, where the lattice remains substantially unchanged (as does the XRPD pattern) despite the compositional variation with respect to water and/or solvent.

An 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, inter alia, the sample preparation technique, crystal size distribution, 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 the machine 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 3% or at least about 4% of the maximum peak

height/intensity. Moreover, instrument variation and other factors can affect the 2-theta values. Thus, peak assignments, such as those reported herein, can vary by plus or minus about 0.2° (2-theta) or about 0.3° (2-theta), and the term "substantially" as used in the context of XRPD herein is meant to encompass the above-mentioned variations.

In the same way, temperature readings in connection with DSC, TGA, or other thermal experiments can vary about ±3 °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.

Crystalline forms of a substance can be obtained by a number of methods, as known in the art. Such methods include, but are not limited to, melt recrystallization, melt cooling, solvent recrystallization, recrystallization in confined spaces such as, e.g., in nanopores or capillaries, recrystallization on surfaces or templates such as, e.g., on polymers,

recrystallization in the presence of additives, such as, e.g., co-crystal counter-molecules, desolvation, dehydration, rapid evaporation, rapid cooling, slow cooling, vapor diffusion, sublimation, exposure to moisture, grinding and solvent-drop grinding.

As used herein, the term "amorphous" or "amorphous form" is intended to mean that the substance, component, or product in question is not crystalline as determined, for instance, by XRPD or where the substance, component, or product in question, for example is not birefringent when viewed microscopically. For example, amorphous means essentially without regularly repeating arrangement of molecules or lacks the long range order of a crystal, i.e., amorphous form is non-crystalline. An amorphous form does not display a defined x-ray diffraction pattern with sharp maxima. In certain embodiments, a sample comprising an amorphous form of a substance may be substantially free of other amorphous forms and/or crystalline forms. For example, an amorphous substance can be identified by an XRPD spectrum having an absence of reflections.

As used herein, the term "substantially amorphous" means a majority of the weight of a sample or preparation of Compound 1 is amorphous and the remainder of the sample is a crystalline form of the same compound. In some embodiments, a substantially amorphous sample has less than about 5% crystallinity (e.g., about 95% of the non-crystalline form of the same compound), less than about 4% crystallinity (e.g., about 96% of the non-crystalline form of the same compound), less than about 3% crystallinity (e.g., about 97% of the noncrystalline form of the same compound), less than about 2% crystallinity (e.g., about 98% of the non-crystalline form of the same compound), less than about 1% crystallinity (e.g., about 99% of the non-crystalline form of the same compound), or about 0% crystallinity (e.g., about 100% of the non-crystalline form of the same compound). In some embodiments, the term "fully amorphous" means less than about 99% or about 0% crystallinity.

Compound 1 can be prepared in batches referred to as batches, samples, or preparations. The batches, samples, or preparations can include Compound 1 in any of the crystalline or non-crystalline forms described herein, including hydrated and non-hydrated forms, and mixtures thereof.

Compounds provided herein (e.g., Compound 1) can also include all isotopes of atoms occurring in the intermediates or final compounds. Isotopes include those atoms having the same atomic number but different mass numbers. For example, isotopes of hydrogen include tritium and deuterium. One or more constituent atoms of the compounds provided herein can be replaced or substituted with isotopes of the atoms in natural or non- natural abundance. In some embodiments, the compound includes at least one deuterium atom. For example, one or more hydrogen atoms in a compound of the present disclosure can be replaced or substituted by deuterium. In some embodiments, the compound includes two or more deuterium atoms. In some embodiments, the compound includes 1, 2, 3, 4, 5, 6, 7 or 8 deuterium atoms. Synthetic methods for including isotopes into organic compounds are known in the art.

In some embodiments, Compound 1 is substantially isolated. The term "substantially isolated" is meant that the compound 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, salts, hydrates, solvates, or solid forms provided herein. 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, salts, hydrates, solvates, or solid forms provided herein.

The term "hydrate," as used herein, is meant to refer to a solid form of Compound 1 that includes water. The water in a hydrate can be present in a stoichiometric amount with respect to the amount of salt in the solid, or can be present in varying amounts, such as can be found in connection with channel hydrates.

As used herein, the term "substantially" when referring to a characteristic figure of a crystal form, such as an XRPD pattern, a DSC thermogram, a TGA thermogram, or the like, means that a subject figure may be non-identical to the reference depicted herein, but it falls within the limits of experimental error and thus may be deemed as derived from the same crystal form as disclosed herein, as judged by a person of ordinary skill in the art.

As used herein, the term "substantially crystalline," means a majority of the weight of a sample or preparation of Compound 1 is crystalline and the remainder of the sample is a non-crystalline form (e.g., amorphous form) of the same compound. In some embodiments, a substantially crystalline sample has at least about 95% crystallinity (e.g., about 5% of the non-crystalline form of the same compound), at least about 96% crystallinity (e.g., about 4% of the non-crystalline form of the same compound), at least about 97% crystallinity (e.g., about 3% of the non-crystalline form of the same compound), at least about 98% crystallinity (e.g., about 2% of the non-crystalline form of the same compound), at least about 99% crystallinity (e.g., about 1% of the non-crystalline form of the same compound), or about 100% crystallinity (e.g., about 0% of the non-crystalline form of the same compound). In some embodiments, the term "fully crystalline" means at least about 99% or about 100% crystallinity.

As used herein, the term "% crystallinity" or "crystalline purity," means percentage of a crystalline form in a preparation or sample which may contain other forms such as an amorphous form of the same compound, or at least one other crystalline form of the compound, or mixtures thereof. In some embodiments, the crystalline forms can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, the crystalline forms can be isolated with a purity greater than about 99%. As used herein, the term "reacting" is used as known in the art and generally refers to the bringing together of chemical reagents in such a manner so as to allow their interaction at the molecular level to achieve a chemical or physical transformation. In some embodiments, the reacting involves at least two reagents, wherein one or more equivalents of second reagent are used with respect to the first reagent. In some embodiments, the reacting step of a synthetic process may involve one or more substances in addition to the reagents such as solvent and/or a catalyst. The reacting steps of the processes described herein can be conducted for a time and under conditions suitable for preparing the identified product.

As used herein, the terms "converting" with respect to changing an intermediate or starting reagent or material in a chemical reaction refers to subjecting the intermediate or starting reagent or material to the suitable reagents and conditions (e.g., temperature, time, solvent, etc.) to effect certain changes (e.g. , breaking or formation of a chemical bond) to generate the desired product.

Compound 1 can be prepared in various solid forms including, e.g., Forms I, Form II, Form III, Form IV, Form V, Form VI, Form VII, Form VIII, Form IX, Form IXa, Form X, and Form XI.

Compound 1 Form I

Provided herein is a solid form of Compound 1 having Form I, which is described below in the Examples. In some embodiments, Form I has one or more characteristic XRPD peaks selected from about 8.1 , about 9.0, and about 12.3 degrees 2-theta

In some embodiments, Form I has at least one characteristic XRPD peaks selected from about 8.1, about 9.0, about 12.3, about 16.0, about 18.0, and about 23.3 degrees 2-theta.

In some embodiments, Form I has at least two characteristic XRPD peaks selected from about 8.1, about 9.0, about 11.5, about 12.3, about 15.1 , about 16.0, about 18.0, about 19.6, about 20.0, about 20.4, about 21.0, about 23.3, about 24.2, about 24.7, and about 27.1 degrees 2-theta.

In some embodiments, Form I has at least three characteristic XRPD peaks selected from about 8.1, about 9.0, about 11.5, about 12.3, about 15.1 , about 16.0, about 18.0, about 19.6, about 20.0, about 20.4, about 21.0, about 23.3, about 24.2, about 24.7, and about 27.1 degrees 2-theta.

In some embodiments, Form I has an XRPD partem with characteristic peaks as substantially shown in Figure 1. In some embodiments, Form I exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form I has a DSC thermogram substantially as depicted in Figure 2. In some embodiments, Form I has a TGA thermogram substantially as depicted in Figure 3.

In some embodiments, Form I has one or more characteristic XRPD peaks selected from about 8.1, about 9.0, and about 12.3 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form I has at least one characteristic XRPD peaks selected from about 8.1 , about 9.0, about 12.3, about 16.0, about 18.0, and about 23.3 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form I has an XRPD partem substantially as depicted in FIG. 1 and a DSC thermogram substantially as depicted in FIG. 2.

Provided herein are also processes for preparing Form I of Compound 1 comprising dissolving Compound 1 in a solvent to form a solution; and isolating Form I from the solution. The isolating can include precipitating or crystallizing Compound 1 from the solution to produce Form I.

In some embodiments, the isolating is carried out by (1) reducing the temperature of the solution of Compound 1, (2) concentrating the solution of Compound 1 , or (3) a combination thereof.

Provided herein are also processes for preparing Form I of Compound 1 comprising mixing Compound 1 with a solvent to generate Form I. The process can further comprise isolating Form I. In some embodiments, the mixing includes stirring a mixture comprising compound 1 and the solvent. In some embodiments, the isolating comprises filtrating the mixture to obtain Form I.

In some embodiments, the solvent comprises acetone, water, or a mixture thereof.

In some embodiments:

the dissolving comprises heating the solution of Compound 1 to a temperature of about 40 °C to about 60 °C; and

the isolating comprises reducing the volume of the solution of Compound 1 to form a reduced-volume solution of Compound 1 ; and cooling the reduced-volume solution of Compound 1 to precipitate Form I. For example, the solution can be cooled to 22 °C.

In some embodiments: the dissolving comprises heating the solution of Compound 1 to a temperature of about 40 °C to about 60 °C, wherein the solution comprises acetone and water as solvent; and the isolating comprises reducing the volume of the solution of Compound 1 to form a reduced-volume solution of Compound 1 ; and cooling the reduced-volume solution of Compound 1 to precipitate Form I.

In some embodiments, Form I can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form I can be isolated with a purity greater than about 99%. Compound 1 Form II

Provided herein is a solid form of Compound 1 having Form II, which is described below in the Examples. In some embodiments, Form II has one or more characteristic XRPD peaks selected from about 11.4, about 12.6, about 14.7, and about 16.1 degrees 2-theta.

In some embodiments, Form II has at least one characteristic XRPD peaks selected from about 11.4, about 12.6, about 14.7, about 16.1, about 18.3, about 21.2, and about 24.8 degrees 2-theta.

In some embodiments, Form II has at least two characteristic XRPD peaks selected from about 11.4, about 12.6, about 14.5, about 14.7, about 16.1, about 18.3, about 21.2, about 24.8, about 27.9, and about 28.3 degrees 2-theta.

In some embodiments, Form II has at least three characteristic XRPD peaks selected from about 11.4, about 12.6, about 14.5, about 14.7, about 16.1, about 18.3, about 21.2, about 24.8, about 27.9, and about 28.3 degrees 2-theta.

In some embodiments, Form II has an XRPD pattern with characteristic peaks as substantially shown in Figure 4.

In some embodiments, Form II exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form II has a DSC

thermogram substantially as depicted in Figure 5. In some embodiments, Form II has a TGA thermogram substantially as depicted in Figure 6.

In some embodiments, Form II has one or move characteristic XRPD peaks selected from about 11.4, about 12.6, about 14.7, and about 16.1 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form II has at least one characteristic XRPD peaks selected from about 11.4, about 12.6, about 14.7, about 16.1, about 18.3, about 21.2, and about 24.8 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form II has an XRPD pattern substantially as depicted in FIG. 4 and a DSC thermogram substantially as depicted in FIG. 5.

In some embodiments, Form II can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form II can be isolated with a purity greater than about 99%.

Compound 1 Form III

Provided herein is a solid form of Compound 1 having Form III, which is described below in the Examples. In some embodiments, Form III has one or more characteristic XRPD peaks selected from about 4.2, about 7.6, and about 15.2 degrees 2-theta

In some embodiments, Form III has at least one characteristic XRPD peak selected from about 4.2, about 7.6, about 13.2, about 15.2, about 19.3, and about 20.7 degrees 2-theta.

In some embodiments, Form III has at least two characteristic XRPD peaks selected from about 4.2, about 7.6, about 8.0, about 12.5, about 13.2, about 15.2, about 16.0, about 19.3, about 20.7, and about 25.6 degrees 2-theta.

In some embodiments, Form III has at least three characteristic XRPD peaks selected from about 4.2, about 7.6, about 8.0, about 12.5, about 13.2, about 15.2, about 16.0, about 19.3, about 20.7, and about 25.6 degrees 2-theta.

In some embodiments, Form III has an XRPD pattern with characteristic peaks as substantially shown in Figure 6.

In some embodiments, Form III exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C. In some embodiments, Form III has a DSC thermogram substantially as depicted in Figure 7.

In some embodiments, Form III has one or more characteristic XRPD peak selected from about 4.2, about 7.6, and about 15.2 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C.

In some embodiments, Form III has at least one characteristic XRPD peaks selected from about 4.2, about 7.6, about 13.2, about 15.2, about 19.3, and about 20.7 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C.

In some embodiments, Form III has an XRPD pattern substantially as depicted in FIG. 6 and a DSC thermogram substantially as depicted in FIG. 7. In some embodiments, Form III can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form III can be isolated with a purity greater than about 99%. Compound 1 Form IV

Provided herein is a solid form of Compound 1 having Form IV, which is described below in the Examples. In some embodiments, Form IV has one or more characteristic XRPD peaks selected from about 4.3, about 11.9, and about 12.9 degrees 2-theta.

In some embodiments, Form IV has at least one characteristic XRPD peak selected from about 4.3, about 11.9, about 12.9, about 14.3, about 15.1, and about 18.0 degrees 2- theta.

In some embodiments, Form IV has at least two characteristic XRPD peaks selected from about 4.3, about 11.9, about 12.9, about 14.3, about 15.1, about 15.5, about 18.0, about 23.3, about 24.5, about 25.1, and about 26.8 degrees 2-theta.

In some embodiments, Form IV has at least three characteristic XRPD peaks selected from about 4.3, about 11.9, about 12.9, about 14.3, about 15.1, about 15.5, about 18.0, about 23.3, about 24.5, about 25.1, and about 26.8 degrees 2-theta.

In some embodiments, Form IV has an XRPD partem with characteristic peaks as substantially shown in Figure 8.

In some embodiments, Form IV exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form IV has a DSC thermogram substantially as depicted in Figure 9.

In some embodiments, Form IV has one or more characteristic XRPD peaks selected from about 4.3, about 11.9, and about 12.9 degrees 2-theta; and exhibits a DSC thermogram having endotherm peak at a temperature of about 208 °C.

In some embodiments, Form IV has at least one characteristic XRPD peaks selected from about 4.3, about 11.9, about 12.9, about 14.3, about 15.1, and about 18.0 degrees 2- theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form IV has an XRPD pattern substantially as depicted in

FIG. 8 and a DSC thermogram substantially as depicted in FIG. 9.

In some embodiments, Form IV can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form IV can be isolated with a purity greater than about 99%. Compound 1 Form V

Provided herein is a solid form of Compound 1 having Form V, which is described below in the Examples. In some embodiments, Form V has one or more characteristic XRPD peaks selected from about 6.6, about 8.2, about 9.2, and about 17.9 degrees 2-theta.

In some embodiments, Form V has at least one characteristic XRPD peak selected from about 6.6, about 8.2, about 9.2, about 11.5, about 13.5, about 15.6, about 17.9, about 19.4, and about 20.7 degrees 2-theta.

In some embodiments, Form V has at least two characteristic XRPD peaks selected from about 6.6, about 8.2, about 9.2, about 11.5, about 13.5, about 15.6, about 17.9, about 19.4, about 20.7, about 23.5, and about 26.8 degrees 2-theta.

In some embodiments, Form V has at least three characteristic XRPD peaks selected from about 6.6, about 8.2, about 9.2, about 11.5, about 13.5, about 15.6, about 17.9, about 19.4, about 20.7, about 23.5, and about 26.8 degrees 2-theta.

In some embodiments, Form V has an XRPD pattern with characteristic peaks as substantially shown in Figure 10.

In some embodiments, Form V exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form V has a DSC

thermogram substantially as depicted in Figure 11. In some embodiments, Form V has a TGA thermogram substantially as depicted in Figure 12.

In some embodiments, Form V has one or more characteristic XRPD peaks selected from about 6.6, about 8.2, about 9.2, and about 17.9 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form V has at least one characteristic XRPD peaks selected from about 6.6, about 8.2, about 9.2, about 11.5, about 13.5, about 15.6, about 17.9, about 19.4, and about 20.7 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form V has an XRPD pattern substantially as depicted in FIG. 10 and a DSC thermogram substantially as depicted in FIG. 11.

In some embodiments, Form V can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form V can be isolated with a purity greater than about 99%. Compound 1 Form VI

Provided herein is a solid form of Compound 1 having Form VI, which is described below in the Examples. In some embodiments, Form VI has one or more characteristic XRPD peaks selected from about 4.4, about 5.2, and about 6.8 degrees 2-theta.

In some embodiments, Form VI has at least one characteristic XRPD peaks selected from about 4.4, about 5.2, about 6.8, about 10.1 , about 10.5, about 13.2, and about 15.8 degrees 2-theta.

In some embodiments, Form VI has at least two characteristic XRPD peaks selected from about 4.4, about 5.2, about 6.8, about 10.1 , about 10.5, about 12.7, about 13.2, about 15.8, about 18.4, about 19.2, about 19.6, and about 20.4 degrees 2-theta.

In some embodiments, Form VI has at least three characteristic XRPD peaks selected from about 4.4, about 5.2, about 6.8, about 10.1, about 10.5, about 12.7, about 13.2, about 15.8, about 18.4, about 19.2, about 19.6, and about 20.4 degrees 2-theta.

In some embodiments, Form VI has an XRPD partem with characteristic peaks as substantially shown in Figure 13.

In some embodiments, Form VI can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form VI can be isolated with a purity greater than about 99%. Compound 1 Form VII

Provided herein is a solid form of Compound 1 having Form VII, which is described below in the Examples. In some embodiments, Form VII has one or more characteristic XRPD peaks selected from about 5.1 , about 8.0, and about 10.2 degrees 2-theta.

In some embodiments, Form VII has at least one characteristic XRPD peaks selected from about 5.1, about 8.0, about 10.2, about 12.3, about 13.0, about 13.5, and about 16.3 degrees 2-theta.

