WO2005077945A2 - Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors - Google Patents

Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors Download PDF

Info

Publication number
WO2005077945A2
WO2005077945A2 PCT/US2005/003728 US2005003728W WO2005077945A2 WO 2005077945 A2 WO2005077945 A2 WO 2005077945A2 US 2005003728 W US2005003728 W US 2005003728W WO 2005077945 A2 WO2005077945 A2 WO 2005077945A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
formula
alkyl
hydrogen
substituted
Prior art date
Application number
PCT/US2005/003728
Other languages
French (fr)
Other versions
WO2005077945A3 (en
Inventor
Bang-Chi Chen
Roberto Droghini
Jean Lajeunesse
John D. Dimarco
Michael Galella
Ramakrishnan Chidambaram
Original Assignee
Bristol-Myers Squibb Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=34864498&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005077945(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to BRPI0507476A priority Critical patent/BRPI0507476B8/en
Priority to KR1020067015839A priority patent/KR101310427B1/en
Priority to DE602005018601T priority patent/DE602005018601D1/en
Priority to AU2005212405A priority patent/AU2005212405B2/en
Priority to DK05722772.0T priority patent/DK1711481T4/en
Priority to CA2555291A priority patent/CA2555291C/en
Priority to JP2006552303A priority patent/JP5148115B2/en
Priority to EP05722772.0A priority patent/EP1711481B2/en
Priority to IN6567DEN2014 priority patent/IN2014DN06567A/en
Priority to SI200530905T priority patent/SI1711481T2/en
Priority to ES05722772T priority patent/ES2337272T5/en
Priority to PL05722772T priority patent/PL1711481T5/en
Priority to AT05722772T priority patent/ATE453630T1/en
Priority to NZ548613A priority patent/NZ548613A/en
Priority to CN2005800119166A priority patent/CN1980909B/en
Application filed by Bristol-Myers Squibb Company filed Critical Bristol-Myers Squibb Company
Publication of WO2005077945A2 publication Critical patent/WO2005077945A2/en
Publication of WO2005077945A3 publication Critical patent/WO2005077945A3/en
Priority to ZA2006/06242A priority patent/ZA200606242B/en
Priority to IL177280A priority patent/IL177280A/en
Priority to NO20063780A priority patent/NO338049B1/en
Priority to HK06112879.5A priority patent/HK1091835A1/en
Priority to HRP20100166TT priority patent/HRP20100166T4/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/56Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/28Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and unsaturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/28Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a non-condensed six-membered aromatic ring of the carbon skeleton
    • C07C237/42Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a non-condensed six-membered aromatic ring of the carbon skeleton having nitrogen atoms of amino groups bound to the carbon skeleton of the acid part, further acylated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings

Definitions

  • the present invention relates to processes for preparing 2-aminothiazole-5- aromatic carboxamides which are useful as kinase inhibitors, such as inhibitors of protein tyrosine kinase and p38 kinase, intermediates and crystalline forms thereof.
  • Ar is aryl or heteroaryl
  • L is an optional alkylene linker
  • R 2 , R 3 , Ri, and R 5 are as defined in the specification herein, are useful as kinase inhibitors, in particular, inhibitors of protein tyrosine kinase and p38 kinase. They are expected to be useful in the treatment of protein tyrosine kinase-associated disorders such as immunologic and oncological disorders [see, US Pat. No. 6,596,746 (the '746 patent), assigned to the present assignee and incorporated herein by reference], and p38 kinase-associated conditions such as inflammatory and immune conditions, as described in US patent application Serial No. 10/773,790, filed February 6, 2004, claiming priority to US Provisional application Serial No. 60/445,410, filed February 6, 2003 (hereinafter the '410 application), both of which are also assigned to the present assignee and incorporated herein by reference.
  • the compound of formula (IN), ' ⁇ -(2-Chloro-6-methylphenyl)-2-[[6-[4-(2- hydroxyethyl)-l-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamide, is an inhibitor of SRC/ABL and is useful in the treatment of oncological diseases.
  • IV Other approaches to preparing 2-aminothiazole-5-carboxamides are described in the '746 patent and in the '410 application.
  • the '746 patent describes a process involving treatment of chlorothiazole with n-BuLi followed by reaction with phenyl isocyanates to give chlorothiazole-benzamides, which are further elaborated to aminothiazole-benzamide final products after protection, chloro-to-amino substitution, and deprotection, e.g.,
  • the '410 application describes a multi-step process involving first, converting N-unsubstituted aminothiazole carboxylic acid methyl or ethyl esters to bromothiazole carboxylic acid esters via diazotization with tert-butyl nitrite and subsequent CuBr 2 treatment, e.g.,
  • This invention is related to processes for the preparation of 2-aminothiazole-5- aromatic amides having the formula (I), wherein L, Ar, R , R 3 , R , R 5 , and m are as defined below, comprising reacting a compound having the formula (H),
  • Q is the group -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, and Ar, L, R 2 , R 3 , and m are as defined below, with a halogenating reagent in the presencp of water followed by a thiourea compound having the formula (HI),
  • Ar is the same in formulae (I) and (IT) and is aryl or heteroaryl;
  • L is the same in formulae (I) and (U) and is optionally-substituted alkylene;
  • R 2 is the same in formulae (I) and (II), and is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo
  • R 3 is the same in formulae (I) and (U), and is selected from hydrogen, halogen, cyano, haloalkyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aryl, heteroaryl, cycloalkyl, and heterocyclo
  • 1 ⁇ is (i) the same in each of formulae (I) and (IH), and (ii) is independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo, or alternatively, R 4 is taken
  • the present invention is directed to crystalline forms of the compound of formula (IV).
  • Figure 1 shows a simulated (bottom) (calculated from atomic coordinates generated at room temperature) and experimental (top) pXRD patterns for crystalline monohydrate of the compound of formula (IV).
  • Figure 2 shows a DSC and TGA of the of the monohydrate crystalline form of the compound of Formula (IV).
  • Figure 3 shows a simulated (bottom) (from atomic parameters refined at room temperature) and experimental (top) pXRD patterns for crystalline butanol solvate of the compound of formula (IN).
  • Figure 4 shows a simulated (bottom) (from atomic parameters refined at - 40°C) and experimental (top) pXRD patterns for crystalline ethanol solvate of the compound of formula (IN).
  • Figure 5 shows a simulated (bottom) (from atomic parameters refined at room temperature) and experimental (top) pXRD patterns for crystalline neat form ( ⁇ -6) of the compound of formula (IN).
