Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
CRYSTALLINE FORMS OF A QUINAZOLE COMPOUND AND ITS HYDROCHLORIDE SALTS
Document Type and Number:
WIPO Patent Application WO/2020/053816
Kind Code:
A1
Abstract:
This patent documentrelates to the crystalline forms of a quinazoline compound and the hydrochloride salts thereof. More particularly, this patent document relates to a preparation method of the crystalline forms of 1-(4-(4-(3,4-dichloro-2-fluorophenylamino) -7-methoxyquinazolin-6-yloxy)piperidin-1-yl)prop-2-en-1-one and its hydrochloride salts.

Inventors:
BAEK JONG OUK (KR)
KIM CHEOL HEE (KR)
HA TAE HEE (KR)
SUH KWEEHYUN (KR)
Application Number:
PCT/IB2019/057720
Publication Date:
March 19, 2020
Filing Date:
September 12, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HANMI PHARMACEUTICAL CO LTD (KR)
International Classes:
C07D239/94; A61K31/517
Domestic Patent References:
WO2015154725A12015-10-15
WO2018094225A12018-05-24
WO2018075823A12018-04-26
Foreign References:
KR20110135685A2011-12-19
KR101013319B12011-02-09
Other References:
See also references of EP 3849964A4
Download PDF:
Claims:
CLAIMS

What is claimed is:

1. Crystalline form of a compound of Chemical formula (1 ):

[Chemical formula (1 )]

wherein the chemical purity of the crystalline form is greater than about 80%, and wherein the crystalline form is selected from the group consisting of

(a) a dihydrate (2H20) crystalline form of the compound of Chemical Formula (1 ) and the crystalline form has an X-ray powder diffraction (XRPD) pattern comprising peaks at diffraction angle 2Q values of 9.4° ± 0.2°, 13.0° ± 0.2° and 18.5° ± 0.2° when irradiated with a Cu-Ka light source;

(b) an anhydrous Form I of the compound of Chemical Formula (1 ) having an XRPD pattern comprising peaks at diffraction angle 2 values Q of 6.0° ± 0.2°, 18.3° ± 0.2° and 22.7° ± 0.2° when irradiated with a Cu-Ka light source;

(c) an anhydrous Form II of the compound of Chemical Formula (1 ) having an XRPD pattern comprising peaks at diffraction angle 2Q values of 4.9° ± 0.2°, 5.9° ± 0.2° and 1 1 .8° ± 0.2° when irradiated with a Cu-Ka light source;

(d) a monohydrochloride monohydrate (1 HCI- 1 H20) crystalline form of the compound of Chemical Formula (1 ) having an XRPD pattern comprising peaks at diffraction angle 2Q values of 8.9° ± 0.2°, 1 3.4° ± 0.2°, 21 .1 ° ± 0.2° and 23.5° ± 0.2° when irradiated with a Cu-Ka light source; and

(e) an anhydrous monohydrochloride crystalline form of the compound of Chemical Formula (1 ) having an XRPD pattern comprising peaks at diffraction angle 2 values Q of 9.5° ± 0.2°, 23.0° ± 0.2°, 23.2° ± 0.2° and 23.5° ± 0.2° when irradiated with a Cu-Ka light source.

2. The crystalline form of the compound of Chemical formula (1 ) of claim 1 , wherein the chemical purity of the crystalline form is greater than 95%.

3. The crystalline form of claim 1 , wherein the crystalline form is as described in

4. The crystalline form of claim 1 , wherein the crystalline form is as described in

(b).

5. The crystalline form of claim 1 , wherein the crystalline form is as described in

(c)~

8. The crystalline form of claim 1 , wherein the crystalline form is as described in

(d)..

7. The crystalline form of claim 1 , wherein the crystalline form is as described in

(e).

8. The crystalline form of claim 1 , wherein the crystalline form is as described in

(a), and wherein the crystalline form has a 13C solid state nuclear magnetic resonance (ssNMR) spectrum comprising peaks at the following chemical shifts: 147.7 ± 0.5, 156.2 ± 0.5 and 165.4 ± 0.5 ppm.

9. The crystalline form of claim 1 , wherein the crystalline form is as described in

(b), and wherein the crystalline form has a 13C ssNMR spectrum comprising peaks at the following chemical shifts: 54.3 ± 0.5, 127.3 ± 0.5, 146.9 ± 0.5 and 156.7 ± 0.5 ppm.

10. The crystalline form of claim 1 , wherein the crystalline form is as described in

(c), and wherein the crystalline form has a 13C ssNMR spectrum comprising peaks at the following chemical shifts: 129.2 ± 0.5, 153.1 ± 0.5, 156.7 ± 0.5 and 165.2 ± 0.5 ppm.

1 1 . The crystalline form of claim 1 , wherein the crystalline form is as described in

(d), and wherein the crystalline form has a 13C ssNMR spectrum comprising peaks at the following chemical shifts: 145.8 ± 0.5, 157.8 ± 0.5 and 164.5 ± 0.5 ppm.

12. The crystalline form of claim 1 , wherein the crystalline form is as described in

(e), and wherein the crystalline form has a 13C ssNMR spectrum comprising peaks at the following chemical shifts: 146.9 ± 0.5, 158.7 ± 0.5 and 163.0 ± 0.5 ppm.

13. A pharmaceutical composition comprising the crystalline form of any one of claims 1 to 12, and at least one pharmaceutically acceptable carrier or diluent.

14. The pharmaceutical composition of claim 13, wherein the pharmaceutical composition is used for treating cancer induced by a tyrosine kinase or a mutant thereof.

15. The pharmaceutical composition of claim 14, wherein the cancer is a solid cancer.

16. The pharmaceutical composition of claim 13, wherein the chemical purity of the crystalline form is greater than about 95%.

17. The pharmaceutical composition of claim 13, further comprising a non-metallic salt lubricant selected from the group consisting of glyceryl behenate, glyceryl palmitostearate, glyceryl monostearate, glyceryl trimyristate, glyceryl tristearate, sucrose fatty acid ester, palmitic acid, palmitoyl alcohol, stearic acid, stearyl alcohol, fumaric acid, polyethyleneglycol 4000, polyethyleneglycol 6000, polytetrafluoroethylene, starch, talc, hydrogenated castor oil, mineral oil, hydrogenated vegetable oil, silicon dioxide, and any combination thereof.

18. The pharmaceutical composition of claim 13, further comprising a metallic salt lubricant.

19. A method to prepare the dihydrate (2H20) crystalline form of claim 3, comprising:

(a) adding the compound of Formula 1 to a mixture of acetone and water, wherein the acetone and the water are in such a ratio that the compound cannot completely dissolve in the mixture at room temperature;

(b) heating the mixture to a temperature wherein the compound completely dissolves; and

(c) cooling down the mixture and removing the acetone and water to obtain the dihydrate (2H20) of the compound.

20. A method to prepare the anhydrous Form I of claim 4, comprising

(a) mixing the dihydrate (2FI20) of the compound of claim 3 with acetone; and

(b) isolating the anhydrous Form I of the compound.

21 . A method to prepare the anhydrous Form II of claim 5, comprising

(a) mixing the anhydrous Form I of the compound of claim 4 with acetonitrile;

(b) heating to above about 80 °C; and

(c) cooling down and isolating the anhydrous Form II of the compound.

22. A method to prepare the monohydrochloride monohydrate (1 HCI- 1 Fl20) of claim 6, comprising

(a) mixing the compound of Formula 1 with ethanol, water and aqueous hydrochloric acid; and

(b) isolating the monohydrochloride monohydrate (1 HCI- 1 H20) of the compound.

23. A method to prepare the anhydrous monohydrochloride crystalline form of claim 7, comprising

(a) mixing an anhydrous compound of formula 1 with a non-protic polar solvent and a hydrochloric acid having a concentration of about 30% or more; and

(b) isolating the anhydrous monohydrochloride of the compound.

24. The method of claim 23, wherein the non-protic polar solvent is DMSO or DMF.

Description:
CRYSTALLINE FORMS OF A QUINAZOLINE COMPOUND AND ITS

HYDROCHLORIDE SALTS

CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit of U.S. Provisional Application No.

62/731 ,500, filed on September 14, 2018, the entire disclosures of which is incorporated herein by reference.

TECHNICAL FIELD

This patent document relates to crystalline forms of a quinazoline compound and its hydrochloride salt forms. More particularly, the crystalline forms are from

1 -(4~(4-(3,4-dichioro-2-fluQropheny!amino)

-7-methoxyquinazolin-6-y!oxy)piperidin-1 -yl)prop-2-en-1 -one, and a pharmaceutical composition containing the same.

BACKGROUND ART

The compound of the following chemical formula (1 ), having general formula of

1 -(4-(4-(3,4-dichloro-2-f!uorophenylamino)-7-methoxyquinazoli n-6-yloxy)piperidin-1 - yl)prop-2-en-1 -one is disclosed in Korean Patent No. 1 ,013,319 and U.S. Patent No. 8,003,658, and these patents disclose that the above compound has

antiproliferative activity such as anticancer activity, and can selectively and effectively treat the drug resistance induced by tyrosine kinase mutation:

[Chemical Formula (1 )]

However, the compound of chemical formula (1 ) prepared in the above cited patents is generally prepared in the form of amorphous solid or incomplete crystal which is less well suited for large-scale pharmaceutical processing, and there is no description with regard to the preparation of specific crystalline forms.

The compound of chemical formula (1 ) prepared by the above cited patents has a disadvantage in that the solubility in water is very low. in addition, since the compound of formula (I) prepared by the cited patents unavailable in the form of uniform crystals, meeting physicochemical stability standards required for pharmaceuticals can be troublesome.

Thus, there is a need to prepare the salts of the compound of chemical formula (1 ) in a crystalline form having improved solubility in water while still being capable of meeting sufficiently the strict requirements and specifications for pharmaceutical formulations.

The above information disclosed in this Background section is only for the enhancement of understanding of the background of the invention and therefore it by no means is intended as an admission that information contained herein constitutes prior art already known to a person of ordinary skill in the art.

SUMMARY OF THE DISCLOSURE

it is an object of this patent document to provide crystalline forms of the above quinazoiine compound of chemical formula (1 ) and its crystalline

hydrochloride salt forms as well as a pharmaceutical composition containing the same.

Formula (1 )

Specifically, the preferred crystalline form of the quinazoiine compound of chemical formula (1 ) is 1 } the crystalline hydrate form of the quinazoline compound of chemical formula (1 ), and

2) the crystalline anhydrous form of the quinazoline compound of chemical formula (1 )

in addition, the crystalline hydrochloride salt form of the preferred quinazoline compound of chemical formula (1 ) is

1 ) the crystalline hydrochloride salt hydrate form of the quinazoline compound of chemical formula (1 ),

2) the crystalline anhydrous hydrochloride salt form of the quinazoline compound of chemical formula (1 ).