In some embodiments, Form VII has at least two characteristic XRPD peaks selected from about 5.1, about 8.0, about 10.2, about 12.3, about 13.0, about 13.5, about 15.6, about 16.3, about 18.2, about 21.3, about 24.7, and about 37.4 degrees 2-theta.

In some embodiments, Form VII has at least three characteristic XRPD peaks selected from about 5.1, about 8.0, about 10.2, about 12.3, about 13.0, about 13.5, about 15.6, about 16.3, about 18.2, about 21.3, about 24.7, and about 37.4 degrees 2-theta.

In some embodiments, Form VII has an XRPD pattern with characteristic peaks as substantially shown in Figure 14. In some embodiments, Form VII exhibits a DSC thermogram having an endotherm peak at a temperature of about 205 °C. In some embodiments, Form VII has a DSC thermogram substantially as depicted in Figure 15. In some embodiments, Form VII has a TGA thermogram substantially as depicted in Figure 16.

In some embodiments, Form VII has one or more characteristic XRPD peaks selected from about 5.1, about 8.0, and about 10.2 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 205 °C.

In some embodiments, Form VII has at least one characteristic XRPD peaks selected from about 5.1, about 8.0, about 10.2, about 12.3, about 13.0, about 13.5, and about 16.3 degrees 2-theta; exhibits a DSC thermogram having an endotherm peak at a temperature of about 205 °C.

In some embodiments, Form VII has an XRPD partem substantially as depicted in FIG. 14 and a DSC thermogram substantially as depicted in FIG. 15.

In some embodiments, Form VII can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form VII can be isolated with a purity greater than about 99%.

Compound 1 Form VIII

Provided herein is a solid form of Compound 1 having Form VIII, which is described below in the Examples. In some embodiments, Form VIII has one or more characteristic

XRPD peaks selected from about 4.5, about 8.0, and about 9.0 degrees 2-theta.

In some embodiments, Form VIII has at least one characteristic XRPD peaks selected from about 4.5, about 8.0, about 9.0, about 12.7, about 13.3, about 14.3, about 15.5, about

16.4, and about 18.1 degrees 2-theta.

In some embodiments, Form VIII has at least two characteristic XRPD peaks selected from about 4.5, about 8.0, about 9.0, about 12.7, about 13.3, about 14.3, about 15.5, about

16.4, about 18.1, about 19.6, about 20.2, about 20.7, about 24.0, about 25.4, and about 26.6 degrees 2-theta.

In some embodiments, Form VIII has at least three characteristic XRPD peaks selected from about 4.5, about 8.0, about 9.0, about 12.7, about 13.3, about 14.3, about 15.5, about 16.4, about 18.1, about 19.6, about 20.2, about 20.7, about 24.0, about 25.4, and about 26.6 degrees 2-theta.

In some embodiments, Form VIII has an XRPD pattern with characteristic peaks as substantially shown in Figure 17. In some embodiments, Form VIII exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C. In some embodiments, Form VIII has a DSC thermogram substantially as depicted in Figure 18. In some embodiments, Form VIII has a TGA thermogram substantially as depicted in Figure 19.

In some embodiments, Form VIII has one or more characteristic XRPD peaks selected from about 4.5, about 8.0, and about 9.0 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C.

In some embodiments, Form VIII has at least one characteristic XRPD peaks selected from about 4.5, about 8.0, about 9.0, about 12.7, about 13.3, about 14.3, about 15.5, about 16.4, and about 18.1 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C.

In some embodiments, Form VIII has an XRPD pattern substantially as depicted in FIG. 17 and a DSC thermogram substantially as depicted in FIG. 18.

In some embodiments, Form VIII can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form VIII can be isolated with a purity greater than about 99%.

Compound 1 Form IX

Provided herein is a solid form of Compound 1 having Form IX, which is described below in the Examples. In some embodiments, Form IX has one or more characteristic XRPD peaks selected from about 6.4, about 8.0, and about 9.6 degrees 2-theta.

In some embodiments, Form IX has at least one characteristic XRPD peaks selected from about 6.4, about 8.0, about 9.6, about 13.3, about 15.3, about 16.0, and about 17.9 degrees 2-theta.

In some embodiments, Form IX has at least two characteristic XRPD peaks selected from about 6.4, about 8.0, about 9.6, about 13.3, about 15.3, about 16.0, about 17.9, about 18.7, about 19.7, about 20.5, about 22.4, about 23.3, about 24.2 degrees 2-theta.

In some embodiments, Form IX has at least three characteristic XRPD peaks selected from about 6.4, about 8.0, about 9.6, about 13.3, about 15.3, about 16.0, about 17.9, about 18.7, about 19.7, about 20.5, about 22.4, about 23.3, about 24.2 degrees 2-theta.

In some embodiments, Form IX has an XRPD pattern with characteristic peaks as substantially shown in Figure 20.

In some embodiments, Form IX exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C. In some embodiments, Form IX has a DSC thermogram substantially as depicted in Figure 21. In some embodiments, Form IX has a TGA thermogram substantially as depicted in Figure 22.

In some embodiments, Form IX has one or more characteristic XRPD peaks selected from about 6.4, about 8.0, and about 9.6 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C.

In some embodiments, Form IX has at least one characteristic XRPD peaks selected from about 6.4, about 8.0, about 9.6, about 13.3, about 15.3, about 16.0, and about 17.9 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 207 °C.

In some embodiments, Form IX has an XRPD pattern substantially as depicted in

FIG. 20 and a DSC thermogram substantially as depicted in FIG. 21.

In some embodiments, Form IX can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form IX can be isolated with a purity greater than about 99%.

Compound 1 Form IXa

Provided herein is a solid form of Compound 1 having Form IXa, which is described below in the Examples. In some embodiments, Form IXa has one or more characteristic

XRPD peaks selected from about 6.4, about 8.1 , and about 11.3 degrees 2-theta.

In some embodiments, Form IXa has at least one characteristic XRPD peaks selected from about 6.4, about 8.1, about 11.3, about 12.4, about 13.2, about 15.4, about 17.8, and about 19.0 degrees 2-theta.

In some embodiments, Form FXa has at least two characteristic XRPD peaks selected from about 6.4, about 8.1, about 11.3, about 12.4, about 13.2, about 15.4, about 17.8, about 19.0, about 20.5, about 21.3, about 22.8, about 23.3, about 23.9, and about 26.6 degrees 2- theta.

In some embodiments, Form IXa has at least three characteristic XRPD peaks selected from about 6.4, about 8.1, about 11.3, about 12.4, about 13.2, about 15.4, about 17.8, about 19.0, about 20.5, about 21.3, about 22.8, about 23.3, about 23.9, and about 26.6 degrees 2-theta.

In some embodiments, Form IXa has an XRPD pattern with characteristic peaks as substantially shown in Figure 23.

In some embodiments, Form IXa exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form IXa has a DSC thermogram substantially as depicted in Figure 24. In some embodiments, Form IXa has a TGA thermogram substantially as depicted in Figure 25.

In some embodiments, Form IXa has one or more characteristic XRPD peaks selected from about 6.4, about 8.1, and about 11.3 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form IXa has at least one characteristic XRPD peaks selected from about 6.4, about 8.1, about 11.3, about 12.4, about 13.2, about 15.4, about 17.8, and about 19.0 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form IXa has an XRPD pattern substantially as depicted in FIG. 23 and a DSC thermogram substantially as depicted in FIG. 24.

In some embodiments, Form IXa can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form IXa can be isolated with a purity greater than about 99%.

Compound 1 Form X

Provided herein is a solid form of Compound 1 having Form X, which is described below in the Examples. In some embodiments, Form X has one or more characteristic XRPD peaks selected from about 4.4, about 6.6, and about 8.2 degrees 2-theta.

In some embodiments, Form X has at least one characteristic XRPD peaks selected from about 4.4, about 6.6, about 8.2, about 8.8, about 12.9, about 21.4, about 22.4, and about 23.3 degrees 2-theta.

In some embodiments, Form X has at least two characteristic XRPD peaks selected from about 4.4, about 6.6, about 8.2, about 8.8, about 12.9, about 16.3, about 21.4, about 22.4, about 23.3, and about 25.8 degrees 2-theta.

In some embodiments, Form X has at least three characteristic XRPD peaks selected from about 4.4, about 6.6, about 8.2, about 8.8, about 12.9, about 16.3, about 21.4, about 22.4, about 23.3, and about 25.8 degrees 2-theta.

In some embodiments, Form X has an XRPD pattern with characteristic peaks as substantially shown in Figure 26.

In some embodiments, Form X can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form X can be isolated with a purity greater than about 99%. Compound 1 Form XI

Provided herein is a solid form of Compound 1 having Form XI, which is described below in the Examples. In some embodiments, Form XI has one or more characteristic XRPD peaks selected from about 8.2, about 9.8, and about 13.5 degrees 2-theta

In some embodiments, Form XI has at least one characteristic XRPD peaks selected from about 8.2, about 9.8, about 13.5, about 16.1, about 18.0, and about 22.7 degrees 2-theta.

In some embodiments, Form XI has at least two characteristic XRPD peaks selected from about 8.2, about 9.8, about 13.5, about 16.1, about 18.0, about 18.9, about 19.7, about 20.5, about 22.7, and about 23.6 degrees 2-theta.

In some embodiments, Form XI has at least three characteristic XRPD peaks selected from about 8.2, about 9.8, about 13.5, about 16.1, about 18.0, about 18.9, about 19.7, about 20.5, about 22.7, and about 23.6 degrees 2-theta.

In some embodiments, Form XI has an XRPD pattern with characteristic peaks as substantially shown in Figure 27.

In some embodiments, Form XI exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C. In some embodiments, Form XI has a DSC thermogram substantially as depicted in Figure 28. In some embodiments, Form XI has a TGA thermogram substantially as depicted in Figure 29.

In some embodiments, Form XI has one or more characteristic XRPD peaks selected from about 8.2, about 9.8, and about 13.5 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form XI has at least one characteristic XRPD peaks selected from about 8.2, about 9.8, about 13.5, about 16.1, about 18.0, and about 22.7 degrees 2-theta; and exhibits a DSC thermogram having an endotherm peak at a temperature of about 208 °C.

In some embodiments, Form XI has an XRPD pattern substantially as depicted in

FIG. 27 and a DSC thermogram substantially as depicted in FIG. 28.

In some embodiments, Form XI can be isolated with a purity of at least about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99%. In some embodiments, Form XI can be isolated with a purity greater than about 99%.

Process for Preparation of Compound 1

The present application further provides a process of preparing Compound 1, where the process can be suitable for scale up. A process of preparing Compound 1 is described in US 2016/0244448, the entirety of which is incorporated herein by reference. In comparison to the process described in US 2016/0244448, the process provided herein has certain advantages making it suitable for scale up. For example, the process provided herein affords high yields and good quality products.

In some embodiments, the process for preparing Compound 1 :

O (Compound 1),

or a salt thereof, comprises:

(i) converting Compound 8:

(Compound 8) to Compound 9:

(11) converting Compound 9 to Compound 10:

(Compound 10);

converting Compound 10 to Compound 11 : (Compound 1 1);

converting Compound 11 to Compound 12 diacetate:

2 HOAc (Compound 12 diacetate);

(v) converting Compound 12 diacetate to Compound 12 hydrochloride:

(Compound 12 hydrochloride); and converting Compound 12 hydrochloride to Compound 1.

In some embodiments, Compound 8 can be prepared by a process comprising: (i) converting Compound 2: (Compound 2) to Compound 4A:

(Compound 4 A);

(ii) converting Compound 4 A to Compound 4: (Compound 4);

converting Compound 4 to Compound 5:

(Compound 5);

(iv) converting Compound 5 to Compound 6:

(Compound 6);

(v) converting Compound 6 to Compound 7:

(Compound 7);

(vi) converting Compound 7 to Compound 8. Provided herein are processes for preparing Compound 1. or a salt thereof, which comprise converting Compound 12 or a salt thereof to Compound 1.

In some embodiments, the salt of Compound 12 is Compound 12 hydrochloride: (Compound 12 hydrochloride).

The conversion of Compound 12 hydrochloride to Compound 1 can comprise reacting Compound 12 hydrochloride with acryloyl chloride in the presence of Bl and SI , wherein B l is a base and SI is a solvent. For example, Bl is an alkali metal hydroxide base (e.g., sodium hydroxide). In some embodiments, S I comprises a halogenated solvent (e.g.,

dichloromethane). The conversion of Compound 12 hydrochloride to Compound 1 can be carried out at a temperature of about 30 °C or lower (e.g., about -10 °C to about 10 °C, or about 0 °C to about 10 °C). In some embodiments, about 2 to about 4 equivalents (e.g, about 3 equivalents) of Bl is used based on 1 equivalent of Compound 12 hydrochloride. In some embodiments, about 1 to about 1.5 equivalents (e.g. , about 1 equivalent) of acryloyl chloride is used based on 1 equivalent of Compound 12 hydrochloride.

In some embodiments, Compound 12 hydrochloride can be prepared by a process comprising converting Compound 12 diacetate:

• 2 HOAc (Compound 12 diacetate)

to Compound 12 hydrochloride.

The conversion of Compound 12 diacetate to Compound 12 hydrochloride can comprise reacting Compound 12 diacetate with B2 and hydrochloric acid in the presence of S2, wherein B2 is a hydroxide base and S2 is a solvent. For example, B2 is an alkali metal hydroxide base such as sodium hydroxide and ammonium hydroxide. In some embodiments, S2 comprises a halogenated solvent, protic solvent, or a mixture thereof. For example, S2 comprises dichloromethane, water, or a mixture thereof.

In some embodiments, Compound 12 diacetate can be prepared by a process comprising converting Compound 1 1 : (Compound 11)

to Compound 12 diacetate.

The conversion of Compound 11 to Compound 12 diacetate can comprise reacting Compound 11 with acetic acid and zinc. In some embodiments, the conversion of Compound 11 to Compound 12 diacetate is carried out at a temperature of about 10 °C to about 30 °C (e.g., about 15 °C to about 25 °C). In some embodiments, about 5 to about 7 equivalents (e.g., about 6 equivalents) of zinc is used based on 1 equivalent of Compound 11.

In some embodiments, Compound 11 can be prepared by a process comprising converting Compound 10:

(Compound 10) to Compound 11.

In some embodiments, the conversion of Compound 10 to Compound 11 comprises reacting Compound 11 with hydroxylamine hydrochloride in the presence of S3, wherein S3 is a solvent. For example, S3 comprises a protic solvent, a basic solvent, or a mixture thereof. In some embodiments, S3 comprises methanol, pyridine, or a mixture thereof. The conversion of Compound 10 to Compound 11 can be carried out at a temperature of about 45 °C or lower (e.g., about 15 °C to about 45 °C).

In some embodiments, Compound 10 can be prepared by a process comprising converting Compound 9:

9) to Compound 10.

The conversion of Compound 9 to Compound 10 can comprise reacting Compound 10 with sodium periodate and osmium tetroxide in the presence of S4, wherein S4 is a solvent. The conversion can further comprise B4, wherein B4 is a base. In some

embodiments, B4 is an alkali metal bicarbonate base (e.g., sodium bicarbonate). In some embodiments, S4 comprises an ether solvent, a protic solvent, an aprotic solvent, or a mixture thereof. In some embodiments, S4 comprises tetrahydrofuran, water, ethyl acetate, or a mixture thereof. The conversion can be carried out at a temperature of about 40 °C or lower (e.g., about 10 °C to about 40 °C). In some embodiments, about 3 to about 4 equivalents (e.g., about 3.5 equivalents) of sodium periodate is used based on 1 equivalent of Compound 9. In some embodiments, about 0.05 to about 0.10 equivalent (e.g., about 0.07 equivalent) of osmium tetroxide is used based on 1 equivalent of Compound 9.

In some embodiments, Compound 9 can be prepared by a process comprising converting Compound 8 :

(Compound 8) to Compound 9.

In some embodiments, the conversion of Compound 8 to Compound 9 comprises reacting Compound 8 with 4,4,5,5-tetramethyl-2-vinyl-l,3,2-dioxaborolane, PI, and B5 in the presence of S5, wherein PI is a transition metal catalyst, B5 is a base, and S5 is a solvent. In some embodiments, PI is a palladium catalyst (e.g., [Ι , - bis(diphenylphosphino)ferrocene]dichloropalladium (II) (Pd(dppf)Ci2)). In some

embodiments, B5 is cesium fluoride. In some embodiments, S5 comprises a protic solvent, an ether solvent, or a mixture thereof. In some embodiments, S5 comprises water, 1,4-dioxane, or a mixture thereof. The conversion can be carried out at a temperature of about 80 °C to about 100 °C (e.g., about 85 °C to about 95 °C). In some embodiments, about 3 to about 5 equivalents (e.g., about 4 equivalents) of B5 is used based on 1 equivalent of Compound 8. In some embodiments, about 0.01 to about 0.05 equivalent (about 0.02 equivalent) of PI is used based on 1 equivalent of Compound 8.

Compound 12 hydrochloride can also be prepared using other processes. For example, Compound 12 hydrochloride can be prepared by a process comprising converting Compound 15:

(Compound 15) to Compound 12 hydrochloride.

In some embodiments, the conversion of Compound 15 to Compound 12

hydrochloride comprises reacting Compound 15 with hydrochloric acid in the presence of S6, wherein S6 is a solvent. In some embodiments, S6 comprises an aprotic solvent, an ether solvent, or a mixture thereof. In some embodiments, S6 comprises dichloromethane, 1,4- dioxane, or a mixture thereof. In some embodiments, about 5 to about 10 equivalents (about 8 equivalents) of hydrochloric acid is used based on 1 equivalent of Compound 15.

In some embodiments, Compound 15 can be prepared by a process comprising converting Compound 8 to Compound 15.

In some embodiments, the conversion of Compound 8 to Compound 15 comprises reacting Compound 8 with potassium N-Boc-aminomethyltrifluoroborate, P2, and B7, wherein P2 is a transitional metal catalyst and B7 is base. In some embodiments, P2 is a palladium catalyst (e.g., cataCXium Pd G4). In some embodiments, B7 is a carbonate base (e.g., cesium carbonate base). In some embodiments, the conversion is carried out in S7, wherein S7 is a solvent. S7 can comprise a protic solvent, an ether solvent, or a mixture thereof. In some embodiments, S7 comprises water, dioxane, or a mixture thereof. The conversion can be carried out at a temperature of about 80 °C to about 90 °C (e.g., about 85 °C). In some embodiments, about 2 to about 3 equivalents (e.g., about 2.5 equivalents) of N- Boc-aminomethyltrifluoroborate is used based on 1 equivalent of Compound 8. In some embodiments, about 4 to about 5 equivalents (about 4.5 equivalents) of B7 is used based on 1 equivalent of Compound 8. In some embodiments, about 0.01 to about 0.05 equivalent (0.03 equivalent) of P2 is used based on 1 equivalent of Compound 8. In some embodiments, Compound 15 can be prepared by a process comprising converting Compound 12 diacetate to Compound 15.