  • Figure 6 shows a simulated (bottom) (from atomic parameters refined at room temperature) and experimental (top) pXRD patterns for crystalline neat form (T1H1-7) of the compound of formula (IN).
  • Ph phenyl
  • Boc tert-butyloxycarbonyl
  • DMSO dimethyl sulf oxide
  • DPPF l,l'-bis(diphenylphosphino)ferrocene
  • HATU O-benzotriazol-1-ylO ⁇ , ⁇ , ⁇ '-tetramethyluronium hexafluorphosphate
  • LDA lithium di-isopropyl amide
  • POCl 3 phosphorous oxychloride
  • EDC or EDCI 3-ethyl-3'-(dimethylamino)propyl- carbodiimide
  • DIPEA diisopropylethylamine
  • HOBt 1-hydroxybenzotriazole hydrate
  • NBS N-bromosuccinamide
  • NMP N-methyl-2-pyrrolidinone
  • NaH sodium hydride
  • NaOH sodium hydroxide
  • Na 2 S 2 O 3 sodium thiosulfate
  • HPLC high performance liquid chromatography
  • LC/MS high performance liquid chromatography/mass spectrometry
  • XRPD x-ray powder diffraction pattern
  • pXRD x-ray powder diffraction pattern
  • alkyl as used herein by itself or as part of another group refers to straight and branched chain saturated hydrocarbons, containing 1 to 20 carbons, 1 to
  • substituted alkyl refers to an alkyl group substituted with one or more substituents (for example 1 to 4 substituents, or 1 to 2 substituents) at any available point of attachment.
  • substituents may be selected from one or more (or 1 to 3) of the following groups:
  • U 3 and U 4 are each independently a single bond, alkylene, alkenylene, or alkynylene; wherein, in group (i),
  • R g R h where R g and R h are each independently hydrogen, alkyl, or alkyl substituted with a group R f ; and; ' wherein, (v) R f is at each occurrence independently selected from alkyl, halogen, cyano, hydroxy, -O(alkyl), SH, -S(alkyl), amino, alkylamino, haloalkyl, haloalkoxy, or a lower alkyl substituted with one to two of halogen, cyano, hydroxy, -O(alkyl), SH, -S(alkyl), amino, alkylamino, haloalkyl, and/or haloalkoxy, and wherein,
  • alkenyl refers to straight or branched chain radicals of 2 to 20 carbons, alternatively 2 to 12 carbons, and/or 1 to 8 carbons in the normal chain, which include one to six double bonds in the normal chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3- pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3- nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8,12-tetradecatrienyl, and the like.
  • a substituted alkenyl refers to an alkenyl having one or more substituents (for example 1 to 3 substituents, or 1 to 2 substituents), selected from those defined above for substituted alkyl.
  • alkynyl refers to straight or branched chain hydrocarbon groups having 2 to 12 carbon atoms, alternatively 2 to 4 carbon atoms, and at least one triple carbon to carbon bond, such as ethynyl, 2-propynyl, 3-butynyl, 2-butynyl, 4-pentynyl, 3-pentynyl, 2-hexynyl, 3- hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonynyl, 4-decynyl,3- undecynyl, 4-dodecynyl and the like.
  • a substituted alkynyl refers to an alkynyl having one or more substituents (for example 1 to 4 substituents, or 1 to 2 substituents), selected from those defined above for substituted alkyl.
  • alkyl When the term “alkyl” is used as a suffix with another group, such as in (aryl)alkyl or arylalkyl, this conjunction is meant to refer to a substituted alkyl group wherein at least one of the substituents is the specifically named group in the conjunction.
  • (aryl)alkyl refers to a substituted alkyl group as defined above wherein at least one of the alkyl substituents is an aryl, such as benzyl.
  • groups designated -O(alkyl) and -S(alkyl) it should be understood that the points of attachment in these instances are to the oxygen and sulfur atoms, respectively.
  • alkylene groups as defined are divalent, i.e., with two single bonds for attachment to two other groups, they are termed “alkylene” groups.
  • alkenyl groups as defined above and alkynyl groups as defined above, respectively are divalent radicals having single bonds for attachment to two other groups, they are termed “alkenylene groups” and “alkynylene groups” respectively.
  • alkylene, alkenylene and alkynylen'e groups include:
  • Alkylene groups may be optionally independently substituted as valence allows with one or more groups as defined for substituted alkyl groups.
  • a substituted alkylene group would be optionally independently substituted as valence allows with one or more groups as defined for substituted alkyl groups.
  • a substituted alkylene group would be optionally independently substituted as valence allows with one or more groups as defined for substituted alkyl groups.
  • I (CH 2 ) 3 -C include CH CH 2 CH 2 anc ⁇ F J and so forth.
  • cycloalkyl refers to optionally-substituted saturated and partially unsaturated (containing 1 or 2 double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, or 3 to 7 carbons, forming the ring.
  • the further rings of multi-ring cycloalkyls may be either fused, bridged and/or joined through one or more spiro unions.
  • Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, cyclopentenyl, cycloheptenyl, cyclooctenyl, cyclohexadienyl, cycloheptadienyl,
  • Each reference to a cycloalkyl is intended to include both substituted and unsubstituted cycloalkyl groups as defined immediately below, unless reference is made to a particular selection of substituents to be made for the cycloalkyl (e.g., wherein cycloalkyl is substituted with one or more groups R f .)
  • substituents for the cycloalkyl groups may be selected from the following:
  • halogen e.g., a single halo substituent or multiple halo substitutents forming, in the latter case, groups such as a perfluoroalkyl group or an alkyl group bearing Cl 3 or CF 3
  • halogen e.g., a
  • R f is at each occurrence independently selected from alkyl, halogen, cyano, hydroxy, -O(alkyl), SH, -S (alkyl), amino, alkylamino, haloalkyl, haloalkoxy, or a lower alkyl substituted with one to two of halogen, cyano, hydroxy, -O(alkyl), SH, -S(alkyl), amino, alkylamino, haloalkyl, and/or haloalkoxy, and wherein,
  • cycloalkylene refers to a "cycloalkyl” group as defined above which is a linking group such as and the like.
  • alkoxy refers to an alkyl or substituted alkyl group as defined above bonded through an oxygen atom (-O-), i.e., the group -OR i5 wherein R ; is alkyl or substituted alkyl.
  • alkylthio refers to an alkyl or substituted alkyl group as defined above bonded through a sulfur atom (-S-), i.e., the group -SR i5 wherein R 4 is alkyl or substituted alkyl.