More preferred examples of the crystalline form are as follows:

the crystalline dihydrate (2H2O) form of the compound of chemical formula (1 ) having the X-ray powder diffraction (XRPD) pattern including peaks at the diffraction angle (2Q±0.2°) of 9.4, 13.0, and 18.5° when irradiated with a Cu-Ka light source; the crystalline anhydrous Form I of the compound of chemical formula (1 ) having the XRPD pattern including peaks at the diffraction angle (2Q±0.2°) of 6.0, 18.3, and 22.7° when irradiated with a Cu-Ka light source;

the crystalline anhydrous Form II of the compound of chemical formula (1 ) having the X-ray powder diffraction pattern including peaks at the diffraction angle (2Q±0.2°) of 4.9, 5.9 and 1 1.8° when irradiated with a Cu-Ka light source;

the crystalline monohydrochloride salt monohydrate (1 HCM H2O) form of the compound of chemical formula (1 ) having the X-ray powder diffraction pattern including peaks at the diffraction angle (2Q±0.2°) of 8.9, 13.4, 21.1 and 23.5° when irradiated with a Cu-Ka light source; and

the crystalline anhydrous monohydrochloride salt (1 HCI) form of the compound of chemical formula (1 ) having the XRPD pattern including peaks at the diffraction angle {2q±0.2 o ) of 9.5, 23.0, 23.2 and 23.5° when irradiated with a Cu-Ka light source; and also, the crystalline di hydrate (2H 2 Q) form of the compound of chemical formula (1 ) having 13 C CP / MAS TOSS (cross polarization/magic angle spinning total suppression of sidebands) solid state nuclear magnetic resonance (ssNMR) spectrum including the chemical shifts {ppm ± 0.5 ppm) of 165.4, 156.2 and 147.7 ppm in the 13 G CP/M AS TOSS ssNMR spectrum;

the crystalline anhydrous Form I of the compound of chemical formula (1 ) having 13 C CP / MAS TOSS solid state nuclear magnetic resonance spectrum including the chemical shifts (ppm±0.5ppm) of 156.7, 146.9, 127.3 and 54.3 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance spectrum;

the crystalline anhydrous Form II of the compound of chemical formula (1 ) having 13 C CP / MAS TOSS ssNMR spectrum including the chemical shifts

(ppm±0.5ppm) of 165.2, 156.7, 153.1 and 129.2 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance spectrum;

the crystalline monohydrochloride salt monohydrate (1 HCI 1 H 2 Q) form of the compound of chemical formula {1 ) having 3 C CP / MAS TOSS solid state nuclear magnetic resonance spectrum including the chemical shifts (ppm±0.5ppm) of 164.5, 157.8 and 145.8 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance spectrum; and

the crystalline anhydrous monohydrochloride salt (1 HCI) form of the compound of chemical formula (1 ) having 13 G CP / MAS TOSS solid state nuclear magnetic resonance spectrum including the chemical shifts {ppm ± 0.5 ppm) of 163.0, 158.7 and 146.9 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance spectrum.

The crystalline form of the compound of chemical formula (1 ) or its hydrochloride salt is "substantially pure” wherein the expression "substantially pure" means at least 95%, preferably 99%.

That is to say, the purity of 95% to 99% means that the particular crystalline form of the compound of chemical formula (1 ) or its hydrochloride salt is 95% to 99% or more and other crystalline forms (amorphous or crystalline forms of the compound of Formula (1 ) other than the particular crystalline form) are 5% to 1 % or less.

In addition, this patent document provides the amorphous

monohydrochloride salt form of the said quinazoline compound of chemical formula (1 )·

According to another object of the present invention, this patent document provides a pharmaceutical composition comprising the crystalline form of the compound of formula (1 ) or the crystalline form of the hydrochloride salt thereof, and at least one pharmaceutically acceptable carrier or diluent.

The pharmaceutical composition has an antiproliferative activity such as anticancer activity and may be used for selective and effective treatment of drug resistance induced by tyrosine kinase mutagenesis.

The crystalline form of the compound of chemical formula (1 ) according to this patent document and its crystalline hydrochloride salt form is superior in terms of various physical and chemical properties such as solubility in water,

hygroscopicity and chemical stability, and thus can be easily used in the production of a pharmaceutical composition containing it as an active ingredient.

Also provided is a method of treating a neoplasm in a subject comprising administering to a subject in need thereof the novel crystalline form of the compound of Formula (1 ), a pharmaceutical composition thereof, or its combination with one or more other agents.

A further aspect of the patent document provides methods of preparing the crystalline forms described herein.

BRIEF DESCRIPTION OF THE DRAWINGS

The above and other features of the crystalline forms will now be described in detail with reference to certain exemplary embodiments thereof illustrated the accompanying drawings which are given hereinbelow by way of illustration only, and thus are not limitative of the present invention, and wherein:

FIGS. 1 A, 1 B, 1 C, 1 D and 1 E show the X-ray powder diffraction (XRPD) spectrums of the compound of chemical formula (1 ) and its crystalline hydrochloride salt form according to the example: Fig. 1 A shows the XRPD for the crystalline form prepared in Example 1 ; Fig. 1 B shows XRPD for the crystalline form prepared in Example 2; Fig. 1 C shows XRPD for the crystalline form prepared in Example 3; Fig.

1 D shows XRPD for the crystalline form prepared in Example 4: and Fig. 1 E shows XRPD for the crystalline form prepared in Example 5.

FIGS. 1 F and 1 G show the XRPD spectrums of the compound of chemical formula (1 ) and its amorphous hydrochloride salt form according to the comparative example; Fig 1 F shows XRPD tor the amorphous form prepared in Example 6; and Fig. 1 G shows XRPD for the compound of chemical formula (1 ) prepared in

Reference Example.

FIGS. 2A, 2B, 2C, 2D and 2E show the solid state nuclear magnetic resonance

(ssNMR) spectrums of the compound of chemical formula (1 ) and its crystalline hydrochloride salt form according to the example; Fig. 2A shows the ssNMRfor the crystalline form prepared in Example 1 ; Fig. 2B shows ssNMR for the crystalline form prepared in Example 2; Fig. 2C shows ssNMR for the crystalline form prepared in Example 3: Fig. 2D shows ssNMR for the crystalline form prepared in Example 4; and Fig. 2E shows ssNMR for the crystalline form prepared in Example 5.

FIGS. 2F and 2G show the ssNMR spectrums of the compound of chemical formula (1 ) and its amorphous hydrochloride salt form according to the comparative example; Fig 2F shows DVS tor the amorphous form prepared in Example 6; and Fig. 2G shows ssNMR for the compound of chemical formula (1 ) prepared in Reference Example.

FIGS. 3A, 3B, 3C, 3D and 3E show the Differential Scanning Calorimetry (DSC) graphs of the compound of chemical formula (1 ) and its crystalline hydrochloride salt form according to the example; Fig. 3A shows the DSC for the crystalline form prepared in Example 1 ; Fig. 3B shows DSC for the crystalline form prepared in Example 2; Fig. 3C shows DSC for the crystalline form prepared in Example 3; Fig. 3D shows DSC for the crystalline form prepared in Example 4; and Fig. 3E shows DSC for the crystalline form prepared in Example 5.

FIGS. 4A, 4B, 4C, 4D and 4E show the dynamic vapor sorption (DVS) graphs of the compound of chemical formula (1 ) and its crystalline hydrochloride salt form according to the example; Fig. 4A shows the DVS for the crystalline form prepared in Example 1 ; Fig. 4B shows DVS for the crystalline form prepared in Example 2; Fig. 4G shows DVS for the crystalline form prepared in Example 3; Fig. 4D shows DVS for the crystalline form prepared in Example 4; Fig. 4E shows DVS for the crystalline form prepared in Example 5.

FIGS. 4F and 4G show the DVS graphs of the compound of chemical formula (1 ) and its amorphous hydrochloride salt form according to the comparative example; Fig 4F shows ssNMR for the amorphous form prepared in Example 8; and Fig. 4G shows DVS for the compound of chemical formula (1 ) prepared in Reference

Example.

Detailed Description

The invention has been described in detail with reference to preferred embodiments thereof. However, it will be appreciated by those skilled in the art that changes may be made in these embodiments without departing from the principles and spirit of the invention, the scope of which is defined in the appended claims and their equivalents.

Definition

Terms not specifically defined in this specification have meanings recognized by those skilled in the art in light of the technology and context. However, unless otherwise specified, throughout the present specification the following terms have the meanings indicated below: The term“abouf as used herein means within 5%, preferably between 1% and 2% of a given value or range. For example, the expression "about 10%" refers to 9.5% to 10.5%, preferably 9.8% to 10.2%. As another example, the expression “about 100 °C” refers to 95 °C to 105 °C, preferably 98 ° C to 102 °C

As used herein the term“chemical purify’ refers to the weight % that is the specified chemical entity, including specified polymorph form. For example, when a crystalline dihydrate {2F½0) of the compound of Formula (1 ) is characterized as having greater than 95% chemical purity, that means that greater than 95% by weight of the substance is the crystalline dihydrate (2H 2 0) of the compound of Formula (1 ) and less than 5% by weight of any other compound including other anhydrous forms and/or polymorphs. Similarly, when a particular

monohydrochloride monohydrate crystalline form (1 HCI- 1 FI2O) of the compound of Formula (1 ) is characterized as having greater than 95% chemical purity, this particular crystalline form is more than 95% by weight among all forms (including for example crystalline or non-crystalline form, salt form or salt-free form, hydrate or anhydrous form) of the compound of Formula (1 ) in the same composition. The term “derived” in this context refers to forming a desirable crystalline form (e.g. an anhydrous form, a hydrate form, or a pharmaceutically acceptable salt) of the compound of Formula (1 ) without changing the chemical structure of the compound. The term“poziotinib” as used herein refers to any of the crystalline forms of the compound of formula (1 ).

The peak value of the diffraction angle (2Q) at the X-ray powder diffraction (XRPD) spectrum reported in this patent document has preferably the experimental error of ± 0.5%, more preferably ± 0.2% which is typically observable in the art.

Also, the chemical shift in the solid state nuclear magnetic resonance spectrum reported in this patent document win be preferably interpreted as within ± 0.5 ppm, more preferably as within ± 0.2%. The crystalline form of the auinazoline compound of chemical formula (1 ) and its crystalline hydrochloride salt form

This patent document provides the following compound of formula (1 ),

1 -(4-(4- (3,4-dichloro-2-f!uoropheny!amino)-7-methoxyquinazolin-6-y!o xy)piperidin-1 -yl)prop -2-en-1 -one and its crystalline hydrochloride salt form:

[Chemical formula (1 )]

The compound of chemical formula (1 ) above may be prepared according to the general procedure described in Korean Patent No. 1 ,013,319 and U.S. Patent No. 8,003,658, all of which are incorporated herein by reference in their entirety.