In some embodiments, the conversion of Compound 12 diacetate to Compound 15 comprises reacting Compound 12 diacetate with di-tert-butyldicarbonate with B8. In some embodiments, B8 is an alkali metal hydroxide base such as sodium hydroxide. The conversion can be carried out in S8, wherein S8 is a solvent. In some embodiments, S8 comprises a protic solvent, an ether solvent, or a mixture thereof. In some embodiments, S8 comprises water, tetrahydrofuran, or a mixture thereof. In some embodiments, about 2 equivalents of di-tert-butyldicarbonate is used based on 1 equivalent of Compound 12 diacetate. In some embodiments, about 2 to about 4 equivalents (e.g., 3 equivalents) of B8 is used based on 1 equivalent of Compound 12 diacetate.

In some embodiments, Compound 8 can be prepared by a process comprising converting Compound 7:

(Compound 7) to Compound 8.

In some embodiments, the conversion of Compound 7 to Compound 8 comprises reacting Compound 7 with l -bromo-2-chloroethane and B9, wherein B9 is a base. In some embodiments, B9 is carbonate base (e.g. , cesium carbonate). The conversion can be carried out in S9, wherein S9 is a solvent. In some embodiments, S9 comprises an aprotic solvent such as Ν,Ν-dimethylformamide. In some embodiments, about 2 equivalents (e.g., about 1.5, about 1.6, about 1.7, about 1.8, about 1.9, or about 2.0 equivalents) of l -bromo-2- chloroethane is used based on 1 equivalent of Compound 7. In some embodiments, about 1 to about 3 equivalents (2 equivalents) of B9 is used based on 1 equivalent of Compound 7.

In some embodiments, Compound 7 can be prepared by a process comprising converting Compound 6: (Compound 6)

to Compound 7.

In some embodiments, the conversion of Compound 6 to Compound 7 comprises reacting Compound 6 with hydrogen chloride. The conversion can be carried out in S 10, wherein S 10 is a solvent. In some embodiments, S 10 comprises a protic solvent, an ether solvent, or a mixture thereof. In some embodiments, S 10 comprises water, 1,4-dioxane, or a mixture thereof. The conversion can be carried out at a temperature of about 70 °C to about 90 °C (e.g., about 80 °C). In some embodiments, about 5 to about 15 equivalents (e.g., about 10 equivalents) of hydrogen chloride is used based on 1 equivalent of Compound 6. The conversion of Compound 6 to Compound 7 can be deemed completed as indicated by HPLC. The pH of the reaction mixture can be adjusted to 8 with a solution of sodium hydroxide in water.

In some embodiments, Compound 6 can be prepared by a process comprising converting Compound 5 :

(Compound 5) to Compound 6.

In some embodiments, the conversion of Compound 5 to Compound 6 comprises reacting Compound 5 with Cul, Bl 1 , and 2-pyridinecarboxylic acid, wherein Bl 1 is a base. In some embodiments, Bl 1 is a carbonate base such as cesium carbonate. The conversion can be carried out in S I 1 , wherein S I 1 is a solvent. In some embodiments, SI 1 comprises an aprotic solvent such as dimethylsulfoxide. The conversion can be carried out at a temperature of about 1 10 °C to about 130 °C (e.g., about 120 °C). In some embodiments, about 0.1 to about 0.3 equivalent (e.g. , about 0.2 equivalent) of Cul is used based on 1 equivalent of Compound 5. In some embodiments, about 2 to about 4 equivalents (e.g. , about 3 equivalents) of Bl l is used based on 1 equivalent of Compound 5. In some embodiments, about 0.5 to about 1 equivalent (e.g., about 0.8 equivalent) of 2-pyridinecarboxylic acid is used based on 1 equivalent of Compound 5.

Alternatively, the conversion of Compound 5 to Compound 6 comprises reacting Compound 5 with B12, wherein B12 is a base. In some embodiments, B12 is a phosphate base such as potassium phosphate. In some embodiments, the conversion can be carried out in S12, wherein S12 is a solvent. In some embodiments, S12 comprises an aprotic solvent (e.g., dimethylsulfoxide). In some embodiments, the conversion of Compound 5 to

Compound 6 is carried out at a temperature of about 70 °C to about 100 °C (e.g., about 80 °C to about 90 °C). In some embodiments, about 3 to about 4 equivalents (e.g., about 3.5 equivalents) of B12 is used based on 1 equivalent of Compound 5.

In some embodiments, Compound 5 can be prepared by a process comprising converting Compound 4:

(Compound 4) to Compound 5.

In some embodiments, the conversion of Compound 4 to Compound 5 comprises reacting Compound 4 with ethyl malonyl chloride and B13, wherein B13 is a base. In some embodiments, B13 is a trimethylamine or sodium bicarbonate. The conversion can be carried out in SI 3, wherein S13 is a solvent. In some embodiments, S13 comprises an aprotic solvent such as dichloromethane. In some embodiments, about 1 to about 1.5 equivalents (e.g., 1 equivalent) of ethyl malonyl chloride is used based on 1 equivalent of Compound 4. In some embodiments, about 1 to about 2 equivalents (e.g., 1.5 equivalents) of trimethylamine is used based on 1 equivalent of Compound 4. In some embodiments, about 2 to about 4 equivalents (e.g., 3 equivalents) of sodium bicarbonate is used based on 1 equivalent of Compound 4.

In some embodiments, Compound 4 can be prepared by a process comprising converting Compound 4A:

to Compound 4.

In some embodiments, the conversion of Compound 4A to Compound 4 comprises reacting Compound 4A with sodium borohydride. The conversion can be carried out in S 14, wherein S14 is a solvent. In some embodiments, S14 comprises a halogenated solvent, a protic solvent, or a mixture thereof. In some embodiments, S14 comprises dichloromethane, methanol, or a mixture thereof. The conversion can be carried out at a temperature of about 5 °C to about 10 °C (e.g., about 8 °C). In some embodiments, about 1 to about 1.5 equivalents (e.g., about 1 equivalent) of sodium borohydride is used based on 1 equivalent of Compound 4A.

In some embodiments, Compound 4A can be prepared by a process comprising converting Compound 2: (Compound 2)

to Compound 4A.

In some embodiments, the conversion of Compound 2 to Compound 4A comprises reacting Compound 2 with 2,6-difluoro-3,5-dimethoxyaniline and acetic acid. The conversion can be carried out in S15, wherein S15 is a solvent. In some embodiments, S15 comprises a protic solvent such as ethanol. In some embodiments, about 1 equivalent of 2,6-difluoro-3,5- dimethoxyaniline is used based on 1 equivalent of Compound 2. In some embodiments, about 3 equivalents of acetic acid is used based on 1 equivalent of Compound 2.

Compound 4 can also be prepared starting from Compound 2 without the isolation of Compound 4A. For example, the conversion of Compound 2 to Compound 4A comprises reacting Compound 2 with 2,6-difluoro-3,5-dimethoxyaniline and dibutyltin dichloride. In some embodiments, the conversion of Compound 2 to Compound 4 further comprises phenylsilane. The conversion can be carried out in SI 6, wherein S 16 is a solvent. In some embodiments, S16 comprises an ether solvent (e.g., tetrahydrofuran). In some embodiments, about 1 equivalent of 2,6-difluoro-3,5-dimethoxyaniline is used based on 1 equivalent of Compound 2. In some embodiments, about 0.1 to about 0.3 equivalent (e.g. , about 0.2 equivalent) of dibutyltin dichloride is used based on 1 equivalent of Compound 2. In some embodiments, about 1 equivalent of phenylsilane is used based on 1 equivalent of Compound 2.

Alternatively, Compound 4 can be prepared starting from Compound 2 without the isolation of Compound 4A as described below. For example, the conversion of Compound 2 to Compound 4A comprises reacting Compound 2 with 2,6-difluoro-3,5-dimethoxyaniline, TMSC1 and borane. The conversion can be carried out in S 17, wherein S 17 is a solvent. In some embodiments, S 17 comprises a pro tic solvent, an ether solvent, or a mixture thereof. In some embodiments, S 17 comprises water, tetrahydrofuran, or a mixture thereof. In some embodiments, about 1 equivalent of 2,6-difluoro-3,5-dimethoxyaniline is used based on 1 equivalent of Compound 2. In some embodiments, about 2 to about 3 equivalents (e.g., about 2.5 equivalents) of TMSC1 is used based on 1 equivalent of Compound 2. In some embodiments, about 1 equivalent of borane is used based on 1 equivalent of Compound 2. In some embodiments, the conversion of Compound 2 to Compound 4 further comprises treating a mixture of Compound 2, 2,6-difluoro-3,5-dimethoxyaniline, TMSCL, and borane with ammonium hydroxide.

In some embodiments provided herein is a compound which is

9), or a salt thereof.

In some embodiments provided herein is a compound which is

(Compound 10), or a salt thereof.

In some embodiments provided herein is a compound which is (Compound 11), or a salt thereof.

In some embodiments rovided herein is a compound which is

(Compound 12 diacetate).

In some embodiments provided herein is a compound which is

(Compound 12 hydrochloride).

The processes described herein can be monitored according to any suitable method known in the art. For example, product formation can be monitored by spectroscopic means, such as nuclear magnetic resonance spectroscopy (e.g., ¾ or 13 C), infrared spectroscopy, spectrophotometry (e.g., UV- visible), or mass spectrometry; or by chromatography such as high performance liquid chromatography (HPLC) or thin layer chromatography. The compounds obtained by the reactions can be purified by any suitable method known in the art. For example, chromatography (medium pressure) on a suitable adsorbent (e.g. , silica gel, alumina and the like), HPLC, or preparative thin layer chromatography; distillation;

sublimation, trituration, or recrystallization. The purity of the compounds, in general, are determined by physical methods such as measuring the melting point (in case of a solid), obtaining a NMR spectrum, or performing a HPLC separation. If the melting point decreases, if unwanted signals in the NMR spectrum are decreased, or if extraneous peaks in an HPLC trace are removed, the compound can be said to have been purified. In some embodiments, the compounds are substantially purified.

Preparation of compounds can involve the protection and deprotection of various chemical groups. The need for protection and deprotection, and the selection of appropriate protecting groups can be readily determined by one skilled in the art. The chemistry of protecting groups can be found, for example, in Wuts and Greene, Greene 's Protective

Groups in Organic Synthesis, 4 th Ed., John Wiley & Sons: New York, 2006, which is incorporated herein by reference in its entirety.

The reactions of the processes described herein can be carried out at appropriate temperatures which can be readily determined by the skilled artisan. Reaction temperatures will depend on, for example, the melting and boiling points of the reagents and solvent, if present; the thermodynamics of the reaction (e.g., vigorously exothermic reactions may need to be carried out at reduced temperatures); and the kinetics of the reaction (e.g., a high activation energy barrier may need elevated temperatures).

In some embodiments, concentrating a solution as described herein refers to a solution where its volume is reduced by letting the solvent evaporate, by heating the solution, by subjecting the solution to reduced pressure, or any combination thereof.

As used herein, the phrase "alkali metal bicarbonate base," employed alone or in combination with other terms, refers to a base having formula M(HCOs), wherein M refers to an alkali metal (e.g. lithium, sodium, or potassium). Example alkali metal bicarbonate bases include, but are not limited to, lithium bicarbonate, sodium bicarbonate, and potassium bicarbonate.

As used herein, the phrase "alkali metal hydroxide base," employed alone or in combination with other terms, refers to a base having formula MOH, wherein M refers to an alkali metal (e.g. lithium, sodium, or potassium). Example alkali metal hydroxide bases include, but are not limited to lithium hydroxide, sodium hydroxide, and potassium hydroxide.

As used herein, the phrase "transition metal catalyst" refers to a metal catalyst (e.g., palladium or nickel catalyst) suitable to catalyze a carbon-carbon coupling reaction. Example transition metal catalysts include, but are not limited to, PdCh(PPh3)2, Pd(PPl¾)4, dichloro(bis{di-tert-butyl[4-(dimethylamino)phenyl]-phosphor anyl})palladium (Pd-132), NiCi2(dppf), and NiCh(dppp), where (dppf) refers to l,l '-bis(diphenylphosphino)ferrocene and (dppp) refers to l,3-bis(diphenylphosphino)propane. Example palladium catalysts include but are not limited to PdCl2(PPh3)2, Pd(PPh3)4, dichloro(bis{di-tert-butyl[4-(dimethylamino)phenyl]-phosphor anyl})palladium (Pd-132), palladium on carbon, PdCl 2 , Pd(OAc) 2 , PdCl 2 (MeCN) 2 ,

tris(dibenzylideneacetone)dipalladium(0) (Pd 2 (dba)3, 4-(di-fert-butylphosphino)-N,N- dimethylaniline-dichloropalladium (2: 1), Pd(dppf)Cl 2 (e.g., Pd(dppf)Cl 2 -CH 2 Cl 2 ), and tetrakis(tri(o-tolyl)phosphine)palladium(0).

In some embodiments, anti-solvent as described herein refers to a solvent where

Compound 1 is less soluble relative to another solvent or solvent mixture in the solution. For example, anti-solvent can include but not limited to benzene, cyclohexane, pentane, hexane, heptane (e.g., n-heptane), toluene, cycloheptane, methylcyclohexane, heptane, ethylbenzene, m-, 0-, or p-xylene, octane, indane, nonane, or naphthalene.

The reactions of the processes described herein can be carried out in suitable solvents which can be readily selected by one of skill in the art of organic synthesis. Suitable solvents can be substantially nonreactive with the starting materials (reactants), the intermediates, or products at the temperatures at which the reactions are carried out, e.g., temperatures which can range from the solvent's freezing temperature to the solvent's boiling temperature. A given reaction can be carried out in one solvent or a mixture of more than one solvent.

Depending on the particular reaction step, suitable solvents for a particular reaction step can be selected. In some embodiments, reactions can be carried out in the absence of solvent, such as when at least one of the reagents is a liquid or gas.

Suitable solvents can include halogenated solvents such as carbon tetrachloride, bromodichloromethane, dibromochloromethane, bromoform, chloroform,

bromochloromethane, dibromomethane, butyl chloride, dichloromethane (methylene chloride), tetrachloroethylene, trichloroethylene, 1,1, 1-trichloroethane, 1,1 ,2-trichloroethane, 1,1-dichloroethane, 2-chloropropane, α,α,α-trifluorotoluene, 1 ,2-dichloroethane, 1,2- dibromo ethane, hexafluorobenzene, 1,2,4-trichlorobenzene, 1,2-dichlorobenzene, chlorobenzene, fluorobenzene, mixtures thereof and the like.

Suitable ether solvents include: dimethoxymethane, tetrahydrofuran, 1,3-dioxane, 1 ,4- dioxane, furan, tetrahydrofuran (THF), diethyl ether, ethylene glycol dimethyl ether, ethylene glycol diethyl ether, diethylene glycol dimethyl ether (diglyme), diethylene glycol diethyl ether, triethylene glycol dimethyl ether, anisole, fert-butyl methyl ether, mixtures thereof and the like.

Suitable protic solvents can include, by way of example and without limitation, water, methanol, ethanol, 2-nitroethanol, 2-fluoroethanol, 2,2,2 -trifluoroethanol, ethylene glycol, 1- propanol, 2-propanol, 2-methoxyethanol, 1-butanol, 2-butanol, wo-butyl alcohol, fert-butyl alcohol, 2-ethoxyethanol, diethylene glycol, 1-, 2-, or 3- pentanol, neo-pentyl alcohol, tert- pentyl alcohol, diethylene glycol monomethyl ether, diethylene glycol monoethyl ether, cyclohexanol, benzyl alcohol, phenol, or glycerol.

Suitable aprotic solvents can include, by way of example and without limitation, N,N- dimethylformamide (DMF), N,N-dimethylacetamide (DMA), l,3-dimethyl-3, 4,5,6- tetrahydro-2(lH)-pyrimidinone (DMPU), l,3-dimethyl-2-imidazolidinone (DMI),

N-methylpyrrolidinone (NMP), formamide, N-methylacetamide, N-methylformamide, acetonitrile, dimethyl sulfoxide, propionitrile, ethyl formate, methyl acetate,

hexachloro acetone, acetone, ethyl methyl ketone, ethyl acetate, sulfolane, N,N- dimethylpropionamide, tetramethylurea, nitromethane, nitrobenzene, or

hexamethylphosphoramide.

Suitable hydrocarbon solvents include benzene, cyclohexane, pentane, hexane, toluene, cycloheptane, methylcyclohexane, heptane, ethylbenzene, m-, o-, or p-xylene, octane, indane, nonane, or naphthalene.

The reactions of the processes described herein can be carried out in air or under an inert atmosphere. Typically, reactions containing reagents or products that are substantially reactive with air can be carried out using air-sensitive synthetic techniques that are well known to the skilled artisan.

The expressions, "ambient temperature" and "room temperature," as used herein, are understood in the art, and refer 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. Methods of Use

Compound 1 or solid forms thereof as described herein can inhibit the activity of the FGFR4 enzyme. For example, Compound 1 can be used to inhibit activity of an FGFR4 enzyme in a cell or in an individual or patient in need of inhibition of the enzyme by administering an inhibiting amount of Compound 1 to the cell, individual, or patient.

In some embodiments, Compound 1 is selective for the enzyme FGFR4 over one or more of FGFRl , FGFR2, and/or FGFR3. In some embodiments, Compound 1 is selective for the enzyme FGFR4 over FGFRl , FGFR2, and FGFR3. In some embodiments, Compound 1 is selective for the enzyme FGFR4 over VEGFR2. In some embodiments, the selectivity is 2-fold or more, 3-fold or more, 5-fold or more, 10-fold or more, 25-fold or more, 50-fold or more, or 100-fold or more.