  • alkoxycarbonyl refers to a carboxy group ( C— O ) linked to an alkyl radical (i.e., to form CO 2 R j ), wherein R j is as defined above for acyl.
  • alkylamino refers to amino groups wherein one or both of the hydrogen atoms is replaced with an alkyl group, i.e., NR k Ri, wherein one of R and Ri is hydrogen and the other is alkyl, or both R k and Rj are alkyl .
  • halo or “halogen” refers to chloro, bromo, fluoro and iodo.
  • haloalkyl means a substituted alkyl having one or more halo substituents.
  • haloalkyl includes mono, bi, and trifluoromethyl.
  • haloalkoxy means an alkoxy group having one or more halo substituents.
  • haloalkoxy includes OCF 3 .
  • aromatic homocyclic i.e., hydrocarbon
  • biphenyl i.e., naphthyl (including 1-naphthyl and 2-naphthyl) and antracenyl
  • aromatic homocyclic i.e., hydrocarbon
  • biphenyl i.e., biphenyl
  • naphthyl including 1-naphthyl and 2-naphthyl
  • antracenyl such as phenyl, biphenyl, naphthyl (including 1-naphthyl and 2-naphthyl) and antracenyl]
  • additional rings either cycloalkyl, heterocyclo or heteroaryl fused thereto. Examples include:
  • Each reference to an aryl is intended to include both substituted and unsubstituted aryl groups as defined herein, unless reference is made to a particular selection of substituents to be made for the aryl (e.g., as when aryl is substituted with one or more groups R f , above).
  • substituents to be made for the aryl e.g., as when aryl is substituted with one or more groups R f , above.
  • the optional substituents for the aryl groups may be selected from those recited above, as valence allows, for cycloalkyl groups.
  • heteroaryl refers to optionally-substituted monocyclic and bicyclic aromatic rings containing from 5 to 10 atoms, which includes 1 to 4 hetero atoms such as nitrogen, oxygen or sulfur, and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocyclo ring, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized.
  • heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, indolyl, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolo
  • Each reference to a heteroaryl is intended to include both substituted and unsubstituted heteroaryl groups as defined herein, unless reference is made to a particular selection of substituents to be made for the heteroaryl (e.g., as when heteroaryl is substituted with one or more groups R f , above).
  • substituents to be made for the heteroaryl e.g., as when heteroaryl is substituted with one or more groups R f , above.
  • the optional substituents for the heteroaryl groups may be selected from those recited above, as valence allows, for cycloalkyl groups.
  • heterocyclic or “heterocyclo” as used herein by itself or as part of another group refer to non-aromatic, optionally substituted, fully saturated or partially unsaturated cyclic groups (for example, 3 to 13 member monocyclic, 7 to 17 member bicyclic, or 10 to 20 member tricyclic ring systems, or containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring.
  • Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized.
  • the heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows.
  • the rings of multi-ring heterocycles may be fused, bridged and/or joined through one or more spiro unions.
  • heterocyclic groups include oxetanyl, imidazolinyl , oxazolidinyl, isoxazolinyl, thiazolidinyl, isothiazolidinyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, -2-oxopiperidinyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, 1,3-dioxolane and tetrahydro-l,l-dioxothienyl,
  • Each reference to a heterocyclo is intended to include both substituted and unsubstituted heterocyclo groups as defined herein, unless reference is made to a particular selection of substituents to be made for the heterocyclo (e.g., as when heterocyclo is substituted with one or more groups R f , above).
  • substituents for the heterocyclo groups may be selected from those recited above, as valence allows, for cycloalkyl groups.
  • ring encompasses homocyclic (i.e., as used herein, all the ring atoms are carbon) or "heterocyclic” (i.e., as used herein, the ring atoms include carbon and one to four heteroatoms selected from N, O and /or S, also referred to as heterocyclo), where, as used herein, each of which (homocyclic or heterocyclic) may be saturated or partially or completely unsaturated.
  • aryl e.g., phenyl
  • cycloalkyl e.g., cyclohexyl
  • heterocyclo e.g., pyrrolidinyl
  • heteroaryl e.g., imidazolyl
  • the reference is intended to include rings having 0 to 3, or 0 to 2, substituents selected from those recited above for the aryl, cycloalkyl, heterocyclo and/or heteroaryl groups, as appropriate.
  • heteroatoms shall include oxygen, sulfur and nitrogen.
  • carbocyclic means a saturated or unsaturated monocyclic or bicyclic ring in which all atoms of all rings are carbon. Thus, the term includes cycloalkyl and aryl rings. The carbocyclic ring may be substituted in which case the substituents are selected from those recited above for cycloalkyl and aryl groups.
  • Base when used herein includes metal oxides, hydroxides or alkoxides, hydrides, or compounds such as ammonia, that accept protons in water or solvent.
  • exemplary bases include, but are not limited to, alkali metal hydroxides and alkoxides (i.e., MOR, wherein M is an alkali metal such as potassium, lithium, or sodium, and R is hydrogen or alkyl, as defined above, or where R is straight or branched chain - 5 alkyl, thus including, without limitation, potassium hydroxide, potassium tert-butoxide, potassium tert-pentoxide, sodium hydroxide, sodium tert- butoxide, lithium hydroxide, etc.); other hydroxides such as magnesium hydroxide (Mg(OH) 2 ) or calcium hydroxide (Ca(OH) 2 ); alkali metal hydrides (i.e., MH, wherein M is as defined above, thus including, without limitation, sodium hydride and lithium hydride); alkylated disilazides, such as, for example, potassium hexamethyldisilazide and lithium hexamethyldisilazide; carbonates such as potassium
  • Coupled reagent refers to a reagent used to couple a carboxylic acid and an amine or an aniline to form an amide bond. It may include a coupling additive, such as CDI, HOBt, HOAt, HODhbt, HOSu, or NEPIS, used in combination with another coupling reagent to speed up coupling process and inhibit side reactions.
  • a coupling additive such as CDI, HOBt, HOAt, HODhbt, HOSu, or NEPIS
  • Particular peptide-coupling reagents may include CDI, DCC, EDC, BBC, BDMP, BOMI, HATU, HAPyU, HBTU, TAPipU, AOP, BDP, BOP, PyAOP, PyBOP, TDBTU, TNTU, TPTU, TSTU, BEMT, BOP-C1, BroP, BTFFH, CIP, EDPBT, Dpp-Cl, EEDQ, FDPP, HOTT-PF6, TOTT-BF4, PyBrop, PyClop, and TFFH.