The compound of chemical formula (1 ) described in the above documents is a poorly soluble compound which is amorphous and has a solubility in water of less than 1 .0 pg/mL.

in general, it is known that the conversion of the free base to the form of salts aids in the solubilization of the water-insoluble drug substance. However, these salts must have various physicochemical properties such as reproducibility of the preparation of specific crystalline form, high crystallinity, stability of crystalline form, chemical stability, and non-hygroscopicity, etc. which are pharmacologically required.

in order to select the suitable salt form of the compound of chemical formula (1 ), various salts of the compound of chemical formula (1 ) were prepared using various acids and solvents according to various conditions and procedures, and their physicochemical properties were evaluated. Among the salts thus prepared, the crystalline forms of the various forms of the hydrochloride salt of the compound of chemical formula (1 ) are the most excellent in terms of various physicochemical properties such as reproducibility of the preparation of specific crystalline form, high crystallinity, stability of crystalline form, chemical stability, and non-hygroscopicity, etc. which are pharmacologically required.

its crystalline hydrochloride salt form

The salt of the compound of chemical formula (1 ) may be prepared in crystalline form, amorphous form or a mixture thereof, but it is preferable that the salt is in crystalline form. The crystalline hydrochloride salt form of the compound of chemical formula (1 ) is preferable in that it has excellent stability and

physicochemical properties that are easy to formulate.

According to the present invention, the compound of chemical formula (1 ) may be in the form of various crystalline forms, for example, its crystalline dihydrate (2HbG) form and crystalline anhydrous form.

Also, according to the present invention, the compound of chemical formula (1 ) may be in the form of various crystalline hydrochloride salts, for example, crystalline monohydrochloride monohydrate (1 HC1- 1 H. ¾ 0) form and their crystalline anhydrous monohydrochloride salt (1 HCl) form.

Among the crystalline hydrochloride salts, as a result of examining in Test Example 1 as described later, the crystalline anhydrous monohydrochloride salt form was the most excellent in solubility in water, and as a result of examining in Test Example 2, it may be advantageous in terms of non-hygroscopicity and stability, and thus may be desirable as a useful active ingredient in pharmaceutical compositions.

Hereinafter, each of the crystalline forms according to this patent document will be described in more detail. As an example, this patent document provides the crystalline dihydrate {2H2O) form of the compound of chemical formula (1 ).

The crystalline dihydrate (2H2O) form of the compound of chemical formula (1 ) has the XRPD spectrum including peaks at the diffraction angle (2Q ± 0 2°) of 9.4, 1 1 .4, 13.0, 16.1 , 1 8.5, 19.3, 24.9 and 26.3° when irradiated with a Gu-Ka light source. These peaks may be peaks with relative intensities of about 10% to 20% or more.

The above crystalline form may have chemical shifts (ppm±0.5ppm) of 147.7, 156.2, and 165.4 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance (cross polarization/magic angle spinning total suppression of sidebands solid state nuclear magnetic resonance, ssNMR) spectrum.

The above crystalline form may have a moisture content (theoretical moisture content of 6.83%) of about 7.5%, a condensation temperature of about 1 17 - 122 °C and a melting point of about 190 - 195 °C.

The above crystalline form may have an endothermic peak of the lowest point at about 1 1 1 °C when running from a starting point of about 79 °C, as measured by the DSC (10 °C / min).

The above crystalline form can be measured to have hygroscopic degree of about 2% to 5% in the relative humidity range of 0 - 90%, as measured by the DVS.

As another example, this patent document provides the crystalline anhydrous Form I of the compound of chemical formula (1 ).

The crystalline anhydrous Form I of the compound of chemical formula (1 ) has the XRPD spectrum including peaks at the diffraction angle (2Q ± 0.2°) of 6.0, 10.6, 10.9, 12.1 , 1 6.0, 17.5, 18.3, 19.2, 20.3, 22.7, 23.7 and 26.3° when irradiated with a Gu-Ka light source. These peaks may be peaks with relative intensities of about 10% to 20% or more. The above crystalline form may have chemical shifts (ppm±0.5ppm) of 54.3, 127.3, 146.9 and 156.7 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance (ssNMR) spectrum.

The above crystalline form may have a moisture content of about 0.1 % and a melting point of about 190 - 195 °C.

The above crystalline form may have an endothermic peak of the lowest point at about 191 °C when running from a starting point of about 186 °C, as measured by the DSC (10 °C / min).

The above crystalline form can be measured to have a hygroscopic degree of about 0.5% in a relative humidity range of 10 - 50% as measured by the DVS and a hygroscopic degree of about 3% in a relative humidity range of 50 - 90%.

As another example, this patent document provides the crystalline anhydrous Form II of the compound of chemical formula (1 ).

The crystalline anhydrous Form II of the compound of chemical formula (1 ) may have the XRPD spectrum including peaks at the diffraction angle (2Q ± 0.2°) of 4.9, 5.9, 1 1 .8, 18.8 and 19.9° when irradiated with a Cu-Ka light source. These peaks may be peaks with relative intensities of about 10% to 20% or more.

The above crystalline form may have chemical shifts (ppm ± Q.5ppm) of 129.2, 153.1 , 156.7 and 165.2 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance (ssNMR) spectrum.

The above crystalline form may have a moisture content of about 0.3% and a melting point of about 183 185 °C.

The above crystalline form may have an endothermic peak of the lowest point at about 185 °C when running from a starting point of about 181 °C, as measured by the DSC (10 °C / min).

The above crystalline form can be measured to have very low hygroscopic degree in a relative humidity range of 0 - 90%, as measured by the DVS. As another example, this patent document provides the crystalline monohydrochioride monohydrate (1 HCI 1 H2O) form of the compound of chemical formula (1 ).

The crystalline monohydrochioride monohydrate (1 HCI 1 H2O) form of the compound of chemical formula (1 ) may have the XRPD spectrum including peaks at the diffraction angle (2Q ± 0.2°) of 8.9, 13.4, 14.1 , 16.0, 19.8, 21 .1 , 21.7, 23.5, 25.7 and 32.7° when irradiated with a Cu-Kcs light source. These peaks may be peaks with relative intensities of about 10% to 20% or more.

The above crystalline form may have chemical shifts (ppm ± O.Sppm) of 145.8, 157.8 and 164.5 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance issNMR) spectrum.

The above crystalline form may have an endothermic peak of the lowest point at about 151 °C and an endothermic peak at about 178 °C when running from a starting point of about 127 °C, as measured by the DSC (10 °C/min).

The above crystalline form may have a moisture content of about 3.2%

(theoretical moisture content 3.30%) and a melting point of about 187 - 193 °C.

The above crystalline form can be measured to have very low hygroscopic degree in a relative humidity range of 10 - 90% as measured by the DVS.

As another example, this patent document provides the crystalline anhydrous monohydrochioride (1 HCI) form of fhe compound of chemical formula (1 ).

The crystalline anhydrous monohydrochioride salt (1 HCI) form of the compound of chemical formula (1 ) may have the XRPD spectrum including peaks at the diffraction angle (2Q ± 0.2°) of 9.5, 12.3, 13.0, 13.5, 14.2, 21 .4, 23.0, 23.2, 23.5, 27.2 and 27.5° when irradiated with a Cu-Ka light source. These peaks may be peaks with relative intensities of about 10% to 20% or more.

The above crystalline form may have chemical shifts (ppm ± O.Sppm) of 146.9, 158.7 and 163.0 ppm in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance (ssNMR) spectrum. The above crystalline form may have an endothermic peak of the lowest point at about 230 °C when running from a starting point of about 201 °C, as measured by the DSC (10 °C / min).

The above crystalline form may have a moisture content of about 0.1 % and a melting point of about 238 - 243 °C.

The above crystalline form can be measured to have very low hygroscopic degree in a relative humidity range of 10 - 90%, as measured by the DVS.

A general process for the preparation of crystalline forms (hydrate or anhydrous) of the compound of chemical formula (1 ) according to this patent document is provided. The process involves:

(a) providing a solution of the compound of chemical formula (1 ) in a solvent system (protic, aprotic or mixed);

(b) cooling the solution to effect formation of a crystal form (hydrate or anhydrous) of the compound of chemical formula (1 ); and

(c) isolating the crystalline form (hydrate or anhydrous) of the compound of chemical formula (1 ).

A process for the preparation crystalline hydrochloride salt forms (hydrate or anhydrous) of the compound of chemical formula (1 ) is also provided. The process involves:

(a) providing a solution of the compound of chemical formula (1 ) in a solvent system (protic, aprotic or mixed);

(b) adding hydrochloric acid to the solution ;

(c) cooling the solution to effect formation of a crystalline hydrochloride salt forms (hydrate or anhydrous) of the compound of chemical formula (1 ); and

(d) isolating the crystalline hydrochloride salt form (hydrate or anhydrous) of the compound of chemical formula (1 ). Non-limiting exampies of the solvent systems are the following: acetone; acetonitrile; acetone/water; acetonitrile/water; ethanol; ethanol/water. DMSO;

DMSO/water, DMF; DM F /water.

This process is highiy-reproducibie and the resulting crystalline product has good fiiterability.

The crystalline forms (hydrate or anhydrous) of the compound of chemical formula (1 ) according to this patent document and its crystalline hydrochloride salt forms (hydrate or anhydrous) do not require particular storage conditions and can be stably maintained for a long period of time. Capable of meeting the

physicochemical properties required for pharmaceuticals including excellent solubility in water, they are readily usable in the manufacture of pharmaceutical compositions containing these as active ingredients.

The crystalline forms (hydrate or anhydrous) of the compound of chemical formula (1 ) according fo this patent document have a high chemical purity in some embodiments, the chemical purify is greater than about 75%, greater than about 80%, greater than about 85%, greater than about 90%, greater than about 95%, or greater than about 99%.

Pharmaceutical composition

As disclosed in Korean Patent No. 1 ,013,319 and U.S. Patent No 8,003,658, incorporated herein in their entirety, it was proven that the compound of chemical formula (1 ) has an antiproliferative activity such as anticancer activity and has an activity of selectively and effectively inhibiting the growth and drug resistance of cancer ceils induced by tyrosine kinase or its variants.

in this respect, the crystalline form of the compound of chemical formula (1 ) and its hydrochloride salt can be used to produce a pharmaceutical composition for the treatment or prevention of various solid cancers, such as cancers or tumors, particularly lung cancer, breast cancer, etc caused by tyrosine kinase or its variants. The dosage of the crystalline form of the compound of chemical formula (1 ) and hydrochloride salts thereof may vary depending on the subject to be treated, the severity of the disease or condition, the rate of administration and the judgment of the prescribing physician, but they can be usually administered to an individual as an active ingredient in an amount of 1 to 2,000 g per kg of body weight 70 kg of free base of the compound of chemical formula (1 ), preferably 5 to 1 ,000 mg based on the compound of chemical formula (1 ), usually on a schedule of one to four times a day, or on/off schedule, via an oral or parenteral route. In some cases, dosage less than the above-mentioned ranges may be more suitable, dosage more than the above- mentioned ranges may be also used without causing harmful side effects, and in the case of the higher dosage, it is administered in divided doses several times per day.