As FGFR4 inhibitors, Compound 1 is useful in the treatment of various diseases associated with abnormal expression or activity of the FGFR4 enzyme or FGFR ligands. Compounds which inhibit FGFR will be useful in providing a means of preventing the growth or inducing apoptosis in tumors, particularly by inhibiting angiogenesis. It is therefore anticipated that Compound 1 will prove useful in treating or preventing proliferative disorders such as cancers. In particular tumors with activating mutants of receptor tyrosine kinases or upregulation of receptor tyrosine kinases may be particularly sensitive to the inhibitors.

In certain embodiments, the FGFR4, or a mutant thereof, activity is inhibited irreversibly. In certain embodiments, FGFR4, or a mutant thereof, activity is inhibited irreversibly by covalently modifying Cys 552 of FGFR4.

In certain embodiments, the disclosure provides a method for treating a FGFR4- mediated disorder in a patient in need thereof, comprising the step of administering to said patient Compound 1, or a pharmaceutically acceptable composition thereof.

For example, Compound 1 or solid forms thereof are useful in the treatment of cancer. Example cancers include bladder cancer, breast cancer, cervical cancer, colorectal cancer, cancer of the small intestine, colon cancer, rectal cancer, cacncer of the anus, endometrial cancer, gastric cancer, head and neck cancer (e.g., cancers of the larynx, hypopharynx, nasopharynx, oropharynx, lips, and mouth), kidney cancer, liver cancer (e.g., hepatocellular carcinoma, cholangiocellular carcinoma), lung cancer (e.g., adenocarcinoma, small cell lung cancer and non-small cell lung carcinomas, parvicellular and non-parvicellular carcinoma, bronchial carcinoma, bronchial adenoma, pleuropulmonary blastoma), ovarian cancer, prostate cancer, testicular cancer, uterine cancer, esophageal cancer, gall bladder cancer, pancreatic cancer (e.g. exocrine pancreatic carcinoma), stomach cancer, thyroid cancer, parathyroid cancer, skin cancer (e.g., squamous cell carcinoma, Kaposi sarcoma, Merkel cell skin cancer), and brain cancer (e.g., astrocytoma, medulloblastoma, ependymoma, neuroectodermal tumors, pineal tumors).

Further example cancers include hematopoietic malignancies such as leukemia or lymphoma, multiple myeloma, chronic lymphocytic lymphoma, adult T cell leukemia, B-cell lymphoma, cutaneous T-cell lymphoma, acute myelogenous leukemia, Hodgkin's or non- Hodgkin's lymphoma, myeloproliferative neoplasms (e.g., polycythemia vera, essential thrombocythemia, and primary myelofibrosis), Waldenstrom's Macroglubulinemia, hairy cell lymphoma, chronic myelogenic lymphoma, acute lymphoblastic lymphoma, AIDS-related lymphomas, and Burkitt's lymphoma.

Other cancers treatable with Compound 1 or solid forms thereof include tumors of the eye, glioblastoma, melanoma, rhabdosarcoma, lymphosarcoma, and osteosarcoma.

Compound 1 or solid forms thereof can also be useful in the inhibition of tumor metastases.

In some embodiments, the present disclosure provides a method for treating hepatocellular carcinoma in a patient in need thereof, comprising the step of administering to said patient Compound 1 , or a pharmaceutically acceptable composition thereof.

In some embodiments, the present disclosure provides a method for treating

Rhabdomyosarcoma, esophageal cancer, breast cancer, or cancer of a head or neck, in a patient in need thereof, comprising the step of administering to said patient Compound 1, or a pharmaceutically acceptable composition thereof.

In some embodiments, the present disclosure provides a method of treating cancer, wherein the cancer is selected from hepatocellular cancer, breast cancer, bladder cancer, colorectal cancer, melanoma, mesothelioma, lung cancer, prostate cancer, pancreatic cancer, testicular cancer, thyroid cancer, squamous cell carcinoma, glioblastoma, neuroblastoma, uterine cancer, and rhabdosarcoma.

As used herein, the term "cell" is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal.

As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" the FGFR4 enzyme with a compound described herein (e.g., Compound 1) includes the administration of a compound described herein to an individual or patient, such as a human, having FGFR, as well as, for example, introducing a compound described herein (e.g., Compound 1) into a sample containing a cellular or purified preparation containing the FGFR4 enzyme.

As used herein, the term "individual" or "patient," used interchangeably, refers to any animal, including mammals, preferably mice, rats, other rodents, rabbits, dogs, cats, swine, cattle, sheep, horses, or primates, and most preferably humans.

As used herein, the phrase "therapeutically effective amount" refers to the amount of active compound or pharmaceutical agent such as an amount of any of the solid forms or salts thereof as disclosed herein 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. An appropriate "effective" amount in any individual case may be determined using techniques known to a person skilled in the art.

The phrase "pharmaceutically acceptable" is used 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, immunogenicity or other problem or complication, commensurate with a reasonable benefit/risk ratio.

As used herein, the phrase "pharmaceutically acceptable carrier or excipient" refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, solvent, or encapsulating material. Excipients or carriers are generally safe, non-toxic and neither biologically nor otherwise undesirable and include excipients or carriers that are acceptable for veterinary use as well as human pharmaceutical use. In one embodiment, each component is "pharmaceutically acceptable" as defined herein. See, e.g., Remington: The Science and Practice of Pharmacy, 21st ed.; Lippincott Williams & Wilkins: Philadelphia, Pa., 2005; Handbook of Pharmaceutical Excipients, 6th ed.; Rowe et al, Eds.; The Pharmaceutical Press and the American Pharmaceutical Association: 2009; Handbook of Pharmaceutical Additives, 3rd ed.; Ash and Ash Eds. ; Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, 2nd ed. ; Gibson Ed.; CRC Press LLC: Boca Raton, Fla., 2009.

As used herein, the term "treating" or "treatment" refers to inhibiting the disease; for example, inhibiting a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e.,, arresting further development of the pathology and/or symptomatology) or ameliorating the disease; for example, ameliorating a disease, condition or disorder in an individual who is experiencing or displaying the pathology or symptomatology of the disease, condition or disorder (i.e. ,, reversing the pathology and/or symptomatology) such as decreasing the severity of disease.

It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, can also be provided in combination in a single embodiment (while the embodiments are intended to be combined as if written in multiply dependent form). Conversely, various features of the invention which are, for brevity, described in the context of a single embodiment, can also be provided separately or in any suitable subcombination.

Combination Therapy

One or more additional pharmaceutical agents or treatment methods such as, for example, anti- viral agents, chemotherapeutics or other anti-cancer agents, immune enhancers, immunosuppressants, radiation, anti-tumor and anti-viral vaccines, cytokine therapy (e.g., IL2, GM-CSF, etc.), and/or tyrosine kinase inhibitors can be used in combination with Compound 1 for treatment of FGFR-associated diseases, disorders or conditions, or diseases or conditions as described herein. The agents can be combined with the present compounds in a single dosage form, or the agents can be administered simultaneously or sequentially as separate dosage forms.

Compound 1 can be used in combination with one or more other kinase inhibitors for the treatment of diseases, such as cancer, that are impacted by multiple signaling pathways. For example, a combination can include one or more inhibitors of the following kinases for the treatment of cancer: Aktl , Akt2, Akt3, TGF-PR, Pirn, PKA, PKG, PKC, CaM-kinase, phosphorylase kinase, MEKK, ERK, MAPK, mTOR, EGFR, HER2, HER3, HER4, INS-R, IGF-1R, IR-R, PDGFaR, PDGFpR, CSFIR, KIT, FLK-II, KDR/FLK-1, FLK-4, flt-1 , FGFR1, FGFR2, FGFR3, FGFR4, c-Met, Ron, Sea, TRKA, TRKB, TRKC, FLT3,

VEGFR/Flt2, Flt4, EphAl, EphA2, EphA3, EphB2, EphB4, Tie2, Src, Fyn, Lck, Fgr, Btk, Fak, SYK, FRK, JAK, ABL, ALK and B-Raf. Additionally, Compound 1 can be combined with inhibitors of kinases associated with the PIK3/Akt/mTOR signaling pathway, such as PI3K, Akt (including Aktl , Akt2 and Akt3) and mTOR kinases.

In some embodiments, Compound 1 can be used in combination with one or more inhibitors of the enzyme or protein receptors such as HPKl , SBLB, TUT4, A2A/A2B, CD47, CDK2, STING, ALK2, LIN28, ADARl , MAT2a, RIOKl, HDAC8, WDR5, SMARCA2, and DCLK1 for the treatment of diseases and disorders. Exemplary diseases and disorders include cancer, infection, inflammation and neurodegenerative disorders.

In some embodiments, Compound 1 can be used in combination with a therapeutic agent that targets an epigenetic regulator. Examples of epigenetic regulators include bromodomain inhibitors, the histone lysine methyltransferases, histone arginine methyl transferases, histone demethylases, histone deacetylases, histone acetylases, and DNA methyltransferases. Histone deacetylase inhibitors include, e.g. , vorinostat. For treating cancer and other proliferative diseases, Compound 1 can be used in combination with targeted therapies, including JAK kinase inhibitors (Ruxolitinib, additional JAK1/2 and JAK1 -selective, baricitinib or INCB39110), Pirn kinase inhibitors (e.g., INCB53914), PI3 kinase inhibitors including PI3K-delta selective and broad spectrum PI3K inhibitors (e.g., INCB50465 and INCB54707), PBK-gamma inhibitors such as PBK-gamma selective inhibitors, MEK inhibitors, CSF1R inhibitors, TAM receptor tyrosine kinases inhibitors (Tyro-3, Axl, and Mer; e.g., INCB81776), angiogenesis inhibitors, interleukin receptor inhibitors, Cyclin Dependent kinase inhibitors, BRAF inhibitors, mTOR inhibitors, proteasome inhibitors (Bortezomib, Carfilzomib), HDAC-inhibitors (panobinostat, vorinostat), DNA methyl transferase inhibitors, dexamethasone, bromo and extra terminal family members inhibitors (for example, bromodomain inhibitors or BET inhibitors, such as INCB54329 or INCB57643), LSD1 inhibitors (e.g., INCB59872 or INCB60003), arginase inhibitors (e.g., INCB1158), indoleamine 2,3-dioxygenase inhibitors (e.g., epacadostat, NLG919 or BMS-986205), and PARP inhibiors (e.g., olaparib or rucaparib).

For treating cancer and other proliferative diseases, Compound 1 can be used in combination with chemotherapeutic agents, agonists or antagonists of nuclear receptors, or other anti-proliferative agents. Compound 1 or solid forms thereof can also be used in combination with a medical therapy such as surgery or radiotherapy, e.g. , gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachy therapy, and systemic radioactive isotopes. Examples of suitable chemotherapeutic agents include any of: abarelix, aldesleukin, alemtuzumab, alitretinoin, allopurinol, altretamine, anastrozole, arsenic trioxide, asparaginase, azacitidine, baricitinib, bendamustine, bevacizumab, bexarotene, bleomycin, bortezombi, bortezomib, busulfan intravenous, busulfan oral, calusterone, capecitabine, carboplatin, carmustine, cetuximab, chlorambucil, cisplatin, cladribine, clofarabine, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, dalteparin sodium, dasatinib, daunorubicin, decitabine, denileukin, denileukin diftitox, dexrazoxane, docetaxel, doxorubicin, dromostanolone propionate, eculizumab, epirubicin, erlotinib, estramustine, etoposide phosphate, etoposide, exemestane, fentanyl citrate, filgrastim, floxuridine, fludarabine, fluorouracil, fulvestrant, gefitinib, gemcitabine, gemtuzumab ozogamicin, goserelin acetate, histrelin acetate, ibritumomab tiuxetan, idarubicin, ifosfamide, imatinib mesylate, interferon alfa 2a, irinotecan, lapatinib ditosylate, lenalidomide, letrozole, leucovorin, leuprolide acetate, levamisole, lomustine, meclorethamine, megestrol acetate, melphalan, mercaptopurine, methotrexate, methoxsalen, mitomycin C, mitotane, mitoxantrone, nandrolone phenpropionate, nelarabine, niraparib, nofetumomab, olaparib, oxaliplatin, paclitaxel, pamidronate, panobinostat, panitumumab, pegaspargase,

pegfilgrastim, pemetrexed disodium, pentostatin, pipobroman, plicamycin, procarbazine, quinacrine, rasburicase, rituximab, rucaparib, ruxolitinib, sorafenib, streptozocin, sunitinib, sunitinib maleate, tamoxifen, temozolomide, teniposide, testolactone, thalidomide, thioguanine, thiotepa, topotecan, toremifene, tositumomab, trastuzumab, tretinoin, uracil mustard, valrubicin, vinblastine, vincristine, vinorelbine, vorinostat, veliparib, talazoparib and zoledronate.

In some embodiments, Compound 1 can be used in combination with immune checkpoint inhibitors. Exemplary immune checkpoint inhibitors include inhibitors against immune checkpoint molecules such as CD27, CD28, CD40, CD122, CD96, CD73, CD47, OX40, GITR, CSF1R, JAK, PI3K delta, PI3K gamma, TAM, arginase, CD137 (also known as 4-1BB), ICOS, A2AR, B7-H3, B7-H4, BTLA, CTLA-4, LAG3 (e.g., INCAGN2385), TIM3 (e.g., INCB2390), VISTA, PD-1, PD-Ll and PD-L2. In some embodiments, the immune checkpoint molecule is a stimulatory checkpoint molecule selected from CD27, CD28, CD40, ICOS, OX40 (e.g., INCAGN1949), GITR (e.g., INCAGN1876) and CD137. In some embodiments, the immune checkpoint molecule is an inhibitory checkpoint molecule selected from A2AR, B7-H3, B7-H4, BTLA, CTLA-4, IDO, KIR, LAG3, PD-1, TIM3, and VISTA. In some embodiments, Compound 1 can be used in combination with one or more agents selected from KIR inhibitors, TIGIT inhibitors, LAIR1 inhibitors, CD160 inhibitors, 2B4 inhibitors and TGFR beta inhibitors.

In some embodiments, the inhibitor of an immune checkpoint molecule is anti-PDl antibody, anti-PD-Ll antibody, or anti-CTLA-4 antibody.

In some embodiments, the inhibitor of an immune checkpoint molecule is a small molecule PD-Ll inhibitor. In some embodiments, the small molecule PD-Ll inhibitor has an IC50 less than 1 μΜ, less than 100 nM, less than 10 nM or less than 1 nM in a PD-Ll assay described in US Patent Publication Nos. US 20170107216, US 20170145025, US

20170174671, US 20170174679, US 20170320875, US 20170342060, US 20170362253, and US 20180016260, each of which is incorporated by reference in its entirety for all purposes.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-1, e.g., an anti-PD-1 monoclonal antibody. In some embodiments, the anti-PD-1 monoclonal antibody is MGA012, nivolumab, pembrolizumab (also known as MK-3475), pidilizumab, SHR-1210, PDR001, ipilumimab or AMP-224. In some embodiments, the anti- PD-1 monoclonal antibody is nivolumab or pembrolizumab. In some embodiments, the anti- PDl antibody is pembrolizumab. In some embodiments, the anti-PDl antibody is pembrolizumab. In some embodiments, the anti-PD-1 monoclonal antibody is MGA012. In some embodiments, the anti-PDl antibody is SHR-1210. Other anti-cancer agent(s) include antibody therapeutics such as 4-1BB (e.g. urelumab, utomilumab.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of PD-L1, e.g., an anti-PD-Ll monoclonal antibody. In some embodiments, the anti-PD-Ll monoclonal antibody is BMS-935559, MEDI4736, MPDL3280A (also known as RG7446), or MSB0010718C. In some embodiments, the anti-PD-Ll monoclonal antibody is MPDL3280A or MEDI4736.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of CTLA-4, e.g., an anti-CTLA-4 antibody. In some embodiments, the anti-CTLA- 4 antibody is ipilimumab.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of LAG3, e.g., an anti-LAG3 antibody. In some embodiments, the anti-LAG3 antibody is BMS-986016 or LAG525.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of GITR, e.g., an anti-GITR antibody. In some embodiments, the anti-GITR antibody is TRX518 or MK-4166.

In some embodiments, the inhibitor of an immune checkpoint molecule is an inhibitor of OX40, e.g., an anti-OX40 antibody or OX40L fusion protein. In some embodiments, the anti-OX40 antibody is MEDI0562. In some embodiments, the OX40L fusion protein is MEDI6383.

In some embodiments, Compound 1 can be used in combination with one or more agents for the treatment of diseases such as cancer. In some embodiments, the agent is an alkylating agent, a proteasome inhibitor, a corticosteroid, or an immunomodulatory agent. Examples of an alkylating agent include cyclophosphamide (CY), melphalan (MEL), and bendamustine. In some embodiments, the proteasome inhibitor is carfilzomib. In some embodiments, the corticosteroid is dexamethasone (DEX). In some embodiments, the immunomodulatory agent is lenalidomide (LEN) or pomalidomide (POM). Suitable antiviral agents contemplated for use in combination with Compound 1 can comprise nucleoside and nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors and other antiviral drugs.

Example suitable NRTIs include zidovudine (AZT); didanosine (ddl); zalcitabine (ddC); stavudine (d4T); lamivudine (3TC); abacavir (1592U89); adefovir dipivoxil [bis(POM)-PMEA]; lobucavir (BMS-180194); BCH-10652; emitricitabine [(-)-FTC]; beta-L- FD4 (also called beta-L-D4C and named beta-L-2', 3'-dicleoxy-5-fluoro-cytidene); DAPD, ((- )-beta-D-2,6,-diamino-purine dioxolane); and lodenosine (FddA). Typical suitable NNRTIs include nevirapine (BI-RG-587); delaviradine (BHAP, U-90152); efavirenz (DMP-266); PNU-142721 ; AG-1549; MKC-442 (l -(ethoxy-methyl)-5-(l -methyl ethyl)-6-(phenylmethyl)- (2,4(1 H,3H)-pyrimidinedione); and (+)-calanolide A (NSC-675451) and B. Typical suitable protease inhibitors include saquinavir (Ro 31 -8959); ritonavir (ABT-538); indinavir (MK- 639); nelfnavir (AG-1343); amprenavir (141W94); lasinavir (BMS-234475); DMP-450; BMS-2322623; ABT-378; and AG-1 549. Other antiviral agents include hydroxyurea, ribavirin, IL-2, IL-12, pentafuside and Yissum Project No.11607.