  • Peptide Coupling Reagents Names, Acronyms and References," Albany Molecular Research, Inc., Technical Reports, Vol. 4, No. 1, incorporated herein by reference.
  • halogenating agent or “halogenating reagent” mean an agent or agents capable of halogenating compounds of formula (H) herein.
  • Halogenating reagents include inorganic and organic halogenating reagents. Examples of inorganic halogenating reagents include chlorine, bromine, iodine, fluorine, and sodium hypochlorite.
  • Organic halogenting reagents include N-chlorosuccinimide (NCS), N- bromosuccinimide (NBS), N-iodosuccinimide (NIS), l,3-dichloro-5,5- dimethylhydantoin, l,3-dibromo-5,5-dimethylhydantoin, and l,3-diiodo-5,5- dimethylhydantoin.
  • High yield as used herein means a yield of greater than 80%, greater than 85%, than 90%, or than 95%.
  • leaving group means groups having the capability of being displaced upon reaction with a nucleophile including I, Br, CI, R 10 SO O- (wherein R 10 is alkyl, substituted alkyl, aryl, or heteroaryl, as defined herein), and weak bases, such as, for example, HSO 4 -.
  • Examples of leaving groups include I, Br, CI, and ions of methyl sulfate, mesylate (methane sulfonate), trifluromethanesulfonate, and tosylate (p- toluenesulf onate) .
  • the group Q is -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, i.e., Q has the capability of being displaced upon reaction with a nucleophile.
  • Suitable solvent as used herein is intended to refer to a single solvent as well as mixtures of solvents.
  • Solvents may be selected, as appropriate for a given reaction step, from, for example, aprotic polar solvents such as DMF, DMA, DMSO, dimethylpropyleneurea, N-methylpyrrolidone (NMP), and hexamethylphosphoric triamide; ether solvents such as diethyl ether, THF, 1,4-dioxane, methyl t-butyl ether, dimethoxymethane, and ethylene glycol dimethyl ether; alcohol solvents such as MeOH, EtOH, and isopropanol; and halogen-containing solvents such as methylene chloride, chloroform, carbon tetrachloride, and 1,2-dichloroethane. Mixtures of solvents may also include biphasic mixtures.
  • slurry as used herein is intended to mean a saturated solution of the compound of Formula (IV) and an additional amount of the compound of Formula (IV) to give a heterogeneous solution of the compound of Formula (IV) and a solvent.
  • the present invention describes crystalline forms of the compound of formula (IN) in substantially pure form.
  • substantially pure means a compound having a purity greater than 90 percent, including 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, and 100 percent.
  • a crystalline form of the compound of the formula (IN) can be substantially pure in having a purity greater than 90 percent, where the remaining less than 10 percent of material comprises other form(s) of the compound of the formula (IV), and/or reaction and/or processing impurities arising from its preparation.
  • a crystalline form of the compound of the formula (IV) in substantially pure form may therefore be employed in pharmaceutical compositions to which other desired components are added, for example, excipients, carriers, or active chemical entities of different molecular structure.
  • crystalline forms of the compound of formula (IN) loses its crystalline structure, and is therefore referred to as a solution of the compound of formula (IN). All forms of the present invention, however, may be used for the preparation of liquid formulations in which the drug is dissolved or suspended. In addition, the crystalline forms of the compound of formula (IV) may be incorporated into solid formulations.
  • a therapeutically effective'amount of the crystalline forms of the compound of formula (IN) is combined with a pharmaceutically acceptable carrier to produce the pharmaceutical compositions of this invention.
  • therapeutically effective amount it is meant an amount that, when administered alone or an amount when administered with an additional therapeutic agent, is effective to prevent, suppress or ameliorate the disease or condition or the progression of the disease or condition.
  • This invention is related to a process for the preparation of 2-aminothiazolyl - 5-aromatic amides which are useful as inhibitors of kinases, particularly protein tyrosine kinase and p38 kinase.
  • the process involves halogenation of ⁇ -(P*)oxy- ⁇ , ⁇ - unsaturated carboxyl aromatic amides (II) (wherein P* is as defined herein), such as ⁇ -(alkyl)oxy- ⁇ , ⁇ -unsaturated carboxyl benzamides, and reaction with thioureas (Iff) to give 2-aminothiazole-5-aromatic amides of formula (I).
  • Desired substituents on the 2- amino group and/or the 5-aromatic group can be attached either before or after the aminothiozole formation.
  • the compound of formula (I) is prepared via reaction of a thiourea wherein I is hydrogen, and the R 4 hydrogen atom is then elaborated to more functionalized groups such as, in one embodiment, substituted pyrimidines.
  • the compound of formula (I) is prepared via reaction of a thiourea wherein R 4 is a pyrimidinyl, and the pyrimidinyl optionally is further elaborated with additional substituents, as desired.
  • Ar is aryl or heteroaryl, more preferably aryl, even more preferably optionally-substituted phenyl.
  • the inventive process may be carried out where a linker group L is present, as in formula I, but advantageously the Ar group is directly attached to the carboxylamide nitrogen atom, as in formula (la).
  • the desired substituents may be attached to the group Ar either before or after the halogenation and cyclization process.
  • the thiourea compounds (IH) may be prepared, prior to the cyclization, having desired groups R and R 5 , corresponding to the groups on the desired final product, or alternatively, the desired groups may be attached to the amino-thiazolyl after cyclization.
  • thiourea compounds (Iff) may be prepared and used in the reaction wherein and R 5 are both hydrogen, or R 4 and R 5 are other groups, different from those of the final desired product, and then, after formation of the aminothiazole (I) or (la), the groups R 4 and R 5 are elaborated to the substituents of the final desired product. All such alternative embodiments and variations thereof are contemplated as within the scope of the present invention.
  • the group R is hydrogen or lower alkyl, more preferably hydrogen, and R 3 is preferably hydrogen.
  • ⁇ -alkyloxy- , ⁇ -unsaturated carboxyl benzamides are thus preferred, including ⁇ -substituted and ⁇ -unsubstituted ⁇ -alkyloxy- , ⁇ -unsaturated carboxyl benzamides, with the latter more preferred, wherein the phenyl group of the benzamide is optionally substituted as recited above for Ar in formula (la).
  • ⁇ -unsubstituted ⁇ -alkyloxy-oc, ⁇ -unsaturated carboxyl benzamides are ⁇ - ethoxy aciyl benzamides, again, wherein the phenyl group of the benzamide is optionally substituted as recited above for Ar.