The pharmaceutical composition according to this patent document may be formulated according to the conventional method and may be prepared in various oral dosage forms such as tablets, pills, powders, capsules, syrups, emulsions or micro-emulsions, etc. or in parenteral dosage forms such as intramuscular, intravenous or subcutaneous administration.

When the pharmaceutical composition according to this patent document is prepared in the form of an oral formulation, examples of the carrier include cellulose, calcium silicate, corn starch, lactose, sucrose, dextrose, calcium phosphate, stearic acid, magnesium stearate, calcium stearate, gelatin, talc, surfactants, suspending agents, emulsifiers, diluents and the like. When the pharmaceutical composition according to this patent document is prepared in the form of an injection, examples of the carrier include water, saline solution, aqueous glucose solution, aqueous pseudosugar solution, alcohols, glycols, ethers (for example, polyethylene glycol 400), oils, fatty acids, fatty acid esters, glycerides, surfactants, suspending agents, emulsifiers and the like. In some embodiments, the pharmaceutical composition further includes a non-metallic salt lubricant selected from the group consisting of glyceryl behenate, glyceryl palmitostearate, glyceryl monostearate, glyceryl trimyristate, glyceryl tristearate, sucrose fatty acid ester, palmitic acid, palmitoyl alcohol, stearic acid, stearyl alcohol, fumaric acid, polyethyleneglycol 4000, polyethyleneglycol 6000, polytetrafluoroethylene, starch, talc, hydrogenated castor oil, mineral oil, hydrogenated vegetable oil, silicon dioxide, and any combination thereof.

in some embodiments, the pharmaceutical composition further includes a metallic salt lubricant. Non-limiting examples include magnesium stearate, magnesium silicate, stearic acid, and calcium stearate.

A related aspect of the invention includes a kit for treating cancer including the crystalline form described herein or a pharmaceutical composition thereof. The kit or the pharmaceutical composition can contain an additional cytotoxic agent or a moiecu!ariy targeted agent. The crystalline form described herein or a

pharmaceutical composition thereof and the additional cytotoxic agent can be administered sequentially or simultaneously, depending on the specific conditions of the subject.

A cytotoxic agent refers to an agent that has a cytotoxic effect on a ceil. A cytotoxic efieet refers to the depletion, elimination and/or the killing of a target ceils (i.e., tumor cells). The cytotoxic agent may be at least one selected from fhe group consisting of an antimetabolite, a mitotic inhibitor, alkylating agent, a platinum-based antineoplastic drug, an mTGR inhibitor, a VEGF inhibitor, an aromatase inhibitor and a GDK4/6 inhibitor. The kit or combination of agents may include at least two cytotoxic agents. For example, the combination may include at least 2, at least 3, or at least 4 selected from the group consisting of an antimetabolite, a mitotic inhibitor, alkylating agent, a platinum-based antineopiastic drug, an mTGR inhibitor, a VEGF inhibitor, an aromatase inhibitor, a CDK4/6 inhibitor and ail of them. The antimetabolite may be a drug that inhibits D A synthesis in cells by suppressing formation of purines or pyrimidines, which are bases of a nucleotide. The antimetabo!ite may be selected from the group consisting of Capecitabine, 5-Fluorouracil, Gemeitabine, Pemetrexed, Methotrexate, 6-Mercaptopurine, Cladribine, Cytarabine, Doxifiudine, Fioxuridine, Fiudarabine, Hydroxycarbamide, decarbazine, hydroxyurea, and asparaginase.

The mitotic inhibitor may be a microtubule-destabilizing agent, a

microtubule-stabilizing agent, or a combination thereof. The mitotic inhibitor may be taxane, vinca alkaloid, epothiione, or a combination thereof.

The mitotic inhibitor may be selected from BT-Q62, HMN-214, eribulin mesylate, vindesine, EC-1089, EC-1458, EC-531 , vintafolide, 2-methoxyestradiol, GTx-230, trastuzumab emtansine, crolibuiin, D1302A-maytansinoid conjugates IMGN-529, lorvotuzumab mertansine, 8AR-3419, SAR-566858, IMP-03138, topotecan/vincristine combinations, BPH-8, fosbretabulin tromethamine, estramustine phosphate sodium, vincristine, vinflunine, vinorelbine, RX-21 101 , cabazitaxel, 8TA-9584, vinblastine, epothiione A, patupilone, ixabepilone,

Epothiione D, paciitaxel, docetaxel, DJ-927, discodermolide, eieutherobin, and pharmaceutically acceptable salts thereof or combinations thereof.

As used herein, an "alkylating agent" is a substance that adds one or more alkyl groups (CnHm, where n and m are integers) to a nucleic acid in the present invention, an alkylating agent is selected from the group consisting of nitrogen mustards, nitrosoureas, alkyl sulfonates, triazines, ethylenimines, and combinations thereof. Non-limiting examples of nitrogen mustards include mechlorethamine, chlorambucil, cyclophosphamide, bendamustine, ifosfamide, meiphaian, meiphaian flufenamide, and pharmaceutically acceptable salts thereof. Non-limiting examples of nitrosoureas include streptozocin, carmustine, iomustine, and pharmaceutically acceptable salts thereof. Non-limiting examples of alkyl sulfonates include busulfan and pharmaceutically acceptable salts thereof. Non-limiting examples of triazines include dacarbazine, temozolomide, and pharmaceutically acceptable salts thereof. Non-limiting examples of ethylenimines include thiotepa, altretamine, and pharmaceutically acceptable salts thereof. Other alkylating agents include

ProLindac, Ac-225 BC-8, ALF-21 11 , trofosfamide, MDX-1203,

thioureidobutyronitrile, mitobronitol, mitolactol, nimustine, glufosfa ide,

HuMax-TAC and PBD ADC combinations, BP-C1 , treosuifan, nifurtimox, improsulfan tosilate, ranimustine, ND-01 , HH-1 , 22P1 G ceils and ifosfa ide combinations, estramustine phosphate, prednimustine, lurbinectedin, trabectedin, altreatamine, 8GN-CD33A, fotemustine, nedaplatin, heptapiatin, apaziquone, SG-20Q0, TLK-58747, laromustine, procarbazine, and pharmaceutically acceptable salts thereof.

The platinum-based antineoplastic drug may be for example, Cisplatin, Carboplatin, Dicyclopiatin, Eptaplatin, Lobaplatin, Miriplatin, Nedaplatin, Oxaliplatin, Picopiatin, or Satraplatin.

The term "mTOR inhibitors (mTQR inhibitor)" as used herein is used for purposes of a material to inhibit the mTOR signaling pathway of the conventional anticancer agents or immunosuppressive agents. The mTOR inhibitor may be rapamycin, temsirolimus, everolimus, ridaforolimus, MLN4924, XL388, GDG-0349, AZD2014, AZD8055, GSK105965, MLNQ128 Ridafor!imus and the like.

“VEGF inhibitor” as used herein is any substance that decreases signaling by the VEGF-VEGFR pathway. VEGF inhibitors can be, to name just a few examples, small molecules, peptides, polypeptides, proteins, including more specifically antibodies, including anti-VEGF antibodies, anti-VEGFR antibodies, intrabodies, maxibodies, minibodies, diabodies, Fc fusion proteins such as peptibodies, receptibodies, soluble VEGF receptor proteins and fragments, and a variety of others. Many VEGF inhibitors work by binding to VEGF or to

a VEGF receptor. Others work more indirectly by binding to factors that bind to VEGF or to a VEGF receptor or to other components of the VEGF signaling pathway. Still other VEGF inhibitors act by altering regulatory posttranslational modifications that modulate VEGF pathway signaling. VEGF inhibitors in

accordance with the invention also may act through more indirect mechanisms. Whatever the mechanism involved, as used herein, a VEGF inhibitor decreases the effective activity of the VEGF signaling pathway in a given circumstance over what if would be in the same circumstance in the absence of the inhibitor.

Non-limiting examples of VEGF inhibitors include: (a) 4TBPPAPG or a closely related compound described in US2003/0125339 or U.S. Pat. No. 8,995,182 which is herein incorporated by reference in its entirety, particularly in parts disclosing 4TBPPAPC and closely related VEGF inhibitors: (b) AMG 706 or a closely related substituted alkylamine derivative described in US2003/0125339 or US2003/0225106 or U.S. Pat. No. 6,995,162 or U.S. Pat. No. 6,878,714 each of which is herein incorporated by reference in its entirety, particularly in parts disclosing AMG 706 and these closely related VEGF inhibitors; (c) Avastin™ or a closely related non-natura!ly occurring humanized monoclonal antibody that binds to VEGF, is a VEGF inhibitor, and is at least 90% identical in sequence to Avastin™; (d) Nexavar® or a closely related substituted omega-carboxyaryl diphenyl urea or derivative thereof described in WO00/42012, WOOO/41698, US20Q5/Q038QSGA1 , US2003/0125359 A1 , US2002/0165394A1 , US2001 /003447A1 ,

US2001/0016659A1 , and US2002/013774A1 which are herein incorporated by reference in their entirety, particularly in parts disclosing these VEGF inhibitors; (e) PTK/ZK or a closely related aniiinophthalazine or derivative thereof that binds to and inhibits the activity of multiple receptor tyrosine kinases including binding to the protein kinase domain and inhibition of VEGFR1 and VEGFR2; (f) Sutent® or a closely related derivative of

(5-[5-fluoro-2-oxo-1 ,2-dihydroindol-(3Z)-ylidenemethyl]-2,4-dimethyl-1 H-pyrrole-3-c arboxylic acid [2-diethylaminoethyl]amide) that is a VEGF inhibitor: and (g) VEGFinhibitors as described in US2006/0241 1 15, including those of Formula IV therein.

Further examples of VEGF inhibitors are the following: (a) 4TBPPAPC, as described in US2003/0125339 or U.S. Pat. No. 6,995,162 which is herein incorporated by reference in its entirety, particularly in parts disclosing 4TBPPAPC; (b) AMG 706, as described in US2003/0125339 or U.S. Pat. No. 6,995,162 or U.S. Pat. No. 6,878,714 which is herein incorporated by reference in its entirety, particularly in parts disclosing AMG 706: (c) Avastin™; id) Nexavar®, as described in W 000/42012, WOOO/41698, US2005/0038080A1 , US2003/0125359A1 , US2002/0165394A1 , US2001/003447A1 , US2001/0016659A1 , and

US2002/013774A1 which are herein incorporated by reference in their entirety, particularly in parts disclosing Nexavar®; (e) PTK/ZK; (f) Sutent®, and

(g) VEGF inhibitors of Formula IV as described in US2006/0241 1 15.