Suitable agents for use in combination with Compound 1 for the treatment of cancer include chemotherapeutic agents, targeted cancer therapies, immunotherapies or radiation therapy. Compound 1 may be effective in combination with anti-hormonal agents for treatment of breast cancer and other tumors. Suitable examples are anti-estrogen agents including but not limited to tamoxifen and toremifene, aromatase inhibitors including but not limited to letrozole, anastrozole, and exemestane, adrenocorticosteroids (e.g. prednisone), progestins (e.g. megastrol acetate), and estrogen receptor antagonists (e.g. fulvestrant).

Suitable anti-hormone agents used for treatment of prostate and other cancers may also be combined with Compound 1. These include anti-androgens including but not limited to flutamide, bicalutamide, and nilutamide, luteinizing hormone-releasing hormone (LHRH) analogs including leuprolide, goserelin, triptorelin, and histrelin, LHRH antagonists (e.g. degarelix), androgen receptor blockers (e.g. enzalutamide) and agents that inhibit androgen production (e.g. abiraterone).

Compound 1 may be combined with or in sequence with other agents against membrane receptor kinases especially for patients who have developed primary or acquired resistance to the targeted therapy. These therapeutic agents include inhibitors or antibodies against EGFR, Her2, VEGFR, c-Met, Ret, IGFR1, or Flt-3 and against cancer-associated fusion protein kinases such as Bcr-Abl and EML4-Alk. Inhibitors against EGFR include gefitinib and erlotinib, and inhibitors against EGFR/Her2 include but are not limited to dacomitinib, afatinib, lapitinib and neratinib. Antibodies against the EGFR include but are not limited to cetuximab, panitumumab and necitumumab. Inhibitors of c-Met may be used in combination with FGFR inhibitors. These include onartumzumab, tivantnib, and INC-280. Agents against Abl (or Bcr-Abl) include imatinib, dasatinib, nilotinib, and ponatinib and those against Alk (or EML4-ALK) include crizotinib. Angiogenesis inhibitors may be efficacious in some tumors in combination with FGFR inhibitors. These include antibodies against VEGF or VEGFR or kinase inhibitors of VEGFR. Antibodies or other therapeutic proteins against VEGF include bevacizumab and aflibercept. Inhibitors of VEGFR kinases and other anti-angiogenesis inhibitors include but are not limited to sunitinib, sorafenib, axitinib, cediranib, pazopanib, regorafenib, brivanib, and vandetanib

Activation of intracellular signaling pathways is frequent in cancer, and agents targeting components of these pathways have been combined with receptor targeting agents to enhance efficacy and reduce resistance. Examples of agents that may be combined with Compound 1 include inhibitors of the PBK-AKT-mTOR pathway, inhibitors of the Raf-

MAPK pathway, inhibitors of JAK-STAT pathway, and inhibitors of protein chaperones and cell cycle progression.

Agents against the PI3 kinase include but are not limited topilaralisib, idelalisib, buparlisib. Inhibitors of mTOR such as rapamycin, sirolimus, temsirolimus, and everolimus may be combined with FGFR inhibitors. Other suitable examples include but are not limited to vemurafenib and dabrafenib (Raf inhibitors) and trametinib, selumetinib and GDC-0973 (MEK inhibitors). Inhibitors of one or more JAKs (e.g., ruxolitinib, baricitinib, tofacitinib), Hsp90 (e.g., tanespimycin), cyclin dependent kinases (e.g., palbociclib), HDACs (e.g., panobinostat), PARP (e.g., olaparib), and proteasomes (e.g., bortezomib, carfilzomib) can also be combined with Compound 1. In some embodiments, the JAK inhibitor is selective for JAK1 over JAK2 and JAK3.

Other suitable agents for use in combination with Compound 1 include chemotherapy combinations such as platinum-based doublets used in lung cancer and other solid tumors (cisplatin or carboplatin plus gemcitabine; cisplatin or carboplatin plus docetaxel; cisplatin or carboplatin plus paclitaxel; cisplatin or carboplatin plus pemetrexed) or gemcitabine plus paclitaxel bound particles (Abraxane®).

Suitable chemotherapeutic or other anti-cancer agents include, for example, alkylating agents (including, without limitation, nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) such as uracil mustard, chlormethine,

cyclophosphamide (Cytoxan™), ifosfamide, melphalan, chlorambucil, pipobroman, triethylene-melamine, triethylenethiophosphoramine, busulfan, carmustine, lomustine, streptozocin, dacarbazine, and temozolomide.

Other suitable agents for use in combination with Compound 1 include: dacarbazine (DTIC), optionally, along with other chemotherapy drugs such as carmustine (BCNU) and cisplatin; the "Dartmouth regimen," which consists of DTIC, BCNU, cisplatin and tamoxifen; a combination of cisplatin, vinblastine, and DTIC; or temozolomide. Compound 1 may also be combined with immunotherapy drugs, including cytokines such as interferon alpha, interleukin 2, and tumor necrosis factor (TNF) in.

Suitable chemotherapeutic or other anti-cancer agents include, for example, antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) such as methotrexate, 5-fluorouracil, floxuridine, cytarabine, 6-mercaptopurine, 6-thioguanine, fludarabine phosphate,

pentostatine, and gemcitabine.

Suitable chemotherapeutic or other anti-cancer agents further include, for example, certain natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) such as vinblastine, vincristine, vindesine, bleomycin, dactinomycin, daunorubicin, doxorubicin, epirubicin, idarubicin, ara- C, paclitaxel (TAXOL™), mithramycin, deoxycoformycin, mitomycin-C, L-asparaginase, interferons (especially IFN-a), etoposide, and teniposide.

Other cytotoxic agents include navelbene, CPT-11, anastrazole, letrazole,

capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.

Also suitable are cytotoxic agents such as epidophyllotoxin; an antineoplastic enzyme; a topoisomerase inhibitor; procarbazine; mitoxantrone; platinum coordination complexes such as cis-platin and carboplatin; biological response modifiers; growth inhibitors; antihormonal therapeutic agents; leucovorin; tegafur; and haematopoietic growth factors.

Other anti-cancer agent(s) include antibody therapeutics such as trastuzumab

(Herceptin), antibodies to costimulatory molecules such as CTLA-4, 4-lBB, PD-Ll and PD-1 antibodies, or antibodies to cytokines (IL-10, TGF-β, etc.).

Other anti-cancer agents also include those that block immune cell migration such as antagonists to chemokine receptors, including CCR2 and CCR4.

Other anti-cancer agents also include those that augment the immune system such as adjuvants or adoptive T cell transfer.

Anti-cancer vaccines include dendritic cells, synthetic peptides, DNA vaccines and recombinant viruses.

Methods for the safe and effective administration of most of these chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, the administration of many of the chemo therapeutic agents is described in the "Physicians' Desk Reference" (PDR, e.g., 1996 edition, Medical Economics Company, Montvale, NJ), the disclosure of which is incorporated herein by reference as if set forth in its entirety.

Pharmaceutical Formulations and Dosage Forms

When employed as pharmaceuticals, Compound 1 as described herein can be administered in the form of pharmaceutical compositions which refers to a combination of Compound 1 as described herein, and at least one pharmaceutically acceptable carrier. These compositions can be prepared in a manner well known in the pharmaceutical art, and can be administered by a variety of routes, depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and to mucous membranes including intranasal, vaginal and rectal delivery), pulmonary (e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), ocular, oral or parenteral. Methods for ocular delivery can include topical administration (eye drops), subconjunctival, periocular or intravitreal injection or introduction by balloon catheter or ophthalmic inserts surgically placed in the conjunctival sac. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal, or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Parenteral administration can be in the form of a single bolus dose, or may be, for example, by a continuous perfusion pump. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable.

This disclosure also includes pharmaceutical compositions which contain, as the active ingredient, Compound 1 in combination with one or more pharmaceutically acceptable carriers. In making the compositions described herein, the active ingredient is typically mixed with an excipient, diluted by an excipient or enclosed within such a carrier in the form of, for example, a capsule, sachet, paper, or other container. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of tablets, pills, powders, lozenges, sachets, cachets, elixirs, suspensions, emulsions, solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 10 % by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.

In preparing a formulation, the active compound can be milled to provide the appropriate particle size prior to combining with the other ingredients. If the active compound is substantially insoluble, it can be milled to a particle size of less than 200 mesh. If the active compound is substantially water soluble, the particle size can be adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.

Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrup, and methyl cellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions described herein can be formulated so as to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.

The compositions can be formulated in a unit dosage form, each dosage containing from about 5 to about 100 mg, more usually about 10 to about 30 mg, of the active ingredient. The term "unit dosage forms" refers to physically discrete units suitable as unitary dosages for human subj ects and other mammals, each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, in association with a suitable pharmaceutical excipient.

The active compound can be effective over a wide dosage range and is generally administered in a pharmaceutically effective amount. It will be understood, however, that the amount of the compound actually administered will usually be determined by a physician, according to the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound administered, the age, weight, and response of the individual patient, the severity of the patient's symptoms, and the like.

For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid pre-formulation composition containing a homogeneous mixture of Compound 1. When referring to these pre-formulation compositions as homogeneous, the active ingredient is typically dispersed evenly throughout the composition so that the composition can be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid pre-formulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present disclosure.

The tablets or pills of the present disclosure 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 permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.

The liquid forms in which the Compound 1 , or compositions as described herein can be incorporated for administration orally or by injection include aqueous solutions, 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.

Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. In some embodiments, the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in can be nebulized by use of inert gases. Nebulized solutions may be breathed directly from the nebulizing device or the nebulizing device can be attached to a face masks tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions can be administered orally or nasally from devices which deliver the formulation in an appropriate manner.

The amount of compound or composition administered to a patient will vary depending upon what is being administered, the purpose of the administration, such as prophylaxis or therapy, the state of the patient, the manner of administration, and the like. In therapeutic applications, compositions can be administered to a patient already suffering from a disease in an amount sufficient to cure or at least partially arrest the symptoms of the disease and its complications. Effective doses will depend on the disease condition being treated as well as by the judgment of the attending clinician depending upon factors such as the severity of the disease, the age, weight and general condition of the patient, and the like. The compositions administered to a patient can be in the form of pharmaceutical compositions described above. These compositions can be sterilized by conventional sterilization techniques, or may be sterile filtered. Aqueous solutions can be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the compound preparations typically will be between 3 and 11, more preferably from 5 to 9 and most preferably from 7 to 8. It will be understood that use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of pharmaceutical salts.

The therapeutic dosage of Compound 1 can vary according to, for example, the particular use for which the treatment is made, the manner of administration of the compound, the health and condition of the patient, and the judgment of the prescribing physician. The proportion or concentration of Compound 1 in a pharmaceutical composition can vary depending upon a number of factors including dosage, chemical characteristics (e.g., hydrophobicity), and the route of administration. For example, Compound 1 can be provided in an aqueous physiological buffer solution containing about 0.1 to about 10% w/v of the compound for parenteral administration. Some typical dose ranges are from about 1 μg/kg to about 1 g/kg of body weight per day. In some embodiments, the dose range is from about 0.01 mg/kg to about 100 mg/kg of body weight per day. The dosage is likely to depend on such variables as the type and extent of progression of the disease or disorder, the overall health status of the particular patient, the relative biological efficacy of the compound selected, formulation of the excipient, and its route of administration. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.

Compound 1 can also be formulated in combination with one or more additional active ingredients which can include any pharmaceutical agent such as anti-viral agents, vaccines, antibodies, immune enhancers, immune suppressants, anti -inflammatory agents and the like.

EXAMPLES Example 1

Synthesis of N-((2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihyd ro-l'H- spiro[cyclopropane-l,4'-[2,7]naphthyridine]-6'-yl)methyl)acr ylamide (Compound 1) Scheme 1

Step 1: 2'-(2, 6-Difluoro-3,5-dimethoxyphenyl)-6'-vinyl-l 2'-dihydro-3 'H-spiro[cyclo- propane-l, 4'-[2, 7]naphthyridin]- '-one (9)

Water (9.5 L), cesium fluoride (CsF, 3.0 kg, 19.75 mol), 1,4-dioxane (8.0 L), 6'- chloro-2'-(2,6-difluoro-3,5-dimethoxyphenyl)-l 2'-dihydro-3'H-spiro[cyclopropane-l,4'- [2,7]naphthyridin]-3'-one (2000 g, 5.25 mol) and 4,4,5,5-tetramethyl-2-vinyl-l ,3,2- dioxaborolane (971 g, 6.30 mol) were charged sequentially to a reactor with agitation. After the mixture was purged with nitrogen, [1,1 '- bis(diphenylphosphino)ferrocene]dichloropalladium (II), complex with dichloromethane (Pd(dppf)Cl2-CH2Cl2) (64 g, 0.08 mol) was charged to the reactor. After the reaction mixture was purged with nitrogen, it was heated to 85-95 °C and agitated at this temperature until reaction completion was indicated by HPLC. The reaction mixture was concentrated by vacuum distillation, then cooled to approximately 40-50 °C. Water (8.0 L) was charged to the reaction mixture. The mixture was cooled to 15-30 °C and stirred for no less than 1 hour. The solids were filtered, washed with n-heptane (10.0 L), partially dried to provide the desired compound (2772 g).

The crude product (7692 g, combined from two batches starting with 5.9 kg of starting chloride) was redissolved in DCM (41.3 L). The water phase was removed and the DCM solution containing the desired product was loaded onto a silica gel column (17.7 kg) with sea sand (5.9 kg) on top of the column. The column was eluted with DCM, followed by a mixture of ethyl acetate (EtOAc) and DCM. The desired fractions were combined and concentrated under reduced pressure to afford the desired compound (5386 g, 93.4% yield). LCMS calculated for C20H19F2N2O3 [M+H] + : 373.14; Found: 373.2; ¾ NMR (300 MHz, CDC1 3 ) 5 8.32 (s, 1 H), 6.80-6.60 (m, 3 H), 6.18 (m, 1 H), 5.45 (m, 1 H), 4.95 (s, 2 H), 3.90 (s, 6 H), 2.0 (m, 2 H), 1.35 (m, 2 H).

Steps 2 and 3: (E)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihyd ro- [cyclopropane- 1 ,4'-[2, 7]naphthyridine]-6'-carbaldehyde oxime (11)

To a solution of 2'-(2,6-Difluoro-3,5-dimethoxyphenyl)-6'-vinyl-r,2'-dihydro- 3'H- spiro[cyclo-propane-l,4'-[2,7]naphthyridin]-3'-one (1700 g, 4.57 mol) in tetrahydrofuran (THF, 30.1 L), were charged sodium periodate (NalCn, 3476 g, 16.25 mol) and water (8.7 L). A solution of osmium tetroxide (OsCn, 80 g, 0.32 mol) in THF (300 mL) was charged in portions to the reactor while maintaining the reaction temperature below 40 °C. The reaction was agitated at approximately 10-40 °C until reaction completion was indicated by HPLC. If necessary, additional Os04 in THF may be added. Ethyl acetate (EtOAc, 8.5 L) was charged to the reactor followed by sodium bicarbonate (NaHCC , 1534 g, 18.26 mol) in portions with agitation to adjust the pH of the resulting mixture to 6-8. The reaction mixture was filtered to remove solids. The reactor and filter cake were rinsed with additional EtOAc (17.0 L). The filtrate and rinsings containing the desired product were charged back to the reactor and the phases were separate. The organic phase was washed sequentially with aqueous NaHCC (238 g in 3.4 L water) and water (3.4 L). To the organic solution, with agitation,

hydroxylamine hydrochloride (698 g, 10.04 mol) was added in portions, followed by pyridine (1.1 L) and a methanol (0.9 L) rinse, maintaining the temperature below 40 °C. The reaction mixture was agitated at approximately 15-45 °C until reaction completion is indicated by HPLC. Methyl t-butyl ether (MTBE, 8.2 L) was added to the reactor and the reaction mixture was cooled to approximately 0-10 °C and stirred at this temperature for no less than 1 hour. The precipitated solid was filtered, washed with MTBE (5.4 L), dried to give the desired compound (1233.0 g).

The filtrate and MTBE rinse were combined and concentrated under reduced pressure. The residue was transferred to a reactor using MeOH (6.1 L). The mixture was cooled and stirred for no less than 1 hour. The precipitated solids were filtered, washed with MTBE (4.3 L) and dried to afford additional desired compound (439.6 g). Total amount of solids: 1672.6 g (94% yield) for two crops. LCMS calculated for C19H18F2N3O4 [M+H] + : 390.13; Found: 390.1 ; ¾ NMR (300 MHz, DMSO-c/e) δ 11.60 (s, 1 H), 8.42 (s, 1 H), 8.05 (s, 1 H), 7.22 (s, 1 H), 7.06 (m, 1 H), 4.96 (s, 2 H), 3.88 (s, 6 H), 1.75 (m, 2 H), 1.49 (m, 2 H).

Step 4: 6'-(aminomethyl)-2 '-(2, 6-difluoro-3, 5-dimethoxyphenyl)-l Ή-spirof cyclopropane- 1, 4 '- [ 2, 7]naphthyridin J-3 '(2 'H)-one diacetate (12 diacetate)

To an agitated mixture of (E)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'- dihydro- H-spiro-[cyclopropane-l ,4'-[2,7]naphthyridine]-6'-carbaldehyde oxime (1700 g, 4.37 mol) and acetic acid (HOAc, 42.5 L) cooled to approximately 15-25 °C, was charged zinc (Zn, 1656 g, 25.32 mol) powder in portions while maintaining the temperature at or below 25 °C. After the reaction was complete as indicated by HPLC, the mixture was filtered through Celite (850 g). The filter cake was rinsed with HOAc (3.4 L). The filtrate and washes were combined and concentrated under reduced pressure, adding n-heptane (17.0 L) to assist the distillation, to afford the desired amine as the diacetate salt, which is used in the next step without further purification.

Steps 5 and 6: 6'-(aminomethyl)-2'-(2, 6-difluoro-3, 5-dimethoxyphenyl)-l 'H- spiro [cyclopropane-1 , 4 '-[2, 7]naphthyridin]-3 '(2 'H)-one hydrochloride (12 HCl)

To a mixture of 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)- H- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'(2'H)-one diacetate (4560 g, 10.47 mol) in DCM (45.6 L) was charged aqueous sodium hydroxide (NaOH, 1823 g in water 22.8 L) solution while maintaining the reaction temperature at or below 30 °C. After the reaction mixture was agitated for no less than 30 minutes, phases were separated. If necessary, the mixture may be filtered through Celite to facilitate phase separation. The aqueous phase was extracted with additional DCM (36.5 L). The organic phases were combined, dried over magnesium sulfate (MgS04, 4560 g) and concentrated under reduced pressure to afford the free base (1532.6 g).