  • the halogenating agent(s) used in the process may be any agent or agents as defined herein capable of halogenating compounds (ff), as previously defined herein.
  • Preferred agents include NBS and the N-halohydantoins.
  • Thiourea compounds (Iff) include unsubstituted thioureas, N-monosubstituted thioureas, and N,N-disubstituted thioureas.
  • the steps of halogenation and cyclization are carried out in a suitable solvent which may include one or more solvents such as hydrocarbons, ethers, esters, amides and ketones with ethers, with dioxane preferred.
  • the ⁇ -(P*)oxy-acryl benzamides (lib), wherein R 2 and R 3 are hydrogen, and P* is as previously defined herein, preferably a lower alkyl, are halogenated with a halogenating agent, such as NBS, in a suitable solvent, in the presence of water, then cyclized with unsubstituted thiourea (Ilia).
  • a halogenating agent such as NBS
  • Leaving groups X and Y are preferably I, Br, CI, or R ⁇ oSO 2 O- (wherein Rio is alkyl, substituted alkyl, aryl, or heteroaryl, as defined herein), more preferably X and Y are selected from I, Br, CI, methyl sulfate, mesylate, trifluoromethanesulfonate, and tosylate, even more preferably from CI and Br.
  • pyrimidines 4 include bis-halogen and sulfonyloxy substituted pyrimidines with the former such as bis-chloro substituted pyrimidines preferred.
  • this step is carried out in the presence of a base, wherein the bases may include alkali hydride and alkoxides with the latter such as sodium t-butoxide preferred.
  • Suitable solvent(s) include solvents such as hydrocarbons, ethers, esters, amides, ketones and alcohols, or mixtures of the above solvents, with ether such as THF preferred.
  • R 2 o and R 2 ⁇ can both be hydrogen, or R 20 and R 2 ⁇ can be independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R 20 and R 2i can be taken together to form a heterocyclo.
  • R 2 o and R 2 ⁇ are taken together so that NHR 20 R ⁇ forms an optionally-substituted piperazine, more preferably a piperazine N'- substituted with substituted alkyl, more preferably hydroxyethyl.
  • this step is carried out in the presence of a base, including inorganic and organic bases, with organic bases such as tertiary amines preferred.
  • Suitable solvent(s) include solvents such as hydrocarbons, halogenated hydrocarbons, ethers, esters, amides, ketones, lactams and alcohols, and mixtures of the above solvents, with alcohols such as n-butanol as one nonlimiting example, and DMF (dimethylformamide), DMA (dimethylacetamide) and NMP (N-methylpyrrolidine) as other examples.
  • the compounds of formula (Id) thus formed may optionally be further elaborated as desired and/or purified and crystallized.
  • the process comprises preparing a compound of the formula (le),
  • Zi and Z 5 are selected from hydrogen, alkyl, halogen, hydroxy, and alkoxy;
  • Q is the group -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, and Zi, Z , Z 3 , Z , and Z5 are as defined above, with a halogenating reagent followed in the presence of water by a thiourea compound having the formula, to provide the compound having the formula (le),
  • R* is hydrogen, whereby the process provides a compound having the formula (If),
  • R 4 may be a group having the formula
  • R 15 , R ⁇ 6 , Zi, Z 2 , Z 3 , Z 4 , Z 5 , R 2 o and R 2 ⁇ are as defined herein.
  • R 4 is a group having the formula, wherein Y, R 15 and R ⁇ 6 are as defined herein, wherein said process provides a compound having the formula (H),
  • R is a group having the formula
  • the process may further comprise reacting the compound of the formula
  • X and Y are leaving groups
  • R 15 and R ⁇ 6 are independently selected from hydrogen, alkyl and substituted alkyl, to provide a compound having the formula, wherein Y, R i5 , R ⁇ 6 , Zi, Z 2 , Z 3 , Z 4 , and Z 5 are as defined above.
  • the process may further comprise reacting the compound of the formula
  • Y, R ⁇ 5 , R ⁇ 6 , Z ls Z 2 , Z 3 , Z 4 , and Z 5 are as defined above.
  • Compounds (Ig) may optionally further be reacted with an amine having the formula ' NHR 2 oR 2 i, wherein R 2 o and R 2 ⁇ are independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R 2 o and R 2 ⁇ can be taken together to form a heterocyclo, to provide a compound having the formula (Hi), wherein R ⁇ 5 , R ⁇ 6 , Zi, Z 2 , Z 3 , Z , Z 5 , R 2 o and R 2i are as defined above.
  • the amine NHR 20 R 2 ⁇ is piperazine in turn optionally substituted with hydroxy(alkyl), more preferably hydroxyethyl.
  • the amine NHR20R21 is NHR20R21
  • the process may further comprise reacting the compound of the formula
  • the present invention provides a crystalline monohydrate of the compound of formula (IV)
  • the monohydrate form is in substantially pure form.
  • the monohydrate form is in substantially pure form, wherein substantially pure is greater than 90 percent pure.
  • (IV) is characterized by an x-ray powder diffraction pattern substantially in accordance with that shown in Figure 1.
  • the monohydrate form of the compound of Formula (TV) is characterized by differential scanning calorimetry thermogram and a thermogravimetric anaylsis substantially in accordance with that shown in Figure 2.
  • the monohydrate form of the compound of Formula (IV) is characterized by unit cell parameters approximately equal to the following:
  • the present invention provides a crystalline butanol solvate of the compound of formula (IV)
  • the present invention is directed to the crystalline ethanol solvate of the compound of formula (IV).
  • the present invention is directed to the crystalline neat form of the compound of formula (TV).
  • the present invention describes a pharmaceutical composition comprising a therapeutically effective amount of at least one of the crystalline forms of the compound of Formula (TV) and a pharmaceutically acceptable carrier.
  • the present invention describes a method for the treatment of cancer which comprises administering to a host in need of such treatment a therapeutically effective amount of at least one of the crystalline forms of the compound of Formula (TV).
  • the present invention describes a method of treating oncological disorders which comprises administering to a host in need of such treatment a therapeutically effective amount of at least one of the crystalline forms of the compound of Formula (IN), wherein the disorders are selected from chronic myelogenous leukemia (CML), gastrointestinal stromal tumor (GIST), small cell lung cancer (SCLC), non-small cell lung cancer ( ⁇ SCLC), ovarian cancer, melanoma, mastocytosis, germ cell tumors, acute myelogenous leukemia (AML), pediatric sarcomas, breast cancer, colorectal cancer, pancreatic cancer, and prostate cancer.