In some embodiments, the VEGF inhibitor is pegaptanib. in one

embodiment, the VEGF inhibitor is bevacizumab. In one embodiment,

the VEGF inhibitor is ranibizumab. In one embodiment, the VEGF inhibitor is lapatinib. in one embodiment, the VEGF inhibitor is sorafenib. In one embodiment, the VEGFinhibitor is sunitinib. In one embodiment, the VEGF inhibitor is axitinib. in one embodiment, the VEGF inhibitor is pazopanib. In one embodiment,

the VEGFinhibitor is afiibercept.

By "aromatase inhibitor” it is meant non-steroidal and steroidal compounds that inhibit the enzyme aromatase thereby preventing the conversion of androgens to estrogens, preferably those which inhibit aromatase activity in vitro with an IC50 value of less than 1 Q~5 M as well as their pharmaceutically acceptable salts.

Exemplary aromatase inhibitors for use in the methods herein described include without limitation anastrozole, letrozole, exemestane, vorozole, formestane, fadrozole, aminogiutethimide, testolactone, 4-hydroxyandrostenedione,

l,4,6-androstatrien-3,17-dione and 4-androstene-3,6,17-trione. The terms "cyclin dependent kinase 4/6 inhibitor" and "CDK4/6 inhibitor" as used herein refer to a compound that selectively targets, decreases, or inhibits at least one activity of CDK4 and/or CDK6 Non-limiting examples of inhibitors of CDK4/6 include Abemaciclib (LY2835219), paibociclib (PD0332991 ), LEE-01 1 (ribociclib), LY2835219 (abemaciclib), G1T28-1 , SHR6390, or P276-00, o a derivative of any one of paibociclib, LEE-01 1 , G1T28-1 , SHR6390, or P278-G0. In certain embodiments, the CDK4/6 inhibitor may be derived from pyridopyrimidine, pyrrolopyrimidine or indolocarbazoie compounds.

As used herein, a "molecuiar!y targeted agent" is a substance that interferes with the function of a single molecule or group of molecules, preferably those that are involved In tumor growth and progression, when administered to a subject. Non-limiting examples of moieculariy targeted agent of this patent document include signal transduction inhibitors, modulators of gene expression and other cellular functions, immune system modulators, antibody-drug conjugates (ADCs), and combinations thereof.

The moieculariy targeted agent may be selected from epidermal growth factor receptor family inhibitors (EGFRi), mammalian target of rapamycin (m f or) inhibitors, immune checkpoint inhibitors, anaplastic lymphoma kinase (ALK) inhibitors, B-cell lymphoma-2 (BCL-2) inhibitors, B-Raf inhibitors, cyciin-dependent kinase inhibitors (CDKi), ERK inhibitors, histone deacetyiase inhibitors (HDACi), heat shock protein-90 inhibitors (HSP90i), Janus kinase inhibitors, mitogen activated protein kinase (MARK) inhibitors, MEK inhibitors, poly ADR ribose polymerase (PARR) inhibitors, phosphoinositide 3-kinase inhibitors (Pi3Ki), Ras inhibitors, and combinations thereof.

The moieculariy targeted agent may be selected from ado-trastuzumab emtansine, a!emtuzumab, cetuximab, ipilimumab, ofatumumab, panitumumab, pertuzumab, rituximab, tositumomab, 131 i-tositumomab, trastuzumab, brentuximab vedotin, denileukin diftitox, ibritumomab tiuxetan, axitinib, bortezomib, bosutinib, cabozantinib, crizotinib, carfiizomib, dasatinib, erlotinib, gefsiSnib, imatinib mesylate, lapatinib, niiotinib, pazopanib, ponatinib, regorafenib, ruxolitinib, sorafenib, sunitinib, tofacitinib, vandetanib, vemurafenib, aiitretinoin, bexarotene, evero!imus, , romidepsin, temsirolimus, tretinoin, vorinostat, and pharmaceutically acceptable salts thereof or combinations thereof. The mo!ecular!y targeted agent may include an antibody or an antibody moiety.

The EGFRi may be selected from erlotinib, gefitinib, lapatinib, canetinib, pelitinib, neratinib,

(R,E)-N-{7-chloro-1 -(1 -(4-(dimethylamino)but-2-enoyl)azepan-3-yl)-1 H-benzoEdjimid azol-2-yi)-2-methy!isonicotinamide, Trastuzumab, Margetuximab, panitumumab, mafuzumab, Necitumumab, pertuzumab, nimotuzumab, zalutumumab,

Necitumumab, cetuximab, icotinib, aiatinib, and pharmaceutically acceptable salt thereof. The molecu!arly targeted agent may be an anti-EGFR family antibody or a complex including the anti-EGFR family antibody. The anti-EGFR family antibody may be an anti-HERt antibody, an an†i-HER2 antibody, or an anti-HER4 antibody.

Method Of Treating Cancer

Another aspect of the invention provides a method of treating a neoplasm in a subject comprising administering to a subject in need thereof the novel crystalline form described herein, the pharmaceutical composition thereof, a combination with one or more agents, or a kit containing the novel crystalline form.

Non-limiting examples of the neoplasm that can be treated according to this patent document include lung cancer including non-small cell lung cancer, breast cancer, stomach cancer, colon cancer, pancreatic cancer, prostate cancer, myeloma, head and neck cancer, ovarian cancer, esophageal cancer, or metastatic cell carcinoma. In some embodimetns, the neoplasm associated with

overexpression or amplification of at least one gene of HER1 , HER2, and HER4 or a mutant thereof may be an abnormal growth of tissue, which if it forms a mass, is commonly referred to as a tumor having overexpression of at least one of HER1 , HER2, HER4 and mutant thereof or amplification of at ieast one gene coding of HER1 , HER2, HER4 or mutant thereof. The mutant may be HER1 having exon 19 deletion, T79QM substitution, L828R substitution, or combination thereof. As used herein, "overexpression" indicates that the protein is expressed at a higher level than normal ceils. The expression level can be measured using

immunohistochemistry, fluorescence in situ hybridization (FISH) or chromogenic in situ hybridization (GISH).

The term "wild type" as used herein is understood in the art refers to a polypeptide or polynucleotide sequence that occurs in a native population without genetic modification. As is also understood in the art, a "mutant" includes a polypeptide or polynucleotide sequence having at ieast one modification to an amino acid or nucleic acid compared to the corresponding amino acid or nucleic acid found in a wild type polypeptide or polynucleotide, respectively included in the term mutant is Single Nucleotide Polymorphism (SNP) where a single base pair distinction exists in the sequence of a nucleic acid strand compared to the most prevalently found (wild type) nucleic acid strand.

Neoplasm including cancers that are either wild type or mutant for HER1 , HER2, or HER4 or have amplification of HER1 , HER2, or HER4 genes or have over expression of HER1 , HER2, or HER4 protein are identified by known methods.

For example, wild type or mutant HER1 , HER2, and HER4 tumor ceils can be identified by DNA amplification and sequencing techniques, DNA and RNA detection techniques, including, but not limited to Northern and Southern blot, respectively, and/or various biochip and array technologies or in-situ hybridization. Wild type and mutant polypeptides can be detected by a variety ot techniques including, but not limited to immunodiagnostic techniques such as ELISA, Western blot or immunocytochemistry.

The cancer to be treated may be associated with EGFR and/or HER2 exon 20 mutations, such as exon 20 insertion mutations. For instance, the novel crystalline form of the compound of this patent document or its combination with one or more agents can be used for the treatment of NSCLC patients with EGFR exon 20 mutations.

In some aspects, the cancer to be treated with the novel crystalline form of the compound of this patent document or its combination with one or more agents is oral cancer, oropharyngeal cancer, nasopharyngeal cancer, respiratory cancer, urogenital cancer, gastrointestinal cancer, central or peripheral nervous system tissue cancer, an endocrine or neuroendocrine cancer or hematopoietic cancer, glioma, sarcoma, carcinoma, lymphoma, melanoma, fibroma, meningioma, brain cancer, oropharyngeal cancer, nasopharyngeal cancer, renal cancer, biliary cancer, pheochromocytoma, pancreatic islet cell cancer, Li-Fraumeni tumors, thyroid cancer, parathyroid cancer, pituitary tumors, adrenal gland tumors, osteogenic sarcoma tumors, multiple neuroendocrine type I and type II tumors, breast cancer, lung cancer, head and neck cancer, prostate cancer, esophageal cancer, tracheal cancer, liver cancer, bladder cancer, stomach cancer, pancreatic cancer, ovarian cancer, uterine cancer, cervical cancer, testicular cancer, colon cancer, rectal cancer or skin cancer. In particular aspects, the cancer is non-small cell lung cancer.

By the term "treating" and derivatives thereof as used herein, is meant therapeutic therapy. In reference to a particular condition, treating means: (1 ) to ameliorate or prevent the condition of one or more of the biological manifestations of the condition, (2) to interfere with (a) one or more points in the biological cascade that leads to or is responsible for the condition or (b) one or more of the biological manifestations of the condition, (3) to alleviate one or more of the symptoms, effects or side effects associated with the condition or treatment thereof, or (4) to slow the progression of the condition or one or more of the biological manifestations of the condition. Prophylactic therapy is also contemplated thereby. The skilled artisan will appreciate that "prevention" is not an absolute term in medicine, "prevention" is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a condition or biological manifestation thereof, or to delay the onset of such condition or biological manifestation thereof.

Prophylactic therapy is appropriate, for example, when a subject is considered at high risk for developing cancer, such as when a subject has a strong family history of cancer or when a subject has been exposed to a carcinogen.

The administration of a therapeutically effective amount of the novel crystalline form of compound of Formula (1 ) or ifs combinations with other agents are advantageous over many other conventional therapies including: i) a greater anticancer effect than the most active single agent, ii) synergistic or highly synergistic anticancer activity, iii) a dosing protocol that provides enhanced anticancer activity with reduced side effect profile, iv) a reduction in the toxic effect profile, v) an increase in the therapeutic window, vi) an increase in the bioavailabiiity of one or both of the component compounds, or vis) an increase in apoptosis over the individual component compounds.

in some embodiments, the kit or combination of the novel crystalline form of the compound of Formula (1 ) of this patent document may include poziotinib and an anti-EGFR family antibody. The anti-EGFR family antibody may be trastuzumab, cetuximab, margetuximab, matuzumab, panitumumab, necitumumab, or pertuzumab. An example of the combination may be poziotinib and trastuzumab; or poziotinib and cetuximab Poziotinib may be hydrochloride. The combination may further include a cytotoxic agent. The cytotoxic agent may be a mitotic inhibitor. The mitotic inhibitor may be taxane, vinca alkaloid, epothilone, or a combination thereof. The vinca alkaloid may be at least one drug selected from the group consisting of vinblastine, vincristine, vindesine and vinoreibine. An example of the combination may include poziotinib; and trastuzumab and vinoreibine. The vinoreibine may be in the form of an injection. The taxane may be paclitaxel or docetaxel. An example of the combination may include poziotinib; and cetuximab and pacliataxe!. The paclitaxel may be in the form of an injection. The novel crystalline form of the compound of Formula (1 ) of this patent document may be administered in an amount of 0.1 mg to 50 mg. Trastuzumab may be administered in an amount of 0.5 mg to 10 g per kg of a body weight.