Alternatively, the diacetate was free based using aqueous ammonium hydroxide solution. To a turbid mixture of crude 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)- ,2'- dihydro-3'H-spiro[cyclopropane-l ,4'-[2,7]naphthyridin]-3'-one (9.70 g, 25.8 mmol) in methylene chloride (150.0 ml) was added water (50 ml, 2775 mmol). Solids precipitated. Concentrated ammonia in H2O (28%) (16.0 mL, 237 mmol) was added. Solids dissolved to give a two phase mixture. Organic phase was separated and washed with a premixed solution of ammonia in H2O (28%) (16.0 mL, 846 mmol) and water (50.0 mL). Aqueous phases were combined and back extracted with dichloromethane (50.0 ml). All organic phases were combined, washed with water (75.0 ml x 2) and concentrated to dryness to afford the free base, which after salt formation step afforded 10.5 g of hydrochloride salt.

To the 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)- H- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'(2'H)-one free base (3352 g, 8.93 mol) was added DCM (70.4 L). The resulting solution was cooled to approximately 15-30 °C. A solution of HCl in 1 ,4-dioxane (4 N, 2.46 L, 9.82 mol) was added to the solution while maintaining the reaction temperature at or below 30 °C. After stirred for no less than 1 hour, the solid was filtered, washed with DCM (25.1 L) and dried to afford the desired HC1 salt of the compound 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)-l Ή- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'(2'H)-one (3552 g, 96.6% yield, 97.55A% purity).

Steps 7,8 and 9: N-((2'-(2, 6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihydro-l Ή- spiro[cyclopropane-l,4'-[2, 7]naphthyridine]-6'-yl)methyl)acrylamide (Compound 1)

To a cooled mixture of 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)- H- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'(2'H)-one HC1 salt (1000 g, 2.43 mol) in DCM (15.0 L) was added aqueous sodium hydroxide (NaOH, 291 g, 7.28 mol, in 3.4 L of water) solution while maintaining the reaction temperature at or below 30 °C. The resulting mixture was agitated for no less than 1 hour and cooled to -10-10 °C. Acryloyl chloride (216 mL, 2.67 mol) was added. The mixture was agitated at approximately 0-10 °C until reaction completion was indicated by HPLC. The reaction mixture was warmed to rt and phases were separated. The aqueous phase was extracted with additional DCM (7.5 L). The organic phases were combined, washed with water and loaded onto a silica gel (15 kg) column with sea sand (5 kg) on top of the column. The column was eluted with a mixture of acetone and DCM. The desired fractions were combined and concentrated under reduced pressure to afford crude amide (782.7 g, 75% yield).

To a mixture of crude acrylamide (2675 g, 6.23 mol), acetonitrile (53.5 L), and water (18.7 L) at 30-40 °C was added aqueous NaOH (234.2 g, 5.86 mol, in 5.65 L of water) solution. The mixture was stirred at 30-40 °C until adequate removal of chloro impurity was demonstrated by HPLC. The reaction mixture was cooled and aqueous phosphoric acid (H3PO4, 85%, 321.5 g, 2.79 mol, diluted in 1.67 L of water) solution was slowly charged to adjust pH of the reaction mixture to between 8-10. The batch was agitated for 1 hour and concentrated under reduced pressure. Water (13.4 L) was charged to the reaction residue and the mixture was agitated. The solids were recovered and washed with water (2.7 L). The wet cake was returned to the reactor and reslurried in water (13.4 L) with agitation for no less than 30 minutes. The solid was recovered and the reslurry operation in water (13.4 L) was repeated one additional time. The solids were recovered and dried under vacuum to afford purified acrylamide (2962.9 g). The solid (2950 g, 6.87 mol) was reslurried in acetone (17.7 L) at 40-60°C for no less than 1 hour. After cooled to 0-10 °C, the solids were recovered by filtration followed with washing with cooled acetone (3.0 L).

The recovered solids were dissolved in acetone (59.0 L)/ USP grade purified water (17.7 L) at 40-60 °C and stirred with activated charcoal (590 g) for 0.5-2 hours. While hot, it was filtered through a bed of Celite (590 g). The Celite bed was rinsed with a hot mixture of acetone (6.64 L) and water (2.21 L). The filtrate and washes were combined and polish filtered into a clean reactor. The mixture was concentrated under reduced pressure and cooled to the room temperature. The solids were filtered, washed with filtered USP grade purified water (14.8 L) and dried under vacuum at 40-50 °C to afford the final compound N- ((2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihydro - H-spiro[cyclopropane-l,4'- [2,7]naphthyridine]-6'-yl)methyl)acrylamide (2301 g, 78% yield). The compound isolated was determined to be Form I.

LC-MS calculated for C22H22F2N3O4 [M+H] + m/z: 430.15; found 430.1 ; ¾ NMR (500 MHz, DMSO-c/e) δ 8.58 (t, J= 5.7 Hz, 1H), 8.36 (s, 1H), 7.06 (t, J= 8.2 Hz, 1H), 6.89 (s, 1H), 6.32 (dd, J= 17.1 , 10.2 Hz, 1H), 6.11 (dd, J = 17.1, 2.1 Hz, 1H), 5.61 (dd, J= 10.2, 2.1 Hz, 1H), 4.94 (s, 2H), 4.41 (d, J= 5.8 Hz, 2H), 3.89 (s, 6H), 1.74 (dd, J= 3.9, 3.9 Hz, 2H), 1.43 (dd, J = 4.0, 4.0 Hz, 2H); 13 C NMR (150 MHz, DMSO-c/e) δ 168.7, 164.6, 157.6, 145.6, 145.1, 143.5, 141.3, 131.6, 125.4, 125.0, 119.7, 114.4, 100.0, 56.9, 49.2, 44.1, 23.6, 20.4; 19 F NMR (376 MHz, DMSO-c/e) δ -150.9.

Alternative synthesis of 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphi spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'(2'H)-one (12)

tert-butyl (2'-(2, 6-difluoro-3, 5-dimethoxyphenyl)-3'-oxo-2 ', 3 '-dihydro-1 Ή- spirof cycloprop ne-1, 4 '-[2, 7]naphthyridine ]-6'-yl)methylcarbamate (15)

A degassed mixture of 6'-chloro-2'-(2,6-difluoro-3,5-dimethoxyphenyl)-r,2'-dihydro - 3'H-spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'-one (2000.0 mg, 5.25 mmol), potassium N- Boc-aminomethyltrifluoroborate (3213 mg, 13.55 mmol), cesium carbonate (7838 mg, 24.06 mmol), cataCXium Pd G4 (116 mg, 0.157 mmol), water (20.0 ml, 1110 mmol) and dioxane (20.0 ml) was heated at 85 °C for 3 hours. The reaction was allowed to cool to room temperature. The organic phase was separated and heptane (10 mL) was added, followed by MTBE (10 mL). The mixture was stirred for 30 min. The solids were filtered, dried under vacuum at 50 °C overnight to give 1.78 g of the desired product as a brown solid. ¾ NMR (400 MHz, CDCls) δ 8.26 (s, 1H), 6.68 - 6.59 (m, 2H), 5.43 (s, 1H), 4.90 (s, 2H), 4.37 (d, J = 5.5 Hz, 2H), 3.87 (s, 6H), 2.00 (m, J= 3.6 Hz, 2H), 1.64 (s, 1H), 1.44 (s, 9H), 1.36 (q, J= 4.2 Hz, 2H).

6'-(aminomethyl)-2 '-(2, 6-difluoro-3, 5-dimethoxyphenyl)-l Ή-spirof cyclopropane-1, 4 '- [2, 7]naphthyridin]-3'(2'H)-one dihydrochloride (12 2HCI)

Into a 500mL round bottom with stir bar was charged tert-butyl { [2'-(2,6-difluoro-3,5- dimethoxyphenyl)-3'-oxo-2',3'-dihydro- H-spiro[cyclopropane-l,4'-[2,7]naphthyridin]- yl]methyl} carbamate (14.7 g, 30.9 mmol) and methylene chloride (250.0 mL) at room temperature to give a thin slurry. 4.0 M Hydrogen chloride in dioxane (59.1 mL, 236 mmol) was added at room temperature. The mixture was stirred for four hours. The solids were filtered, washed with a generous volume of DCM and dried on the filter then overnight under vacuum to give 14.14 g of the desired bis-HCl salt (102% yield). ¾ NMR (400 MHz, DMSO-c/e) δ 8.73 (s, 3H), 8.52 (s, 1H), 7.45 (s, 1H), 7.08 (t, J= 8.2 Hz, 1H), 6.81 (s, 1H), 5.01 (s, 2H), 4.32 - 4.08 (m, 2H), 3.89 (s, 6H), 1.82 (q, J= 3.9 Hz, 2H), 1.59 (q, J= 4.0 Hz, 2H).

Alternative synthesis of 6'-(aminomethyl)-2'-(2,6-difluoro-3,5-dimethoxyphenyl)- H- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'(2'H)-one (12)

2 '-(2, 6-difluoro-3, 5-dimethoxyphenyl)-3 '-oxo-2 ' 3 '-dihydro-1 Ή-spirof cyclopropane-1, 4 '- [2, 7]naphthyridine]-6'-carbonitrile (14)

A degassed mixture of 6'-chloro-2'-(2,6-difluoro-3,5-dimethoxyphenyl)- ,2'-dihydro- 3'H-spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'-one (2.25 g, 5.91 mmol), zinc cyanide (1.39 g, 11.8 mmol), dppf (0.15 g, 0.26 mmol), Pd(PPh 3 ) 4 (0.1 g, 0.089 mmol) and N,N- dimethylacetamide (25 mL, 270 mmol) was stirred at 100 °C for 1 hour. The reaction was cooled to room temperature and filtered through a pad of Celite. The Celite bed was washed with 25 mL of DMAC. The filtrate and washes were combined and cooled to 5 °C and 100 mL of water was added. The resulting yellow slurry was stirred for 1.5 hours and filtered. The solids were washed with water (3 x 30 mL) and heptane (3 x 20 mL) and dried to give 2.3 g of the desired crude product. LC-MS calculated for C19H16F2N3O3 [M+H] + m/z:

372.11 ; found 372.1. ¾ NMR (400 MHz, DMSO-c/e) δ 8.55 (s, 1H), 7.70 (s, 1H), 7.08 (t, J = 8.2 Hz, 1H), 5.06 (s, 2H), 3.89 (s, 6H), 1.79 (q, J= 4.0, 3.6 Hz, 2H), 1.64 (q, J= 4.1 Hz, 2H).

6'-(aminomethyl)-2 '-(2, 6-difluoro-3, 5-dimethoxyphenyl)-l Ή-spirof cyclopropane-1, 4 '- [2, 7]naphthyridin] -3'(2'H)-one (12)

2'-(2,6-difluoro-3,5-dimethoxyphenyl)-3'-oxo-2',3'-dihydro- H-spiro[cyclopropane- l,4'-[2,7]naphthyridine]-6'-carbonitrile (37.1 mg, 0.100 mmol) in EtOH (2.0 ml) was stirred under N2 at room temperature. Potassium borohydride (32.3 mg, 0.600 mmol) and Raney Nickel (11.7 mg, 0.200 mmol) were added. The reaction was stirred at room temperature for 15 minute and then 60 °C for 4 hours. The mixture was cooled to room temperature and filtered through a pad of Celite. The Celite was washed with DCM. The filtrate and washes were combined and concentrated to give crude product. LC-MS calculated for C19H20F2N3O3 [M+H] + m/z: 376.1 ; found: 376.1.

Alternative purification of 6'-(aminomethyl)-2'-(2, 6-difluoro-3,5-dimethoxyphenyl)- -spiro [cyclopropane- 1 ,4'-[2, 7]naphthyridin]-3'(2'H)-one (12)

tert-butyl (2'-(2, 6-difluoro-3, 5-dimethoxyphenyl)-3'-oxo-2 ', 3 '-dihydro-1 Ή- spirof cyclopropane-1, 4 '-[2, 7]naphthyridine ]-6'-yl)methylcarbamate (15)

Into a 1L round bottom with stir bar was charged [A] 6'-(aminomethyl)-2'-(2,6- difluoro-3,5-dimethoxyphenyl)- ,2'-dihydro-3'H-spiro[cyclopropane-l,4'-[2,7]naphthyridin]- 3'-one acetate (17.5 g, 40.2 mmol) and Tetrahydrofuran (350.0 mL) to give a slurry. 2.0 M Sodium hydroxide in water (60.3 mL, 1.20E2 mmol) was added and the resulting mixture was stirred for 5 minutes to give a homogeneous solution. Di-tert-Butyldicarbonate (17.5 g, 80.4 mmol) was added at room temperature in one portion. The reaction was complete after 2 hours as indicated by HPLC. The reaction was diluted with water (350.0 mL) and ethyl acetate (350.0 mL). After phase separation, the aqueous fraction was extracted with ethyl acetate (100.0 mL). The organic extracts were combined, dried over MgS04, filtered and concentrated to a thick slurry. MTBE was added and the mixture was concentrated again to a fairly thick slurry. Heptane was added. The mixture was stirred at room temperature for 1 hour. The solids were filtered, rinsed with fresh heptane and dried under vacuum to give 15.3 g of off-white solids (80.1 % yield). ¾ NMR (400 MHz, CDCb) δ 8.26 (s, 1H), 6.68 - 6.59 (m, 2H), 5.43 (s, 1H), 4.90 (s, 2H), 4.37 (d, J= 5.5 Hz, 2H), 3.87 (s, 6H), 2.00 (m, J = 3.6 Hz, 2H), 1.64 (s, 1H), 1.44 (s, 9H), 1.36 (q, J= 4.2 Hz, 2H).

Synthesis of 6'-chloro-2'-(2,6-difluoro-3,5-dimethoxyphenyl)-r,2'-dihydro -3'H- spiro[cyclopropane-l,4'-[2,7]naphthyridin]-3'-one (8)

N-[ ( 4, 6-dichloropyridin-3-yl)methyl ]-2, 6-difluoro-3, 5 -dimethox aniline

Procedure 1 :

To a stirred solution of 4,6-dichloronicotinaldehyde (105.7 g, 546.5 mmol) in anhydrous THF (200 mL) under N2 was added 2,6-difluoro-3,5-dimethoxyaniline (109.6 g, 573.8 mmol) and dibutyltin dichloride (3.46 g, 10.9 mmol). After stirred 6 min at rt, phenylsilane (65.1 g, 601.2 mmol) was added in portions, and the mixture was stirred at rt for 48 h. The reaction mixture was diluted with hexanes (1400 mL), stirred for 2 h, and the resulting precipitate was filtered to afford the crude product (141 g). The product was purified by filtration via a pack of silica gel on a disposable funnel eluting with EtOAc to afford the desired product (138.6 g, 72.6%). LC-MS calculated for C14H13CI2F2N2O2 m/z [M+H] + ; 349; found 349.

Procedure 2:

To a stirred solution of 2,6-difluoro-3,5-dimethoxyaniline (10.09 g, 53.3 mmol) and 4,6-dichloronicotinaldehyde (10.0 g, 52.3 mmol) and DMF (150 ml) in oven-dried 100 mL RBF under N2 in a water bath was added TMSC1 (16.70 ml, 131 mmol) drop wise at rt over 15 minutes to give a solution. The mixture was stirred at room temperature for 2.5 hours to give a thick slurry. Borane in THF (1.0 M, 52.3 ml, 52.3 mmol) was added. The mixture was stirred at room temperature for 3 hours and cooled to 0 °C. Water (110 ml) was added and the mixture was stirred for 20 minutes. Ammonium hydroxide (14.2 ml, 89 mmol) in water (21.6 ml, 1197 mmol) was added drop wise to the reaction and the mixture was stirred for additional 20 minutes. Water (184 ml) was added and the stirring was continued. The solid was filtered, washed sequentially with water (3 x 15 mL) and heptane (2 x 15 mL) and dried under vacuum at 50 °C to give the crude product (17.1 g, 94% yield, 99.55A% purity). Procedure 3 :

To a stirred solution of 2,6-difluoro-3,5-dimethoxyaniline (9.03 g, 47.7 mmol) and sodium triacetoxyborohydride (38.0 g, 180 mmol) in methylene chloride (60 mL)

/trifluoroacetic acid (30 mL) was added 4,6-dichloronicotinaldehyde (8.00 g, 45.5 mmol) in small portions at room temperature. After 1 hour, the volatiles were removed under vacuo and saturated aqueous NaHCC (200 mL) was added. The resulting mixture was extracted with DCM (3 x 150 mL). The organic layers were combined, dried over Na2S04, and concentrated. The residue was purified on silica gel (eluting with 0 to 0-40% EtOAc in hexanes) to afford the desired product (15.0 g). LC-MS calculated for C14H13CI2F2N2O2 [M+H] + m/z: 349.0; found 349.1.

Procedure 4:

(E)-l-(4,6-dichloropyridin-3-yl)-N-(2,6-difluoro-3,5-dimetho xyphenyl)met n-imine

(4A).

4,6-Dichloronicotinaldehyde (1293 g, 7.347 moles, 1.00 equiv) and acetic acid (1300 mL, 22.86 mol, 3.1 equiv) were sequentially added to a solution of 2,6-difluoro-3,5- dimethoxyaniline (1409 g, 7.449 mol, 1.014 equiv) in ethanol (9 L) at room temperature. After stirring for 18 hours, the solids were collected by filtration and rinsed with ethanol (4 L). The filtrates were concentrated under reduced pressure to a volume of 3L. The solids were collected by filtration and rinsed with ethanol (2 L). The filtrates were concentrated under reduced pressure to a volume of 1.5 L. The solids were collected by filtration and rinsed with ethanol (1 L). The three batches of solids were dried separately under vacuum at 40 °C overnight to give the desired compound (1509 g, 360 g and 419 g respectively). The three batches were combined and dry blended to give the desired compound (2287 g, 89.7% yield, >97% purity). LCMS calculated for C14H11CI2F2N2O2 (M+H): 347.02; Found: 347.1. ¾ NMR (300 MHz, CDC1 3 ) 5 9.23 (s, 1 H), 8.95 (s, 1 H), 7.45 (s, 1 H), 6.50 (m, 1H), 3.90 (s, 6H).