  • CML chronic myelogenous leukemia
  • GIST gastrointestinal stromal tumor
  • SCLC small cell lung cancer
  • ⁇ SCLC non-small cell lung cancer
  • ovarian cancer melanoma
  • mastocytosis germ cell tumors
  • AML acute myelogenous leukemia
  • pediatric sarcomas breast cancer, colorectal cancer
  • the present invention is directed to a use of the at least one of the crystalline forms of the compound of Formula (IN), in the preparation of a medicament for the treatment of oncological disorders, such as those described herein.
  • the present invention is directed to a method of treating of oncological disorders, as described herein, which are resistant or intolerant to Gleevec ® (STI-571), comprising administering to a host in need of such treatment a therapeutically effective amount of the compound of Formula (IN) or at least one of the crystalline forms of the compound of Formula (TV).
  • the compounds of formula (I) prepared according to the inventive process herein inhibit protein tyrosine kinases, especially Src-family kinases such as Lck, Fyn, Lyn, Src, Yes, Hck, Fgr and Blk, and are thus useful in the treatment, including prevention and therapy, of protein tyrosine kinase-associated disorders such as immunologic and oncologic disorders.
  • the compounds of formula (I) also may inhibit receptor tyrosine kinases including HER1 and HER2 and therefore be useful in the treatment of proliferative disorders such as psoriasis and cancer. The ability of these compounds to inhibit HER1 and other receptor kinases will permit their use as anti-angiogenic agents to treat disorders such as cancer and diabetic retinopathy.
  • Protein tyrosine kinase-associated disorders are those disorders which result from aberrant tyrosine kinase activity, and/or which are alleviated by the inhibition of one or more of these enzymes.
  • Lck inhibitors are of value in the treatment of a number of such disorders (for example, the treatment of autoimmune diseases), as Lck inhibition blocks T cell activation.
  • the treatment of T cell mediated diseases, including inhibition of T cell activation and proliferation, is a particularly preferred use for compounds of formula (I) prepared according to the process herein.
  • transplant such as organ transplant, acute transplant or heterograft or homograft (such as is employed in burn treatment)
  • protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes
  • transplantation tolerance induction arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; chronic obstructive pulmonary disease (COPD), such as emphysema; inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft vs.
  • transplant such as organ transplant, acute transplant or heterograft or homograft (such as is employed in burn treatment)
  • protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes
  • transplantation tolerance induction arthritis (such as rhe
  • T-cell mediated hypersensitivity diseases including contact hypersensitivity, delayed-type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; cancers, including cancers where Lck or other Src-family kinases such as Src are activated or overexpressed, such as colon carcinoma and thymoma, and cancers where Src-family kinase activity facilitates tumor growth or survival; glomerulonephritis; serum sickness; uticaria
  • the compounds of the present invention are useful for the treatment of cancers such as chronic myelogenous leukemia (CML), gastrointestinal stromal tumor (GIST), small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), ovarian cancer, melanoma, mastocytosis, germ cell tumors, acute myelogenous leukemia (AML), pediatric sarcomas, breast cancer, colorectal cancer, pancreatic cancer, prostate cancer and others known to be associated with protein tyrosine kinases such as, for example, SRC, BCR-ABL and c-KIT.
  • CML chronic myelogenous leukemia
  • GIST gastrointestinal stromal tumor
  • SCLC small cell lung cancer
  • NSCLC non-small cell lung cancer
  • ovarian cancer melanoma
  • mastocytosis germ cell tumors
  • AML acute myelogenous leukemia
  • pediatric sarcomas breast cancer
  • colorectal cancer pancreatic cancer
  • the compounds of the present invention are also useful in the treatment of cancers that are sensitive to and resistant to chemotherapeutic agents that target BCR-ABL and c-KIT, such as, for example, Gleevec® (STI-571).
  • the compound of the formula (IN) (including, but not limited to the crystalline forms of that compound described herein, such as the crystalline monohydrate) is useful in the treatment of patients resistant or intolerant to Gleevec ® (STI-571) for diseases such as chronic myelogenous leukemias (CML), or other cancers (including other leukemias) as described herein.
  • a compound of Formulas I is administered in conjunction with at least one anti-neoplastic agent.
  • anti-neoplastic agent or "anti-cancer agent” is synonymous with "chemotherapeutic agent” and/or “anti-proliferative agent” and refers to compounds that prevent cancer, or hyperproliferative cells from multiplying.
  • Anti-proliferative agents prevent cancer cells from multiplying by: (1) interfering with the cell's ability to replicate D ⁇ A and (2) inducing cell death and/or apoptosis in the cancer cells.
  • Classes of compounds that may be used as anti-proliferative cytotoxic agents and/or anti-proliferative agents include the following: Alkylating agents (including, without limitation, nitrogen mustards, ethyleni ine derivatives, alkyl sulfonates, nitrosoureas and triazenes): Uracil mustard, Chlormethine, Cyclophosphamide (Cytoxan®), ffosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylene-melamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, dacarbazine, and Temozolomide.
  • Alkylating agents including, without limitation, nitrogen mustards, ethyleni ine derivatives, alkyl sulfonates, nitrosoureas and triazenes
  • Uracil mustard including, without limitation, nitrogen mustards, ethyleni ine derivatives, al
  • Antimetabolites including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors: Methotrexate, 5-Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6- Thioguanine, Fludarabine phosphate, Pentostatine, and Gemcitabine.
  • Natural products and their derivatives for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins: Ninblastine, Nincristine,
  • Idarubicin, Ara-C, paclitaxel (paclitaxel is commercially available as Taxol®), Mithramycin, Deoxyco-formycin, Mitomycin-C, L-Asparaginase, H terferons (especially TFN-a), Etoposide, and Teniposide.
  • anti-proliferative cytotoxic agents and/or anti-proliferative agents are navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
  • radiation therapy includes, but is not limited to, x-rays or gamma rays which are delivered from either an externally applied source such as a beam or by implantation of small radioactive sources. Radiation therapy may be useful in combination with compounds of the present invention. The following may also be useful when administered in combination with compounds of the present invention.
  • Microtubule affecting agents interfere with cellular mitosis and are well known in the art for their anti-proliferative cytotoxic activity.