Cetuximab may be administered in an amount of from 100 mg/m2 to 500 mg/m2 of a surface area of the body.

Vinorelbine may be administered in an amount of 0.5 mg/m2 to 50 mg/m2 of a surface area of the body. Also, paclitaxel may be administered in an amount of 100 mg/m2 to 300 mg/m2 of a surface area of the body.

Trastuzumab, sold under the brand name HerceptinTM among others, is a monoclonal antibody used to treat breast cancer. Specifically it is used for breast cancer that is HER2 receptor positive. Trastuzumab is given by slow injection into a vein and injection just under the skin.

Cetuximab is an epidermal growth factor receptor (EGFR) inhibitor used for the treatment of metastatic colorectal cancer, metastatic non-small cell lung cancer and head and neck cancer. Cetuximab is a chimeric (mouse/human) monoclonal antibody given by intravenous infusion that is distributed under the trade name Erbitux in the U.S. and Canada by the drug company Bristol-Myers Squibb and outside the U.S. and Canada by the drug company Merck KGaA. In Japan, Merck KGaA, Bristol-Myers Squibb and Eli Lilly have a co-distribution.

Paclitaxel (PTX). sold under the brand name Taxol among others, is a chemotherapy medication used to treat a number of types of cancer. This includes ovarian cancer, breast cancer, lung cancer, Kaposi sarcoma, cervical cancer, and pancreatic cancer. It is given by injection into a vein.

in one embodiment, the kit / combination may include the novel crystalline form of the compound of Formula ( 1 ) of this patent document and a mitotic inhibitor. The mitotic inhibitor may be selected from BT-062, HMN-214, eribulin mesylate, vindesine, EC-1069, EC-1456, EC-531 , vintafolide, 2-methoxyestradiol. GTx-230, trastuzumab emtansine, crolibulin, D1302A-maytansinoid conjugates, IMGN-529, lorvotuzumab mertansine, SAR-3419, SAR-566658, IMP-03138, topotecan/vincristine combinations, BPH-8, fosbretabuiin tromethamine, estramustine phosphate sodium, vincristine, vinflunine, vinoreibine, RX-21 101 , cabazitaxel, 8TA-9584, vinblastine, epothilone A, patupilone, ixabepilone,

Epothilone D, paclitaxel, docetaxel, DJ-927, discodermolide, eieutherobin, and pharmaceutically acceptable salts thereof or combinations thereof. An example of the combination may include poziotinib and taxane, vinca alkaloid, or a combination thereof. The vinca alkaloid may be at least one drug selected from the group consisting of vinblastine, vincristine, vindesine and vinoreibine. The taxane may be paclitaxel or docetaxel. An example of the combination may include poziotinib and pacliataxei; or poziotinib and vinoreibine. The neoplasm may be a breast cancer in which Her2 is overexpressed.

Poziotinib may be administered in an amount of 0.1 mg to 50 mg. Also, vinoreibine may be administered in an amount of 0.5 mg/ni2 to 50 mg/m2 of a surface area of the body. Also, paclitaxel may be administered in an amount of from 100 mg/m2 to 300 mg/m2 of a surface area of the body.

Vinoreibine (NVB), sold under the brand name Navelbine among others, is a chemotherapy medication used to treat a number of types of cancer. This includes breast cancer and non-small ceil lung cancer. lt is given by injection into a vein or by mouth. Vinoreibine is in the vinca alkaloid family of medications. It is believed to work by disrupting the normal function of microtubules and thereby stopping cell division.

in one embodiment, the combination may include the novel crystalline form of the compound of Formula (1 ) of fhis patent document and an mTGR inhibitor. The mTOR inhibitor may be selected from zotarolimus, umirolimus, temsirolimus, siro!imus, sirolimus NanoCrystal, siroiimus TransDerm, sirolimus-PNP, everolimus, biolimus A9, ridaforolimus, rapamycin, TCD-10023, DE-109, MS-R001 , MS-R002, MS-R003, Perceiva, XL-765, quinacrine, PKI-587, PF-04691502, GDC-0980, dactolisib, CC-223, PWT-33597, P-7170, LY-3023414, INK-128, GDC-0Q84, D8-7423, D8-3078, CC-1 15, CBLC-137, AZD-2014, X-480, X-414, EC-0371 , VS-5584, PQR-401 , PQR-316, PQR-31 1 , PQR-309, PF-06465603, NV-128, nPT-MTOR, BC-210, WAY-600, WYE-354, WYE-687, LGR-22Q, HMPL-518, GNE-317, EC-0565, CC-214, ABTL-Q812, and pharmaceutically acceptable salts thereof or combinations thereof. An example of the combination may include poziotinib and rapamycin. The rapamycin may be in the form of an injection.

Rapamycin, also known as sirolimus, is a compound produced by the bacterium Streptomyces hygroscopicus.

The novel crystalline form of the compound of Formula ( 1 ) of this patent document may be administered in an amount of 0.1 mg to 50 mg. Also, rapamycin may be administered in an amount of 0.5 mg/m2 to 10 mg/ni2 of a surface area of the body.

in one embodiment, the kit / combination may include the novel crystalline form of the compound of Formula (1 ) of this patent document and antimetabolite. The antimetabolite may be selected from the group consisting of capecitabine, 5-fiuorouracii, gemcitabine, pemetrexed, methotrexate, 6-mercaptopurine, ciadribine, cytarabine, doxif!udine, fioxuridine, f!udarabine, hydroxycarbamide, decarbazine, hydroxyurea, and asparaginase. An example of the combination may include poziotinib and 5-iiuorouracii. The 5-fluorouracil may be in the form of an injection.

The novel crystalline form of the compound of Formula (1 ) of this patent document may be administered in an amount of 0.1 mg to 50 mg. 5-Fiuorouracii may be administered in an amount of 100 mg/m2 to 3,000 mg/m2 of a surface area of the body.

Fluorouracil (5-FU), sold under the brand name Adrucii among others, is a medication used to treat cancer. By injection into a vein it is used for colon cancer, esophageal cancer, stomach cancer, pancreatic cancer, breast cancer, and cervical cancer.[2] As a cream it is used for basal cell carcinoma. Fluorouracil is in the antimetaboiite and pyrimidine analog families of medications. How it works is not entirely clear but believed to involve blocking the action of thymidylate synthase and thus stopping the production of DNA.

in one embodiment, the kit /combination may include the novel crystalline form of the compound of Formula (1 ) of this patent document and a platinum-based antineoplastic drug. The platinum-based antineoplastic drug may be selected from the group consisting of cispiatin, carbopiatin, dicyclopiatin, eptapiatin, iobapiatin, miriplatin, nedaplatin, oxaliplatin, picoplatin, and satraplatin. An example of the combination may include poziotinib and cispiatin. The cispiatin may be in the form of an injection.

Poziotinib may be administered in an amount of 0.1 mg to 50 mg. Cispiatin may be administered in an amount of 1 mg/m2 to 100 mg/m2 of a surface area of the body.

Cispiatin is a chemotherapy medication used to treat a nu ber of cancers.

This includes testicular cancer, ovarian cancer, cervical cancer, breast cancer, bladder cancer, head and neck cancer, esophageal cancer, lung cancer, mesothelioma, brain tumors and neuroblastoma it is used by injection info a vein. Cispiatin is in the platinum-based antineoplastic family of medications it works in part by binding to, and inhibiting DNA replication.

Pharmaceutical formulations or agents in a kit may be presented in unit dose forms containing a predetermined amount of active ingredient per unit dose. As is known to those skilled in the art, the amount of active ingredient per dose will depend on the condition being treated, the route of administration and the age, weight and condition of the patient. Preferred unit dosage formulations are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of an active ingredient. Furthermore, such pharmaceutical formulations may be prepared by any of the methods well known in the pharmacy art. The novel crystalline forms or combinations of the current invention are incorporated into convenient dosage forms such as capsules, tablets, or injectable preparations. Solid or liquid pharmaceutical carriers are employed. Solid carriers include, starch, lactose, calcium sulfate dihydrate, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Liquid carriers include syrup, peanut oil, olive oil, saline, and water. Similarly, the carrier may include a prolonged release material, such as glyceryl monostearate or glyceryl dlstearate, alone or with a wax. The amount of solid carrier varies widely but, suitably, may be from about 25 mg to about 1 g per dosage unit. When a liquid carrier is used, the preparation will suitably be in the form of a syrup, elixir, emulsion, soft gelatin capsule, sterile injectable liquid such as an ampoule, or an aqueous or nonaqueous liquid suspension.

For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing with a similarly comminuted pharmaceutical carrier such as an edible carbohydrate, as, tor example, starch or mannitol. Flavoring, preservative, dispersing and coloring agent can also be present.

it should be understood that in addition to the ingredients mentioned above, the formulations may include other agents conventional in the art having regard to the type of formulation in question, for example those suitable for oral administration may include flavoring agents.

Hereinafter, this patent document will be described with reference to specific examples. However, the following embodiments are only illustrative of the present invention, and the scope of this patent document is not limited thereto. Analytical instruments and measuring methods

1. X~ray Powder Diffraction (XRPD)

X-ray Powder Diffraction (XRPD) spectroscopy was conducted on the D8 Advance (Brisker ASX, Germany) analyzer from 3°2Q to 40°2Q for the sample. When the amount of the sample is <100 mg, about 5 - 1 0 mg of the sample Is gently squeezed onto the glass slide fitted to the sample holder. When the amount of sample was >100 mg, about 100 mg of sample was gently squeezed onto the plastic sample holder so that sample surface was smooth and was just above the sample holder level.

The measurements were made as follows:

Anode material (Ka): Cu Ka (1 .54Q56A)

Scan range: 3 - 40 degrees

Generator settings: 100 mA, 40.0 kV

Scan speed: 1 sec/step

Divergent slit size (Diver slit): 0.3 degree

Anti-scatter slit: 0.3 degree

Temperature: 20 °C

Step size: 0.02 deg 2Q

Rotating: Use

Goniometer radius: 435mm

2. Differential Scanning Calorimetry (DSC)

Differential Scanning Calorimeter (DSC) analysis was conducted on the ST - 1000 (Scinco, Korea) analyzer at 30 - 350 °C 5 - 10 mg of sample was weighed into the aluminum DSC pan and sealed non-hermetica!ly with the perforated aluminum lid, and then the heat flow reaction (DSC) generated by heating the sample from 30 C G to 350 °C at a scan rate of 10 °C/min was monitored. 3. Dynamic Vapor Sorption (DVS)

Dynamic Vapor Sorption (DVS) analysis was conducted on the DVS advantage {Surface measurement system, United Kingdom) analyzer at 25 °C and relative humidity of 0 - 90%.