N-((4, 6-dichloropyridin-3-yl)methyl)-2, 6-difluoro-3,5-dimethoxyaniline (4)

Solid sodium borohydride (88 g, 2.33 moles, 1.1 equiv) was added portionwise keeping temperature between 27-33 °C to a solution of compound (E)-l -(4,6-dichloropyridin- 3-yl)-N-(2,6-difluoro-3,5-dimethoxyphenyl)methan-imine (732 g, 2.11 moles, 1.0 equiv) in dichloromethane (3.65 L) and methanol (1.8 L) at 8 °C. The reaction mixture was stirred at room temperature overnight, at which point LCMS indicated the reaction was complete. The reaction was diluted with dichloromethane (1.5 L) and water (3.0 L) and stirred for one hour. The organic layer was separated and the aqueous layer was back-extracted with dichloro- methane (2 x 1 L). The combined organic layers were concentrated under reduced pressure to give crude product (739.5 g, over theory), which was used subsequently. LCMS calculated for C14H13CI2F2N2O2 (M+H): 349.03 ; Found: 349.2. ¾ NMR (300 MHz, CDCI3) δ 8.32 (s, 1 H), 7.35 (s, 1 H), 6.10 (m, 1 H), 4.62 (s, 2H), 4.25 (s, 1H), 3.85 (s, 6H). ethyl 3-[[(4, 6-dichloropyridin-3-yl)methyl](2, 6-difluoro-3, 5- dimethoxyphenyl)

oxopropanoate (5)

Procedure 1 :

To a stirred solution of N-[(4,6-dichloropyridin-3-yl)methyl]-2,6-difluoro-3,5- dimethoxyaniline (138.6 g, 397.0 mmol) in THF (500 mL) was added NaH (60% w/w in mineral oil, 10.0 g, 417 mmol) in portions at rt. After stirred at rt forl 5 minutes, the solution was cooled to 0 °C, and ethyl malonyl chloride (92.5 g, 614.4 mmol) was added dropwise under N 2 . The ice-bath was removed after 10 min, and the reaction mixture was stirred at rt for 1.5 h. The reaction mixture was diluted with DCM and was quenched with saturated aqueous NH4CI, and extracted with DCM (3 χ 1000 mL). The organic layers were combined, washed with 5% NaHCCb solution, dried over Na 2 S04, and concentrated. The crude was added 25% MTBE in hexane, and colorless solid formed. Filtration gave the product as a colorless crystalline solid (177.5 g, 96.5%). LC-MS calculated for C19H19CI2F2N2O5 m/z [M+H] + : 463; found 463.

Procedure 2:

To a stirred solution of N-[(4,6-dichloropyridin-3-yl)methyl]-2,6-difluoro-3,5- dimethoxyaniline (138.6 g, 397.0 mmol) and triethylamine (44 g, 430 mmol) in DCM (500 mL) was added ethyl malonyl chloride ( 65 g, 430 mmol) in dropwise at rt. After stirred at rt for 10 minutes, LC-MS showed the completion of the reaction. The reaction mixture was partitioned between DCM and 1.0 N HC1 solution. The organic layer was washed with saturated NaHCCb solution, brine, dried and concentrated to give the crude oil product. Treatment with 25% MTBE in hexane gave the product (131 g, 98.7% yield) for the next step.

Procedure 3 :

Ethyl malonyl chloride (477 g, 3.17 moles, 1.5 equiv) was added dropwise at room temperature to a mixture of crude N-[(4,6-dichloropyridin-3-yl)methyl]-2,6-difluoro-3,5- dimethoxyaniline (739.5 g, 2.11 moles, 1 equiv) and sodium bicarbonate (550 g, 6.55 moles, 3.1 equiv) in dichloromethane (3 L). After stirring for 1 hour, water (3 L) was added and the layers were separated. The aqueous layer was back extracted with dichloromethane (1 L). The combined organic layers were concentrated under reduced pressure. The crude product was purified by over silica gel (6.5 kg), eluting with a gradient of 20 to 50% ethyl acetate in heptanes to give the desired product (754.5 g, 77% yield, >95% purity). LCMS calculated for C19H19CI2F2N2O5 [M+H] + : 463.06; Found: 462.9. ¾ NMR (300 MHz, CDCI3) δ 8.35 (s, 1 H), 7.30 (s, 1 H), 6.67 (m, 1 H), 5.02 (s, 2H), 4.10 (m, 2H), 3.82 (s, 6H), 3.23, (s, 2H), 1.21 (m, 3H).

6-chloro-2-(2, 6-difluoro-3, 5-dimethoxyphenyl)-3-oxo-l , 2, 3, 4-tetrahydro-2, 7-naphthyridine- 4-carboxylate (6)

Procedure 1 :

A mixture of ethyl 3-[[(4,6-dichloropyridin-3-yl)methyl](2,6-difluoro-3,5- dimethoxyphenyl)amino]-3-oxopropanoate (36.0 g, 77.7 mmol), Cu(I)I (2.80 g, 14.7 mmol), CS2CO3 (70.0 g, 215 mmol), and 2-pyridinecarboxylic acid (7.23 g, 58.7 mmol) in DMSO (204 mL) was stirred and degassed with N2 three times, then heated to 120 °C for 2 h, HPLC indicated no starting material left. The heat was removed, the reaction was cooled to rt, and then slowly filtered the reaction mixture into 500 mL of aqueous 2N HC1 in ice-bath with stirring, and rinsed with more 2N HC1 solution. The resulting solid in aqueous HC1 solution was filtered, washed with water (3 x 300 mL), dried in air to afford the crude product. The crude was stirred and slurried in 60% EtOAc in hexane (400 mL). Filtration, washed with more 60% EtOAc in hexane gave the product as an off-white solid (28.6 g, 86.2% yield). LC-MS calculated for C19H18CIF2N2O5 m/z [M+H] + : 427; found 427.

Procedure 2:

Into a 2L, 4-neck round bottom flask equipped with stir bar, thermocouple, condenser and 3-way valve were charged ethyl 3-[[(4,6-dichloropyridin-3-yl)methyl](2,6-difluoro-3,5- dimethoxyphenyl)amino]-3-oxopropanoate (107.9 g, 232.9 mmol), dimethyl sulfoxide (647 mL) and K3PO4 (173 g, 813 mmol) to give a slurry. After degassed 4x backfilling with nitrogen each time, the reaction mixture was stir at 80 °C overnight and 90 °C for 1 hour, at which time the reaction was complete as indicated by HPLC. The reaction was allowed to cool to room temperature and slowly added in portions over 52 minutes to a cooled 1.0 M Hydrogen chloride in water (1295 mL, 1295 mmol) in a 5L, 4-neck round bottom flask with overhead stirring, while maintaining the temperature < 15 °C. Following the addition, the slurry was stirred for 90 minutes in the ice bath. The solid was filtered, washed with water (1000 mL) and dried on the filter under vacuum overnight to afford 141.2 g of tan solids. The solids were redissolved in methylene chloride (720 mL) and ethyl acetate (500 mL). The resulting solution was washed with water (1000 mL x 2) and concentrate on a rotovap under mild vacuum (~200mbar) to 314 g. Heptane (3500 mL) was added dropwise over 35 minutes. The slurry was stirred overnight at room temperature. The solids were filtered, washed with a solution of ethyl acetate (135 mL) and heptane (180 mL) and dried on the funnel under vacuum to afford 88 g (88.5% yield) of yellow solids.

6-chloro-2-(2,6-difluoro-3,5-dimethoxyphenyl)-l,2-dihydro -2 -na (7)

Into 5L, 4-neck round bottom flask with overhead stirring, thermocouple, heating mantle, condenser and nitrogen inlet were charged ethyl 6-chloro-2-(2,6-difluoro-3,5- dimethoxyphenyl)-3-oxo-l ,2,3,4-tetrahydro-2,7-naphthyridine-4-carboxylate (227.1 g, 532.1 mmol) and 1 ,4-dioxane (900 mL) followed by 12.0 M hydrogen chloride in water (448 mL, 5380 mmol) and water (1695 mL). The reaction mixture was stirred at 80 °C for 6 hours and then allowed to cool to room temperature and stirred overnight. The reaction was complete after stirring overnight as indicated by HPLC. The mixture was cooled in an ice bath to 7.5 °C and the pH was adjusted to 8 with a solution of 6.0 M Sodium hydroxide in water (941 mL, 5646 mmol) added dropwise over 75 minutes via an addition funnel, while maintaining the internal temperature < 15 °C. After the pH adjustment, the cold bath was removed and water (1695 mL) was added dropwise via addition funnel over 17 minutes. The mixture was stirred at room temperature for 3.5 hours. The solids were filtered, washed with water (1695 niL), dried on the filter and then at 50 °C in vacuum oven overnight to afford 180.5 g (85.1% yield) of tan/light yellow solid. Purity was 99.2%.

6'-chloro-2 '-(2, 6-difluoro-3, 5-dimethoxyphenyl)-l ' 2 '-dihydro-3 Ή-spiro [cyclopropane- 1 , 4 '- [2, 7]naphthyridin ]-3 '-one (8)

Into a 5L, 3-neck round bottom flask equipped with stir bar, thermocouple, addition funnel and nitrogen inlet were charged 6-chloro-2-(2,6-difluoro-3,5-dimethoxyphenyl)-l,4- dihydro-2,7-naphthyridin-3(2H)-one (189.8 g, 519.0 mmol), N,N-dimethylformamide (570 mL) and cesium carbonate (377.3 g, 1157 mmol) under nitrogen. The mixture was cooled in an ice bath to 12 °C. l-Bromo-2-chloroethane (130.6 g, 892.1 mmol) was charged via the addition funnel dropwise over 12 minutes. Internal temperature rose to 19.5 °C at the end of the addition. After the addition, the cold bath was removed and the reaction was stirred at room temperature for 3 hours and 30 °C for 1 hour. Additional l-bromo-2-chloroethane (5.0 mL, 60.0 mmol) was added and the reaction was stirred for additional 5 hours at 30 °C and overnight at room temperature. Additional cesium carbonate (12.0 g, 36.8 mmol) was charged and the reaction was stirred at 35 °C for 2 hours followed 40 °C for 4 hours. The reaction was found to be complete by HPLC.

To the reaction mixture was added heptane (750 mL) followed by water (2300 mL). The mixture was stirred for 1 hour. The solids were filtered, washed water (2300 mL), dried on the filter under vacuum to afford 207 g (105%) of the desired product, which was used in the next reaction without further purification.

Preparation of 4,6-Dichloronicotinaldehyde (2)

1 A 1 B 1 C 2, 4-Dichloro-3-( trimethylsilyl)pyridine ( IB

A solution of 2,4-dichloropyridine (650 g, 4.39 mol, 1 equiv) in THF (2 L) was added dropwise over one hour, while maintaining the internal temperature below -65 °C, to a solution of 2.5 M ft-butyllithium in hexanes (1.95 L, 4.875 mol, 1,1 equiv) in THF (5.2 L) at - 78 °C. After stirring at -78 °C for 30 minutes, trimethylchlorosilane (580 g, 680 mL, 5.34 mol, 1.2 equiv) was added over one minute. The internal temperature increased to -50 °C. The reaction mixture was stirred cold for 10 minutes and quenched with 20% aqueous ammonium chloride (4.3 L) and saturated brine (1.45 L). The product was extracted with MTBE (2 x 6 L). The combined organic layers were washed with brine ( 2x 0.9 L) and concentrated under reduced pressure. The residue was purified over silica gel (4 kg), eluting witth a gradient of 0 to 20% ethyl acetate in heptanes, to give the desired compound (841 g, yield 87%, purity 97%) as a dark oil. LCMS calculated for C 8 Hi 2 Cl 2 NSi [M+H] + : 220.01; Found: 219.9. ¾ NMR (300 MHz, CDCls) δ 8.20 (d, 5.27 Hz, 1 H), 7.20 (d, 5.27 Hz, 1 H), 0.52 (s, 9 H).

4, 6-Dichloro-5-(trimethylsilyl)nicotin

2.5 M ft-Butyllithium in hexane (1540 mL, 3.85 mol, 1.4 equiv) was added over 10 minutes, while maintaining temperature below -10 °C, to a solution of 2,2,6,6- tetramethylpiperidine (568 g, 4.02 mol, 1.46 equiv) in THF (5 L) at -40 °C. The reaction mixture was allowed to warm-up to 0 °C and was stirred at this temperature for 30 minutes. The mixture was cooled to -78 °C and a solution of 2,4-dichloro-3-(trimethylsilyl)pyridine (604 g, 2.74 mol, 1 equiv) in THF (2.9 L) was added over 30 minutes maintaining the internal temperature below -70 °C. The reaction was stirred at -78 °C for an additional 90 minutes. 4- Formylmorpholine (552 g, 4.8 mol. 1.75 equiv) was added over 30 minutes, while maintaining the internal temperature below -70 °C, and the reaction was stirred at -78 °C for 30 minutes. The reaction was quenched with IN HC1 and the product was extracted with MTBE (2 x 8 L). The combined organic layers were washed with saturated brine (2 x 4 L) and the solvent was removed under reduced pressure. The crude product was purified by over silica gel (3 kg), eluting with a gradient of 0 to 20% ethyl acetate in heptanes, to give the desired compound (538 g, 79% yield, 95% purity) as light yellow oil which partialy solidified upon standing. LCMS calculated for C9H12CI2NOS- [M+H] + : 248.01 ; Found: 248.1. ¾ NMR (300 MHz, CDCb) δ 10.49 (s, 1 H), 8.74 (s, 1 H), 0.57 (s, 9 H).

4, 6-Dichloronicotinaldehyde (2)

A solution of 4,6-dichloro-5-(trimethylsilyl)-nicotinaldehyde (1112 g, 4.48 mol, 1 equiv) in THF (8.8 L) and water (4.4 L) was cooled in ice/water bath to 5 °C.

Tetrabutylammonium fluoride trihydrate (1556 g, 4.92 mol, 1.1 equiv) was added portion- wise over 5 minutes. THF (2 L) was used to rinse reagents into the flask. A mild exothermic effect was observed (temperature increased to 15 °C). The cooling bath was removed and the reaction mixture was stirred at room temperature overnight, at which point LCMS indicated the reaction was complete. Water (11 L) was added and the product was extracted with ethyl acetate (3 x 12 L). The combined organic layers were washed with 10% sodium bicarbonate (10 L) and saturated brine (10 L). The solvent was evaporated under reduced pressure and the residue was purified over silica gel (3 kg), eluting with a gradient of 0 to 6% ethyl acetate in heptanes. The product was dried under vacuum at 30 °C for one hour to give the desired compound (596 g, 75% yield, 99% purity) as white solid. LCMS calculated for CeHeChNC [M+H 2 0+H] + : 193.98; Found: 194.1. ¾ NMR (300 MHz, CDCI3) δ 10.44 (s, 1 H), 8.85 (s, 1 H), 7.49 (s, 1 H).

Alternative syntheis of 4,6-Dichloronicotinaldehyde (2)

1 2

4, 6-Dichloronicotinaldehyde (2)

To a stirred solution of 2,4-dichloro-5-carbethoxypyridine (199.0 g, 904.3 mmol) in methylene chloride (2000.0 mL) at -78 °C under N2 was added a solution of

diisobutylaluminum hydride in methylene chloride (994.8 mL, 1.0 M, 994.8 mmol) dropwise over 85 min. After 2 h, LCMS indicated the reaction was complete. The reaction was quenched with aqueous 2 N HC1 solution (600 mL), warmed up to room temperature, and stirred for 30 min. The organic layer was separated, and the aqueous layer was extracted with DCM (300 ml). The combined organic layers were washed with water (500 mL), dried over Na2S04 and concentrated in vacuo to afford the crude aldehyde (161.5 g with 91.7% purity), which was used in the next step without further purification. LC-MS calculated for

C6H4CI2NO m/z [M+H] + : 176; found 176.

Example 2

Preparation and Characterization of Form I

In one experiment, Form I was prepared according to the procedures in Example 1.

Form I was characterized by XRPD. The XRPD was obtained from Rigaku MiniFlex X-ray Powder Diffractometer (XRPD). The general experimental procedures for XRPD were: (1) X-ray radiation from copper at 1.054056 A with Κβ filter; (2) X-ray power at 30 KV, 15 mA; and (3) the sample powder was dispersed on a zero-background sample holder. The general measurement conditions for XRPD were: Start Angle 3 degrees; Stop Angle 45 degrees;

Sampling 0.02 degrees; and Scan speed 2 degree/min. The XRPD pattern is shown in Figure 1 and the XRPD data are provided in the table below.

Table 1

20.4 85 8.1

21.1 142 13.5

21.6 49 4.7

22.6 83 7.9

23.3 1053 100

24.2 342 32.5

24.7 446 42.4

25.5 47 4.5

26.5 147 14

27.1 374 35.5

28.1 42 4

28.6 67 6.4

29.3 196 18.6

30.1 45 4.3

31.7 81 7.7

32.2 120 11.4

33.8 51 4.8

35.1 42 4

36.7 115 10.9

37.3 69 6.6

38.8 38 3.6

40.1 41 3.9

41.4 240 22.8

42.2 73 6.9

42.6 179 17

43.3 40 3.8

43.8 42 4

Form I was characterized by DSC. The DSC was obtained from TA Instruments Differential Scanning Calorimetry, Model Q200 with autosampler. The DSC instrument conditions were as follows: 30 - 300°C at 10°C/min; Tzero aluminum sample pan and lid; and nitrogen gas flow at 50 mL/min. The DSC thermogram is shown in Figure 2. The DSC thermogram revealed an endothermic event at an onset temperature of 206.9 °C with a peak temperature of 207.7 °C.

Form I was characterized by TGA. The TGA was obtained from TA Instrument Thermogravimetric Analyzer, Model Q500. The general experimental conditions for TGA were: ramp from 20°C to 600 °C at 20°C/min; nitrogen purge, gas flow at 40 mL/min followed by balance of the purge flow; sample purge flow at 60 mL/min; platinum sample pan. The TGA thermogram is shown in Figure 3.

Example 3

Preparation and Characterization of Form II In one experiment, Form II was prepared by adding about 30 mg of Form I to about 3 mL of saturated or cloudy solution of Form I prepared in chloroform followed by stirring at 22 ± 1 °C for 3 days, which was filtered. Form II was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Examples 15 and 19.

Form II was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD partem is shown in Figure 4 and the XRPD data are provided in the table below.