10 g of sample was placed in the wire mesh steam sorption balance pan and attached to the DVS-advantage dynamic vapor adsorption balance by the Surface Measurement Systems. The sample was applied to ramping profile of relative humidity (RH) of 10 - 90% in 10% increments while maintaining the sample in each step until a stable weight was achieved (99.5% step completion). After the completion of the sorption cycle, the sample was dried using the same procedure, during which the relative humidity was returned to a relative humidity below 0% all the time. The change in weight during the sorption/desorption cycle (repetition of 3 times) was recorded to determine the hygroscopicity of the sample.

4. Solid State Nuclear Magnetic Resonance (ssNMR)

For the purpose of comparing crystalline polymorphs in solid state using the nuclear magnetic resonance spectrometer, the solid state nuclear magnetic resonance (ssNMR) analysis was conducted at room temperature using a Broker Avance P 500 MHz Solid NMR system (Bruker, Germany) analyzer after placing 100 mg of sample in the 4 mm sample tube. The analysis conditions of the 13 C NMR spectra ( 13 C CP / MAS TOSS ssNMR) are as follows.

Frequency: 125.76 MHz

Spectrum width: 20 kHz

Sample rotation speed at magic angle: 5 kHz

Pulse sequence: CP (Cross Polarization) SPINAL 64 with decoupling

(Decoupling power of 80 kHz)

Repetition delay: 5 seconds

Contact time: 2 ms

Number of scans: 4096 External standard: adamantane

5. High Performance Liquid Chromatography (HPLC)

High performance liquid chromatography (HPLC) was conducted on the Agilent 1 100/1200 series HPLC Systems (Agilent, USA) analyzer for the purpose of purity and content analysis including stability test and the like. The analysis conditions of the HPLC are as follows.

Conditions of purity and content analysis: Thienopyrimidine compound of chemical formula (1 )

Column: Hydrosphere C18 (YMC), 5 pm (150 mm x 4.6 mm)

Column temperature: 30 °C

Detector: Ultraviolet absorptiometer

Detection wavelength: 254 nm

Flow rate: 1 .0 mL/min

Analysis time: 35 minutes

Eluent: NaC!C Na^PCVphosphate buffer solution (pH 2.5 ± 0.1 ) / GH 3 CN = 40/60 (v/v%)

6. ton Chromatography (IC)

Ion Chromatograph (!C) analysis was conducted on the Thermo Fisher Scientific ICS-2500 series IC Systems (Thermo Fisher Scientific, USA) analyzer for the purpose of content analysis of hydrochloric acid in fhe hydrochloride salt. The analysis conditions of the IC are as follows.

Conditions of content analysis: Thienopyrimidine compound of chemical formula (1 )

Column: lonPac AS19 (Dionex), (250 mm x 4 mm), Guard (50 mm x 4 mm)

Column temperature: 30 °C

Detector: Electrical Conductivity Detector (CD)

Suppressor: ASRS 4mm, current 40 mA

Flow rate: 1 .0 ml / min Analysis time: 30 minutes

Eluent: 10 mM potassium hydroxide solution

7. Moisture measurement

Moisture measurement was conducted using the 795KFT Titrino (Metrohm, Switzerland) Karl-Fischer moisture analyzer.

8. Melting point measurement

Melting point measurement was conducted using the IA9200 (Electrothermal, UK) melting point apparatus.

Reference Example: Preparation of the compound of chemical for uSa 111

100 g of the compound of chemical formula (1 ) prepared according to the method of Korean Patent Registration No. 1 ,013,319 and U.S. Patent No. 8,003,658 cited in the present specification or a similar method was dissolved in the mixed solution of 300 mi of dichloromethane and 300 ml of methanol, stirred at 40 °C for 30 minutes, and the insoluble solid was filtered out using a filter paper. Distillation under reduced pressure gave 93 g (yield 93%) of the title compound.

Moisture content: 2.1 °/o

Anaivsis of characteristics

The analysis results of XRPD, ssNMR, DSC and DVS for the compound of chemical formula (1 ) prepared in Reference Example are shown in FIG. 1 G, FIG. 2G and FIG. 4G, respectively.

The compound of chemical formula (1 ) prepared in Reference Example did not show any particular diffraction value in the XRPD spectrum, which shows a typical pattern of amorphous substances.

Also, the compound of Reference Example showed a broad peak in the spectrum of ssNMR, which is a typical peak pattern of amorphous structure.

Also, the compound of Reference Example did not exhibit any specific endotherm exotherm curve, as measured by the DSC (10 °C /min). Also, the compound of Reference Example exhibited a tendency to continuously absorb moisture of 1 - 3% at a relative humidity range of 10 - 90%, as measured by the DVS.

Also, the moisture content of the amorphous form was 2 1 %, as measured by the Kar!-Fischer moisture analyzer, and no characteristic melting point was observed.

Examples: Preparation of crystalline polymorphs of the compound of formula (I) and its hydrochloride salts

Example 1. Preparation of the crystalline dihydrate (2H 2 0) form of the compound of chemical formula (1)

To 10.0 g of the compound of chemical formula (1 ) of Reference Example, 80 mL of acetone and 20 ml of water were added, followed by completely dissolved the compound of chemical formula (1 } by heating under reflux and then cooled to 20 - 25 °C and stirred for 4 hours. The resulting solid was filtered and washed with 20 mL of mixed solvent of 4 : 1 of acetone : water. The filtered solid was dried at 50 °C to give 10 g (yield: 93%) of the title compound.

Moisture content: 7 5% (theoretical value of the dihydrate: 6.83%)

Analysis of characteristics

The analysis results of XRPD, ssNMR, DSC and DVS for the crystalline form prepared in Example 1 are shown in FIGS. 1 A, 2A, 3A and 4A, respectively.

Peaks with a relative intensity (i/i 0 ) of 15% or more of the crystalline form in the XRPD spectrum are summarized in Table 1 below. In the case of a peak of !/! o ³30% or more, peaks at the diffraction angle (2Q ± 0.2°) of 9.4, 1 1 .4, 13.0, 16.1 , 18.5, 19.3, 24.9 and 26.3° appeared. Table 1 :

2 Q: diffraction angle, d: distance between crystal faces,

!/!o {%): relative intensity (i: intensity of each peak; ! 0 : intensity of the largest peak).

Peaks of the chemical shift (ppm ± 0.5 ppm) in the 13 C CP/MAS TOSS ssNMR spectrum of the crystalline form are summarized in Table 2 below.

Table 2:

chemical shift (ppm ± 0.5 ppm).

The above crystalline form showed an endothermic peak of the lowest point at about 100.7 °C when running from a starting point of about 79 °C as measured by the DSC (10 °C / min) and an endothermic peak of the lowest point at about 190.8 °C when running from a starting point of about 186.5 °C. The endothermic peak at about 100.7 °C, as measured by the DSC, means the dehydration point of the crystalline hydrate form of crystalline Form I of the compound of chemical formula (1 ), and the endothermic peak at about 190.8 C G means the melting point.

The crystalline form exhibited a moisture content (theoretical moisture content of 6.83%) of about 7.5% in the Kar!-Fischer moisture analyzer and showed a condensation temperature of about 1 17 - 122 °C and a melting point of about 190 - 195 °C.

The hygroscopic degree of the crystalline form was about 2% to 5% in the relative humidity range of 10% - 90%, as measured by the DVS.

Example 2. Preparation of crystalline anhydrous Form I of the compound of chemical formula (1)

To 5.0 g of the compound of chemical formula {1 ) obtained by the method of Example 1 , 50 mL of acetone was added, and then stirred at 20 - 25 °C for 8 hours. The resulting solid was filtered and washed with 7.5 mL of acetone. The filtered solid was dried at 50 °C to obtain 3.7 g (yield: 80%) of the title compound.

Moisture content: 0.1%

Analysis of characteristics

Analysis results of XRPD, ssNMR, DSC and DVS for the crystalline form prepared in Example 2 are shown in FIGS. 1 B, 2B, 3B and 4B, respectively.

Peaks with a relative intensity (!/! 0 ) of 15% or more of the crystalline form in the XRPD spectrum are summarized in Table 3 below. In the case of a peak of i/! o >30% or more, peaks at the diffraction angle (2Q ± 0.2°) of 6.0, 10.6, 10.9, 12.1 . 16.0, 17.5, 18.3, 1 9.2, 20.3, 22.7, 23.7 and 26.3° appeared.

Table 3:

2 Q: diffraction angle, d: distance between crystal faces,

i/l o (%): relative intensity (I: intensity of each peak; l 0 : intensity of the largest peak). Peaks of the chemical shift (ppm ± 0.5 ppm) in the l 3 C CP/MAS TOSS solid state nuclear magnetic resonance(ssNMR) spectrum of the crystalline form are summarized in Table 4 below.

Table 4:

chemical shift (ppm ± 0.5 ppm).

The above crystalline form exhibited an endothermic peak of the lowest point at about 190.8 °C when running from a starting point of about 185.8 °G, as measured by the DSC {10 °C/min), and the endothermic peak at about 190.8 °C means the melting point.

The crystalline form exhibited a moisture content of about 0.1 %, as measured by the Karl-Fischer moisture analyzer, and showed a melting point at about 190 - 195 °C.

The hygroscopic degree of the crystalline form was a level of about 0.3 - 0.5% in the relative humidity range of 10% - 50%, as measured by the DVS, which is very low, and the hygroscopic degree in the range of 50 - 90% was measured to be a level of about 3%.

Example 3, Preparation of crystalline anhydrous Form I! of the compound of chemical formula (1)

To 2.0 g of the compound of chemical formula (1 ) of Example 2, 20 mL of acetonitrile was added, followed by heated and stirred at 70 - 80 °C for 2 hours and then stirred at 20 - 25 °C for 12 hours. The resulting solid was filtered and washed with 5 mL of acetonitrile. The filtered solid was dried at 50 °C to give 1 .7 g (yield:

85%) of the title compound. Moisture content: 0.3%.

Analysis of characteristics

Analysis results of XRPD, ssNMR, DSC and DVS for the crystalline form prepared in Example 3 are shown in FIGS. 1 C, 2C, 3G and 4G, respectively.

Peaks with a relative intensity (!/! 0 ) of 15% or more of the crystalline form in the XRPD spectrum are summarized in Table 5 below. In the case of a peak of l/! 0 ³3Q% or more, peaks at the diffraction angle (2Q ± 0.2°) of 4.9, 5.9, 1 1 .8, 18.8 and 19.9° appeared.