Table 2

Form II was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 5. The DSC thermogram revealed an endothermic event at an onset temperature of 206.5 °C with a peak temperature of 207.8 °C. Example 4

Preparation and Characterization of Form III

In one experiment, Form III was prepared as follows. To about 3 mL of saturated or cloudy solutions of Form I prepared in 1,4-dioxane was added about 30 mg of Form I followed by stirring at 22 ± 1 °C for 3 days, which was filtered. Form III was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Examples 15, 19, and 20.

Form III was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattem is shown in Figure 6 and the XRPD data are provided in the table below.

Table 3

Form III was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 7. The DSC thermogram revealed an endothermic event at an onset temperature of 205.5 °C with a peak temperature of 206.9 °C Example 5

Preparation and Characterization of Form IV

In one experiment, Form IV was prepared as follows. To about 3 mL of saturated or cloudy solutions of Form I prepared in toluene was added about 30 mg of Form I followed by stirring at 25 ± 1 °C for 3 days, which was filtered. Form rv was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Example 15.

Form IV was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattem is shown in Figure 8 and the XRPD data are provided in the table below.

Table 4

Form IV was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 9. The DSC thermogram revealed an endothermic event at an onset temperature of 206.7 °C with a peak temperature of 208.2 °C.

Example 6

Preparation and Characterization of Form V

In one experiment, Form V was prepared according to the procedures below.

Approximately 3-4 mL of saturated solution of Form I in chloroform were evaporated under air without stirring at 22 ± 1 °C. Form V was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Examples 16 and 20.

Form V was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 10 and the XRPD data are provided in the table below.

Table 5

41.1 68 2.2

42.2 90 3

42.7 64 2.1

Form V was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 11. The DSC thermogram revealed an endothermic event at an onset temperature of 206.6 °C with a peak temperature of 208.1 °C.

Form V was characterized by TGA. The TGA was obtained using similar conditions as Form I. The TGA thermogram is shown in Figure 12.

Example 7

Preparation and Characterization of Form VI

In one experiment, Form VI was prepared according to the procedures below.

Approximately 3-4 mL of saturated solution of Form I in 1,4-dioxane were evaporated under air without stirring at 22 ± 1 °C. Form VI was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Example 16.

Form VI was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 13 and the XRPD data are provided in the table below.

Table 6

23.6 44 4.3

24.2 50 4.9

24.5 47 4.5

24.7 34 3.3

25.5 75 7.2

26.0 34 3.3

26.7 58 5.6

27.6 53 5.2

28.9 31 3

29.6 50 4.8

31.2 39 3.7

32.1 34 3.3

35.4 30 2.9

42.5 36 3.5

Example 8

Preparation and Characterization of Form VII

In one experiment, Form VII was prepared according to the procedures below.

Approximately 3-4 mL of saturated solution of Form I in THF were evaporated under air without stirring at 22 ± 1 °C. Form VII was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Examples 16, 17, and 18 .

Form VII was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 14 and the XRPD data are provided in the table below.

Table 7

24.1 209 24.1

24.7 856 99

25.7 71 8.2

26.5 148 17.2

27.3 121 13.9

28.2 63 7.3

29.7 71 8.2

31.6 69 8

32.2 36 4.1

33.3 36 4.1

37.4 430 49.7

40.0 57 6.5

42.2 38 4.4

43.7 74 8.5

Form VII was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 15. The DSC thermogram revealed an endothermic event at an onset temperature of 201.7 °C with a peak temperature of 204.9 °C.

Form VII was characterized by TGA. The TGA was obtained using similar conditions as Form I. The TGA thermogram is shown in Figure 16.

Example 9

Preparation and Characterization of Form VIII

In one experiment, Form VIII was prepared according to the procedures below. Approximately 3-4 mL of saturated solution of Form I in 1,4-dioxane was evaporated under air without stirring at 50 ± 1 °C. Form VIII was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Example 18.

Form VIII was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 17 and the XRPD data are provided in the table below.

Table 8

16.4 573 55.4

16.9 104 10

18.1 902 87.3

19.6 664 64.2

20.2 281 27.2

20.7 157 15.2

21.7 135 13.1

23.1 191 18.5

24.0 1034 100

24.4 421 40.7

25.4 490 47.4

26.6 476 46.1

27.3 151 14.6

27.9 270 26.1

29.8 40 3.8

30.5 48 4.6

31.2 199 19.3

32.2 86 8.3

32.8 53 5.2

33.4 55 5.3

34.0 135 13

38.6 41 4

39.0 69 6.6

39.6 41 3.9

40.2 44 4.3

41.7 108 10.5

Form VIII was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 18. The DSC thermogram revealed an endothermic event at an onset temperature of 205.6 °C with a peak temperature of 207.3 °C.

Form VIII was characterized by TGA. The TGA was obtained using similar conditions as Form I. The TGA thermogram is shown in Figure 19.

Example 10

Preparation and Characterization of Form IX

In one experiment, Form IX was prepared according to the procedures below. To 1 mL of saturated solution of Form I prepared in chloroform was added 5.0 mL hexane to give slurry, which was filtered. Form IX was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Example 17. Form IX was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 20 and the XRPD data are provided in the table below.

Table 9

Form IX was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 21. The DSC thermogram revealed an endothermic event at an onset temperature of 205.5 °C with a peak temperature of 207.2 °C.

Form IX was characterized by TGA. The TGA was obtained using similar conditions as Form I. The TGA thermogram is shown in Figure 22. Example 11

Preparation and Characterization of Form IXa

In one experiment, Form IXa was prepared according to the procedures below. To 1 mL of saturated solution of Form I prepared in chloroform was added 7.5 mL MTBE to give slurry, which was filtered. Form IXa was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Example 17.

Form IXa was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 23 and the XRPD data are provided in the table below.

Table 10

Form IXa was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 24. The DSC thermogram revealed an endothermic event at an onset temperature of 206.7 °C with a peak temperature of 208.0 °C.

Form IXa was characterized by TGA. The TGA was obtained using similar conditions as Form I. The TGA thermogram is shown in Figure 25.

Example 12

Preparation and Characterization of Form X

In one experiment, Form X was prepared according to the procedures below. To 1 mL of saturated solution of Form I prepared in 1 ,4-dioxane was added 5.0 mL heptane to give slurry, which was filtered. Form X was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Examples 17 and 18.

Form X was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 26 and the XRPD data are provided in the table below.

Table 11

2-Theta Height H%

4.4 5308 100

6.6 293 5.5

8.2 779 14.7

8.7 164 3.1

9.5 51 1

10.1 56 1.1

12.5 77 1.4

12.9 409 7.7

16.3 158 3

16.5 141 2.6

17.2 58 1.1

18.3 127 2.4

19.8 74 1.4

21.4 494 9.3

22.4 502 9.5

23.3 324 6.1

25.8 767 14.5

29.8 61 1.2

30.8 206 3.9

32.5 54 1

33.0 56 1.1

35.1 81 1.5

38.1 49 0.9 39.0 88 1.7

39.5 60 1.1

42.4 139 2.6

43.6 94 1.8

43.8 76 1.4

Example 13

Preparation and Characterization of Form XI

In one experiment, Form XI was prepared according to the procedures below. To 5.0 mL of heptane was added 1.5 mL of saturated solution of Form I prepared in

dichloromethane to give slurry, which was filtered. Form XI was also prepared in accordance with the procedures set forth in the examples provided herein, e.g., Example 18.

Form XI was characterized by XRPD. The XRPD was obtained using similar conditions as those for Form I. The XRPD pattern is shown in Figure 27 and the XRPD data are provided in the table below.

Table 12

30.2 44 0.8

31.4 76 1.3

32.5 572 10.2

34.0 112 2

35.2 58 1

36.4 61 1.1

38.1 37 0.7

41.4 52 0.9

42.4 311 5.5

Form XI was characterized by DSC. The DSC was obtained using similar conditions as those for Form I. The DSC thermogram is shown in Figure 28. The DSC thermogram revealed an endothermic event at an onset temperature of 206.5 °C with a peak temperature of 207.6 °C.

Form XI was characterized by TGA. The TGA was obtained using similar conditions as Form I. The TGA thermogram is shown in Figure 29.

Example 14

Solubility Measurement

The solubility of the Compound 1 Form I was measured according to Procedure 1 for solubility in 23 ± 1 °C (Table 13) and Procedure 2 for 50 ± 1 °C (Table 14) and the results are summarized in Table 15.

Compound 1 Form I is completely soluble (>50 mg/mL) in DMSO, DMF,

DCM/MeOH (1 :1), and THF/water (4: 1) at 23 °C and 50 °C, and in acetone/water (4:1) and chloroform at 50 °C. It is insoluble (<1 mg/mL) in heptane, hexane, toluene, MTBE, isobutyl acetate, IP Ac, water, and acetone water (1:4) at 23 °C and 50 °C, and in MIBK, EtOAc, IP A, and acetone water (1:2) at 23 °C. It is slightly soluble (1-50 mg/mL) in MeCN, 1,4-dioxane, MeOH, 2-methoxyethanol, THF, acetone, w-BuOH, EtOH, ethyl formate, 1-propanol, MEK, acetone water (2: 1), and acetone water (1 : 1) at 23 °C and 50 °C, and in chloroform and dichloromethane at 23 °C, and in MIBK, EtOAc, IP A, and acetone water (1:2) at 50 °C.

Table 13. Procedure 1 for solubility measurement of Compound 1 Form I in various solvents at 23 ± 1 °C

4 Agitated the mixture at 23 ± 1 °C over the weekend, which is controlled by IKA ® ETS-D5 temperature controller and IKA ® RCT basic safety control

5 Filtered the supernatant using syringe filter (PTFE, 0.22 μιτι, 13 mm, Agela Technologies Inc.)

6 Pipetted the saturated solution into HPLC vials.

7 Diluted the saturated solution in HPLC vials with acetonitrile.

8 HPLC analysis

9 Calculated solubility as indicated in Table 15

Table 14. Procedure 2 for solubility measurement of Compound 1 Form I in various solvents at 50 ± 1 °C

Table 15. Solubility of Compound 1 Form I in various solvents

Note: 1. Solubility of Form II in chloroform; 2. Solubility of Form III in 1,4-Dioxane; 3. Solubility of Form IV in Toluene.

Example 15

Phase Equilibration at 23± 1 °C and 50± 1 °C

Phase equilibration studies were designed to provide information on a predominant crystal form for phase identification. Based on its solubility in various solvent systems in Example 14, Compound 1 was equilibrated in a representative group of solvents at 23± 1 °C (Table 16) and 50 ± 1 °C (Table 17). To the solvents listed in Table 16 (23 ± 1 °C) and Table 17 (50 ± 1 °C) was added Compound 1 Form I until a cloudy solution was obtained, then about 20-40 mg of

Compound 1 Form I was added to the cloudy solution. The mixture was stirred at 23 ± 1 °C over weekend and 50 ± 1 °C for 2 days. The solid was filtered and analyzed by XRPD to give the results in Table 16 and Table 17.

Three new polymorphic forms were found by XRPD in the phase equilibration at 23 ± 1 °C including Form II (chloroform), Form III (1,4-dioxane), and Form IV (toluene). Form III (1 ,4-dioxane) was also identified in the phase equilibration at 50 ± 1 °C. Table 16: Crystal form for phase equilibration at 23 ± 1 °C

Table 17: Crystal form for phase equilibration at 50 ± 1 °C

Example 16

Evaporation at 23 ± 1 °C and 50± 1 °C

Evaporation studies were carried out to identify the predominant crystal form during uncontrolled precipitation. Experiments that did not result in any particulate solids (i.e. clear thin films and oils) were not studied further. XRPD was used to study the solid-state morphology of the crystalline forms of the evaporation samples at 23 ± 1 °C and 50 ± 1 °C. The results are presented in Table 18 (23 ± 1 °C) and Table 19 (50 ± 1 °C).

Evaporation at 23 ± 1 °C (Table 18) resulted in new polymorphic Form V

(chloroform), Form VI (1,4-dioxane), and Form VII (THF). Evaporation at 50 ± 1 °C (Table 19) resulted in new polymorphic Form VIII (1,4-dioxane). Table 18: Crystal form identification from evaporation at 23 ± 1 °C

rystal form identification from evaporation at 50 ± l °C

Solid State

No. Solvent

Form

1 Acetonitrile I

2 Chloroform I

3 DCM I

4 DMF I

5 1,4-Dioxane VIII

6 Methanol I

2-

7 I

Methoxyethanol

8 MIBK N/A

9 THF I

10 Acetone I

11 «-BuOH I

12 EtOH I

13 EtOAc I

Example 17

Anti-solvent addition

Saturated solutions of Compound 1 were prepared by adding the compound

(Compound 1 Form I) to the solvents respectively. An anti-solvent was added to induce precipitation. MTBE, toluene, water, heptane, and hexane were selected as the anti-solvents. Experiments that did not produce any particulate solids on anti-solvent addition were not studied further. The results are presented in Table 20. Antisolvent addition resulted in Form IX (chloroform-hexane), Form IXa (chloroform-MTBE), Form X (1,4-dioxane-heptane and 1 ,4-dioxane- hexane), and Form VII (THF -hexane).

Table 20. Antisolvent addition of Compound 1 Form I in various solvents

The saturated solution was prepared at 30 °C Example 18

Reverse Addition

Saturated solutions of Compound 1 Form I were prepared in solvents listed in Table 21, and added to a larger volume of a miscible anti-solvent. Experiments that did not produce any particulate solids upon addition to the anti-solvent were not studied further. Reverse addition resulted in Form XI (dichloro methane-heptane), Form X (1,4-dioxane- heptane and 1 ,4-dioxane-hexane), and Form VII (THF-hexane).

Table 21. Reverse addition of INCB062079 in various solvents

water (4: 1, 1.0)

THF/

26 water (4: 1, 1.0) Hexane (5.0) I

THF/

27 water (4: 1, 1.0) MTBE (5.0) I

THF/

28 water (4: 1, 1.0) Toluene (5.0) I

THF/

29 water (4: 1, 1.0) Water (5.0) I

Example 19

Quench Cool of saturated solution

Saturated solutions of Compound Form I prepared at about 35 °C were quenched cooled to induce precipitation of higher energy forms. Representative solvents were chosen based on solubility data measured at 23 °C and 50 °C. The studied solvents and the crystal form of the sample in each of the solvent were shown in Table 22. Quench cooling resulted in Form II (chloroform) and Form III (1,4-dioxane).

Table 22. Crystal form identification for Compound 1 Form I from quench cool

Example 20 Crystallization of saturated solution with heating and cooling cycles

Saturated solutions of Compound 1 Form I were prepared at 50 °C, and cooled in a bath slowly by using a programmed circulating bath. To the clear solution was added about 10 mg of Compound 1 Form I to give slurry. The formed slurry was then heated to 50 °C over 2 hours and then cooled down to 5 °C over 2 hours. This process was repeated for 3 days and the solid was filtered for further analysis. The results are presented in Table 23. Heating and cooling cycles resulted in Form V (chloroform) and Form III (1,4-dioxane).

Table 23: Crystallization of saturated solution of Compound 1 with heating and cooling recycles

Example 21

Studies of Stability Relationship of Compound 1 Forms

To evaluate the transformation of Compound 1 solid forms, competitive slurry experiments in production crystallization solvents at 23-25 °C were performed with a mixture of twelve (12) polymorphs (Form I through Form XI) according to the procedures in Table 25 (acetone/water 2:1) and Table 26 (acetone/water 1 :1). The mixture of forms (Form I through Form XI) was converted to Form I after stirring for 3.5 hours as shown by XRPD (data not shown). These results indicate that the Form I is the most stable polymorphic form in acetone/ water (2: 1) and acetone/water (1 : 1). Table 24. Mixed samples in different solvents (competitive slurries) 23-25 °C

Table 25. Procedure for competitive experiment in Acetone/water (2:1) at 23-25 °C

Example A

FGFR Enzymatic Assay

The inhibitor potency of the exemplified compounds is measured in an enzyme assay that measures peptide phosphorylation using FRET measurements to detect product formation. Inhibitors are serially diluted in DMSO and a volume of 0.5 is transferred to the wells of a 384-well plate. For FGFR3, a 10 volume of FGFR3 enzyme (Millipore) diluted in assay buffer (50 mM HEPES, 10 mM MgCh, 1 mM EGTA, 0.01% Tween-20, 5 mM DTT, pH 7.5) is added to the plate and pre-incubated for a time between 5-10 minutes and up to 4 hours. Appropriate controls (enzyme blank and enzyme with no inhibitor) are included on the plate. The assay is initiated by the addition of a 10 solution containing biotinylated EQEDEPEGDYFEWLE peptide substrate (SEQ ID NO: 1) and ATP (final concentrations of 500 nM and 140 μΜ respectively) in assay buffer to the wells. The plate was incubated at 25 °C for 1 hr. The reactions are ended with the addition of 10 μίΛ βΙΙ of quench solution (50 mM Tris, 150 mM NaCl, 0.5 mg/mL BSA, pH 7.8; 30 mM EDTA with Perkin Elmer Lance Reagents at 3.75 nM Eu-antibody PY20 and 180 nM APC-Streptavidin). The plate is allowed to equilibrate for ~1 hr before scanning the wells on a PheraStar plate reader (BMG Labtech).

FGFR1, FGFR2, and FGFR4 are measured under equivalent conditions with the following changes in enzyme and ATP concentrations: FGFR1 , 0.02 nM and 210 uM respectively, FGFR2, 0.01 nM and 100 uM, respectively, and FGFR4, 0.04 nM and 600 uM respectively. The enzymes can be purchased from Millipore or Invitrogen.

GraphPad prism3 is used to analyze the data. The IC50 values are derived by fitting the data to the equation for a sigmoidal dose-response with a variable slope. Y=Bottom + (Top-Bottom)/(l+10 A ((LogIC5o-X)*HillSlope)) where X is the logarithm of concentration and Y is the response. Compounds having an IC50 of 1 μΜ or less are considered active.

Example B

FGFR4 Cellular and In Vivo Assays

The FGFR4 inhibitory activity of the example compounds in cells, tissues, and/or animals can be demonstrated according to one or more assays or models described in the art such as, for example, in French et al. "Targeting FGFR4 Inihibits Hepatocellular Carcinoma in Preclinical Mouse Models," PLoS ONE, May 2012, Vol. 7, Issue 5, e36713, which is incorporated herein by reference in its entirety.

Various modifications of the invention, in addition to those described herein, will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including all patent, patent applications, and publications, cited in the present application is incorporated herein by reference in its entirety.