Table 5:

2 Q: diffraction angle, d: distance between crystal faces,

i/i 0 (%): relative intensity (I: intensity of each peak; l 0 : intensity of the largest peak).

Peaks of the chemical shift (ppm ± 0.5 ppm) in the l 3 C CP/MAS TOSS solid state nuclear magnetic resonance (ssNMR) spectrum of the crystalline form are summarized in Table 6 below.

Table 6:

chemical shift (ppm ± 0.5 ppm).

The above crystalline form exhibited an endothermic peak of fhe lowest point at about 184.8 °C when running from a starting point of about 181 .3 °C, as measured by the DSC {10 °C / min), and the endothermic peak at about 184.8 °C means the melting point.

The crystalline form exhibited a moisture content of abouf 0.3%, as measured by the Karl-Fischer moisture analyzer and showed a melting point of abouf 183 - 185 °C.

The hygroscopic degree of the crystalline form was a level of abouf 0.7% in the relative humidity range of 10% - 90%, as measured by the DVS, which is very low. The crystalline form was sufficiently stable at the long-term storage conditions (e.g., 25 °C. and relative humidity of 60%), accelerated conditions (e.g., 40 °C. and relative humidity of 75%), and harsh conditions (e.g., 60 °C ).

Example 4. Preparation of crystalline monohydrochSoride monohydrate (I HCI.H2O) form of the compound of chemical formula (1)

To 10 g of the compound prepared by the method of Reference Example or Examples 1 to 3, 100 mi of mixed solvent of ethanol: water (9: 1 ) was added. 4.9 ml of 35% concentrated hydrochloric acid was added, and stirred at room temperature for 6 hours. The resulting solid was filtered and washed with 30 mL of ethanol. The filtered solid was dried at 50 °C to obtain 9.1 g (yield: 82%) of fhe title compound.

Moisture content: 3.2% (theoretical value of the monohydrate: 3.3%)

Ion chromatography: 6.5% (theoretical value of the monohydrochloride:

6.92%)

Analysis of characteristics

Analysis results of XRPD, ssNMR, DSC and DVS for the crystalline form prepared in Example 4 are shown in FIGS. 1 D, 2D, 3D and 4D, respectively.

Peaks with a relative intensity (!/! 0 ) of 15% or more of the crystalline form in the XRPD spectrum are summarized in Table 7 below. In the case of a peak of !/! o ³30% or more, peaks at the diffraction angle (20 ± 0.2°) of 8.9, 13 4, 14.1 , 16.0,

19.8, 21.1 . 21 .7, 23 5, 25.7 and 32 7° appeared.

Table 7:

2 Q: diffraction angle, d: distance between crystal faces,

l/lo (%): relative intensity (I: intensity of each peak: l 0 : intensity of the largest peak).

Peaks of the chemical shift (ppm ± 0 5 ppm) in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance (ssNMR) spectrum of the crystalline form are summarized in Table 8 below.

Table 8

chemical shift (ppm ± 0 5 ppm).

The above crystalline form exhibited an endothermic peak of the lowest point at about 151 .0 °C when running from a starting point of about 126.7 °C, as measured by the DSC (10 °C/min) and an endothermic peak at about 177 7 °C. The endothermic peak at about 151 0 °C as measured by the DSC, means the dehydration point of the crystalline monohydrochloride monohydrate form, and the endothermic peak at about 177.7 °C indicates the melting point.

The crystalline form exhibited a moisture content of about 3.2% as measured by the Karl-Fischer moisture analyzer and showed a melting point of about 187 - 193 °C.

The hygroscopic degree of the crystalline form was a level of about 0.4% in the relative humidity range of 10% - 90%, as measured by the DVS, which is very low. It can be expected that the crystalline form absorbs moisture in long-term storage conditions (e.g., temperature of 25 °C. and relative humidity of 60%) and accelerated conditions (e.g., temperature of 40 °C. and relative humidity of 75%) to maintain the crystalline form of monohydrate.

Example 5. Preparation of crystalline anhydrous monohydrochloride (1 HCI) form of the compound of chemical formula (1)

To 20 g of the anhydrous compound 1 prepared by a method similar to that of Example 2, 60 ml. of DMSO was added. 5.1 ml. of 35% concentrated

hydrochloric acid was added, and the mixture was stirred at room temperature for 6 hours. The resulting solid was filtered and washed with 40 ml. of DMSO. The filtered solid was then dried at 50 °C to give 18.3 g (yield: 85%) of the title compound.

Moisture content: 0.1 %

Ion chromatography: 6.6% (theoretical value of the monohydrochloride salt:

6.92%)

Additionally, to 20 g of the anhydrous compound 1 prepared by a method similar to that of Example 2, 60 ml. of DMF was added. 5.1 ml. of 35% concentrated hydrochloric acid was added, and stirred at room temperature for 6 hours. The resulting solid was filtered and washed with 40 ml. of DMF. The filtered solid was dried at 50 °C to give 16.6 g (yield: 77%) of the title compound.

Analysis of characteristics Analysis results of XRPD, ssNMR, DSC and DVS for the crystalline form prepared in Example 5 are shown in FIGS 1 E, 2E, 3E and 4E, respectively.

Peaks with a relative intensity (i/i 0 ) of 15% or more of the crystalline form in the XRPD spectrum are summarized in Table 9 below. In the case of a peak of l/! o ³30% or more, peaks at the diffraction angle (2Q ± 0.2°) of 9.5, 12.3, 13.0, 13.5,

14.2, 21.4, 23.0, 23.2, 23.5, 27.2 and 27.5 ° appeared.

Table 9:

2 Q: diffraction angle, d: distance between crystal faces,

!/!o (%): relative intensity ( I : intensity of each peak; ! 0 : intensity of the largest peak).

Peaks of the chemical shift (ppm ± 0.5 ppm) in the 13 C CP/MAS TOSS solid state nuclear magnetic resonance (ssNMR) spectrum of the crystalline form are summarized in Table 10 below.

Table 10:

chemical shift (ppm ± 0.5 ppm).

The above crystalline form exhibited an endothermic peak of fhe lowest point at about 230.1 °C when running from a starting point of about 200.7 °C, as measured by the DSC {10 °C / min). The endothermic peak at about 230.1 C G means the melting point.

The crystalline form exhibited a moisture content of abouf 0.1 % in the Karl -Fischer moisture analyzer and showed a melting point of about 238 - 243 °C.

The hygroscopic degree of the crystalline form was at a level of about 0.35% in the relative humidity range of 10% - 90%, as measured by the DVS, which is very low. The crystalline form did not absorb moisture in long-term storage conditions (e.g., temperature of 25 °C. and relative humidity of 60%) and accelerated conditions (e.g., temperature of 40 °C. and relative humidity of 75%) to maintain the crystalline form anhydrous.

Example 6. Preparation of the amorphous form of the

monohydroch!onde (1 HCI) of the compound of chemical formula (1 )

5 g of fhe crystalline anhydrous form of the compound of formula 1 obtained in Example 5 was dissolved in 150 mL of methanol. The solution was filtered through the filter to remove foreign substances, and the filtrate was concentrated under reduced pressure to obtain 4.9 g (yield: 98%) of the title compound as a solid.

Moisture content: 1 .2 %

Analysis results of XRPD, DVS and ssNMR for the amorphous form prepared in Example 6 are shown in FIGS. 1 F, 2F, and 4F, respectively.

The amorphous form did not show any diffraction value in the XRPD spectrum.

in addition, the amorphous form exhibited a very high hygroscopic degree at the relative humidity range of 10 - 90%, as measured by the DVS. Through this, it is expected to be unstable by absorbing moisture under long-term storage conditions (25 °C temperature and relative humidity of 80%) and accelerated conditions (40 °C temperature and relative humidity of 75%). Actually, a moisture absorption of 7 - 9% was confirmed under the 25 °C., 60% relative humidity condition and the 40 °C, 75% relative humidity condition.

in addition, the moisture content of the amorphous form was 1 .2%, as measured by the Karl-Fischer moisture analyzer, and no characteristic melting point was observed.

Test Example 1 : Comparative test of solubility o! amorphous form and crystalline polymorphs of the hydrochloride salt

To compare the solubility of amorphous hydrochloride salt form and crystalline hydrochloride salt polymorphs, each of the polymorphs and amorphous form of fhe hydrochloride salt of the compound of chemical formula (1 ) prepared in Examples 4 to 6 was used to prepare samples under the following conditions according to non-lonized water and acidity (pH). Thereafter, each solution was analyzed by high performance liquid chromatography (HPLG) according to the measurement conditions of the content of the compound of chemical formula (1 ) to measure the dissolved amount (LOD: 0.1 pg/mL) based on the compound of chemical formula (1 ). The results calculated from the measured values are shown in Table 1 1 below.

Specifically, 5 mg of each polymorph was added to 5 ml of water and mixed using a Voltamixer at 20 25 °C. Thereafter, the filtrate obtained by filtrating using GH Polypro membrane Acrodisc, PALL (pore size 0.2 pm) was diluted with a dilution solvent for high performance liquid chromatography (HPLG) at a ratio of 1/100, to obtain the samples.

Table 1 1 :

As shown in the above Table 1 1 , the solubility of the hydrochloride salt of the compound of chemical formula (1 ) was remarkably higher than that of the compound of chemical formula (1 ) (less than 1.0 pg/mL), and the crystalline anhydrous form of the hydrochloride salt among crystalline polymorphs showed the highest solubility in wafer.

Accordingly, the crystalline anhydrous hydrochloride salt form of the compound of chemical formula (1 ) is expected to be the most advantageous in terms of pharmaceutical composition when considering elution etc.

Test Example 2: Comparative test of stability of amorphous form and crystalline polymorphs of the hydrochloride sait

To compare the stability of amorphous hydrochloride sait form and crystalline hydrochloride salt polymorphs, each of samples of the polymorphs and amorphous form of the hydrochloride salt of the compound of chemical formula (1 ) prepared in Examples 4 to 6 was left for 4 weeks under long term conditions (e.g., temperature of 25 ± 2 °C and relative humidity of 60 ± 5%) and accelerated conditions (e.g., temperature of 40 °C and relative humidity of 75%). Each sample was analyzed by high performance liquid chromatography (HPLC) according to the purity measurement conditions of the compound of chemical formula (1 ). The purity measurement values (%) are shown in Table 12 below.

Table 12:

As shown in Table 12, the crystalline hydrochloride sait form of the compound of chemical formula (1 ) was stable compared to the amorphous hydrochloride salt form of the compound of chemical formula (1 ), and in particular, the crystalline anhydrous form of the hydrochloride salt of the compound of chemical formula (1 ) showed the best results.

Accordingly, through Comparative Tests 1 and 2, the crystalline anhydrous hydrochloride salt form of the compound of chemical formula (1 ) is expected to be the most advantageous in terms of the pharmaceutical composition when considering various physicochemical properties such as solubility, purity, stability, hygroscopicity, and melting point, etc.