  • Microtubule affecting agents useful in the invention include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolastatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®, NSC 125973), Taxol® derivatives (e.g., derivatives (e.g., NSC 608832), thiocolchicine NSC 361792), trityl cysteine (NSC 83265), vinblastine sulfate (NSC 49842), vincristine sulfate (NSC 67574), natural and synthetic epothilones
  • antineoplastic agents include, discodermolide (see Service, (1996) Science, 274:2009) estramustine, nocodazole, MAP4, and the like. Examples of such agents are also described in the scientific and patent literature, see, e.g., Bulinski (1997) J. Cell Sci. 110:3055 3064; Panda (1997) Proc. Natl. Acad. Sci.
  • hormones and steroids include synthetic analogs: 17a-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, hlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, Zoladex can also be administered to the patient.
  • antiangiogenics such as matrix metalloproteinase inhibitors, and other VEGF inhibitors, such as anti-VEGF antibodies and small molecules such as ZD6474 ; and SU6668 are also included.
  • VEGF inhibitors such as anti-VEGF antibodies and small molecules such as ZD6474 ; and SU6668 are also included.
  • Anti- Her2 antibodies from Genetech may also be utilized.
  • a suitable EGFR inhibitor is EKB-569 (an irreversible inhibitor).
  • Imclone antibody C225 immunospecific for the EGFR, and src inhibitors are also included.
  • cytostatic agent is CasodexTM which renders androgen-dependent carcinomas non-proliferative.
  • cytostatic agent is the antiestrogen Tamoxifen which inhibits the proliferation or growth of estrogen dependent breast cancer.
  • Inhibitors of the transduction of cellular proliferative signals are cytostatic agents. Examples are epidermal growth factor inhibitors, Her-2 inhibitors, MEK-1 kinase inhibitors, MAPK kinase inhibitors, PI3 inhibitors, Src kinase inhibitors, and PDGF inhibitors.
  • certain anti-proliferative agents are anti-angiogenic and antivascular agents and, by interrupting blood flow to solid tumors, render cancer cells quiescent by depriving them of nutrition. Castration, which also renders androgen dependent carcinomas non-proliferative, may also be utilized. Starvation by means other than surgical disruption of blood flow is another example of a cytostatic agent.
  • a particular class of antivascular cytostatic agents is the combretastatins.
  • Other exemplary cytostatic agents include MET kinase inhibitors, MAP kinase inhibitors, inhibitors of non-receptor and receptor tyrosine kinases, inhibitors of integrin signaling, and inhibitors of insulin-like growth factor receptors.
  • anthracyclines e.g., daunorubicin, doxorubicin, cytarabine (ara-C; Cytosar-U®); 6-thioguanine (Tabloid®), mitoxantrone (Novantrone®) and etoposide (NePesid®),amsacrine (AMSA), and all-trans retinoic acid (ATRA).
  • anthracyclines e.g., daunorubicin, doxorubicin
  • ara-C Cytosar-U®
  • 6-thioguanine Tabloid®
  • mitoxantrone Novantrone®
  • NePesid® etoposide
  • AMSA amsacrine
  • ATRA all-trans retinoic acid
  • BCR-ABL inhibitors such as, but not limited to, Gleevec® (imatinib, STI-571) or AM ⁇ -107, the compound shown below
  • the compounds of the present invention may be useful in combination with anti-cancer compounds such as fentanyl, doxorubicin, interferon alfa-n3, palonosetron dolasetron anastrozole, exemestane, bevacizumab, bicalutamide, cisplatin, dacarbazine, cytarabine, clonidine, epirubicin, levamisole, toremifene, fulvestrant, letrozole, tamsulosin, gallium nitrate, trastuzumab, altretamine, hydroxycarbamide, ifosfamide, interferon alfacon-1, gefitinib, granisetron, leuprorelin, dronabinol, megestrol, pethidine, promethazine, morphine, vinorelbine, pegfilgrastim, filgrastim, nilutamide, thiethylperazine,
  • the present invention provides methods for the treatment of a variety of cancers, including, but not limited to, the following: carcinoma including that of the bladder (including accelerated and metastatic bladder cancer), breast, colon (including colorectal cancer), kidney, liver, lung (including small and non-small cell lung cancer and lung adenocarcinoma), ovary, prostate, testes, genitourinary tract, lymphatic system, rectum, larynx, pancreas (including exocrine pancreatic carcinoma), esophagus, stomach, gall bladder, cervix, thyroid, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma, histiocytic lympho
  • the invention is useful to treat GIST, breast cancer, pancreatic cancer, colon cancer, NSCLC, CML, and ALL, sarcoma, and various pediatric cancers.
  • the compounds of the present invention are protein tyrosine kinase inhibitors and as such are useful in the treatment of immunological disorders in addition to oncological disorders.
  • U.S. Patent No. 6,596,746 describes the utility of the compound in immunological disorders and is hereby incorporated by reference for the description of the compound in such immunological disorders.
  • the present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the administration of a therapeutically effective amount of the combinations of this invention, with or without pharmaceutically acceptable carriers or diluents.
  • the pharmaceutical compositions of this invention comprise an anti-proliferative agent or agents, a formula I compound, and a pharmaceutically acceptable carrier.
  • the methods entail the use of a neoplastic agent in combination with a Formula I compound.
  • the compositions of the present invention may further comprise one or more pharmaceutically acceptable additional ingredient(s) such as alum, stabilizers, antimicrobial agents, buffers, coloring agents, flavoring agents, adjuvants, and the like.
  • the antineoplastic agents, Formula I, compounds and compositions of the present invention may be administered orally or parenterally including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration.
  • the present invention also provides using the compounds obtained with the inventive process to further prepare pharmaceutical compositions capable of treating Src-kinase associated conditions, including the conditions described above.
  • the said compositions may contain other therapeutic agents.
  • Pharmaceutical compositions may be formulated by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (e.g., excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulations.
  • compositions may be administered by any means suitable for the condition to be treated, which may depend on the need for site-specific treatment or quantity of drug to be delivered.
  • Topical administration is generally preferred for skin-related diseases, and systematic treatment preferred for cancerous or pre-cancerous conditions, although other modes of delivery are contemplated.
  • the compounds of formula (1) may be delivered orally, such as in the form of tablets, capsules, granules, powders, or liquid formulations including syrups; topically, such as in the form of solutions, suspensions, gels or ointments; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular or intrasternal injection or infusion techniques (e.g., as sterile injectable aq. or non-aq. solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; rectally such as in the form of suppositories; or liposomally.
  • Dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents may be administered.
  • the compounds of formula (I), prepared according to the inventive process may be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved with suitable pharmaceutical compositions or, particularly in the case of extended release, with devices such as subcutaneous implants or osmotic pumps.
  • exemplary compositions for topical administration include a topical carrier such as PLASTIBASE® (mineral oil gelled with polyethylene).
  • compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art.
  • the compounds of formula (I) may also be orally delivered by sublingual and/or buccal administration, e.g., with molded, compressed, or freeze-dried tablets.
  • compositions may include fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins.
  • fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins.
  • high molecular weight excipients such as celluloses (ANICEL®) or polyethylene glycols (PEG); an excipient to aid mucosal adhesion such as hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), sodium carboxymethyl cellulose (SCMC), and/or maleic anhydride copolymer (e.g., GA ⁇ TREZ®); and agents to control release such as polyacrylic copolymer (e.g., CARBOPOL 934®).
  • HPC hydroxypropyl cellulose
  • HPMC hydroxypropyl methyl cellulose
  • SCMC sodium carboxymethyl cellulose
  • Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use.
  • An example of a composition for oral administration is the compound of formula (IN), lactose monohydrate (intra-granular phase), microcrystalline cellulose(intra-granular phase), croscarmellose sodium(intra-granular phase), hydroxypropyl cellulose(intra-granular phase), microcrystalline cellulose (extra- ' granular phase), croscarmellose sodium (extra-granular phase), and magnesium stearate (extragranular phase).
  • compositions for nasal aerosol or inhalation administration include solutions which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance absorption and/or bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
  • compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • suitable non-toxic, parenterally acceptable diluents or solvents such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
  • compositions for rectal administration include suppositories which may contain, for example, suitable non-irritating excipients, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • suitable non-irritating excipients such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
  • the effective amount of a compound of formula (I) may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a mammal of from about 0.05 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition.
  • Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats, horses, and the like.
  • this term is intended to include all subjects, most preferably mammalian species, that are affected by mediation of Src kinase levels.
  • the compounds of the present invention are administered using the formulations of the invention.
  • the compounds of the present invention are administered by IN infusion over a period of from about 10 minutes to about 3 hours, preferably about 30 minutes to about 2 hours, more preferably about 45 minutes to 90 minutes, and most preferably about 1 hour.
  • the compounds are administered intravenously in a dose of from about 0.5 mg/m 2 to 65 mg/m 2 , preferably about 1 mg/m 2 to 50 mg/m 2 , more preferably about 2.5 mg/m 2 to 30 mg/m 2 , and most preferably about 25 mg/m 2 .
  • compounds of the present invention including the crystalline forms of the compounds of formula IN can be administered orally, intravenously, or both.
  • the methods of the invention encompass dosing protocols such as once a day for 2 to 10 days, preferably every 3 to 9 days, more preferably every 4 to 8 days and most preferably every 5 days.
  • the compounds of the present invention, including the crystalline forms of the compounds of formula TV can be administered orally, intravenously, or both, once a day for 3 days, with a period of 1 week to 3 weeks in between cycles where there is no treatment.
  • the compounds of the present invention, the crystalline forms of the compounds of formula TV can be administered orally, intravenously, or both, once a day for 5 days, with a period of 1 week to 3 weeks in between cycles where there is no treatment.
  • the treatment cycle for administration of the compounds of the present invention, the crystalline forms of the compounds of formula IN is once daily for 5 consecutive days and the period between treatment cycles is from 2 to 10 days, or alternatively one week.
  • a compound of the present invention for example, a compound of formula IN, is administered once daily for 5 consecutive days, followed by 2 days when there is no treatment.
  • the compounds of the present invention, the crystalline forms of the compounds of formula TV can also be administered orally, intravenously, or both once every 1 to 10 weeks, every 2 to 8 weeks, every 3 to 6 weeks, alternatively every 3 weeks.
  • the compounds of the present invention, the crystalline forms of the compounds of formula IN are administered in a 28 day
  • the compounds of the present invention are administered in a 28 day cycle wherein the compound of formulae TV are orally administered on day 1 and intravenously administered on days 7, 14, and 28.
  • the compounds of the present invention are administered until the patient shows a response, for example, a reduction in tumor size, or until dose limiting toxicity is reached.
  • Jurkat T cells are incubated with the test compound and then stimulated by the addition of antibody to CD3 (monoclonal antibody G19-4). Cells are lysed after 4 minutes or at another desired time by the addition of a lysis buffer containing ⁇ P-40 detergent. Phosphorylation. of proteins is detected by anti-phosphotyrosine immunoblotting. Detection of phosphorylation of specific proteins of interest such as
  • ZAP-70 is detected by immunoprecipitation with anti-ZAP-70 antibody followed by anti-phosphotyrosine immunoblotting.
  • Such procedures are described in Schieven, G.L., Mittler, R.S., Nadler, S.G., Kirihara, J.M., Bolen, J.B., Kanner, S.B., and
  • the Lck inhibitors inhibit the tyrosine phosphorylation of cellular proteins induced by anti-CD3 antibodies.
  • G19-4 See Hansen, J.A., Martin, P.J., Beatty, P.G.,
  • Lck inhibitors block calcium mobilization in T cells stimulated with anti-CD3 antibodies.
  • Cells are loaded with the calcium indicator dye indo-1, treated with anti- CD3 antibody such as the monoclonal antibody G19-4, and calcium mobilization is measured using flow cytometry by recording changes in the blue/violet indo-1 ratio as described in Schieven, G.L., Mittler, R.S., Nadler, S.G., Kirihara, J.M., Bolen, J.B., Kanner, S.B., and Ledbetter, J.A., "ZAP-70 tyrosine kinase, CD45 and T cell receptor involvement in UV and H 2 O 2 induced T cell signal transduction", /. Biol. Chem., 269, 20718-20726 (1994), and the references incorporated therein.
  • Lck inhibitors inhibit the proliferation of normal human peripheral blood T cells stimulated to grow with anti-CD3 plus anti-CD28 antibodies.
  • a 96 well plate is coated with a monoclonal antibody to CD3 (such as G19-4), the antibody is allowed to bind, and then the plate is washed. The antibody bound to the plate serves to stimulate the cells.
  • Normal human peripheral blood T cells are added to the wells along with test compound plus anti-CD28 antibody to provide co-stimulation. After a desired period of time (e.g., 3 days), the [3H]-thymidine is added to the cells, and after further incubation to allow incorporation of the label into newly synthesized DNA, the cells are harvested and counted in a scintillation counter to measure cell proliferation.
  • Example 3 To a 50 mL RBF containing the above compound 3 A (0.5g, 2.0 mmol) was added THF (2.5 mL) and water (2 mL), followed by NBS (0.40 g, 2.22 mmol), and the solution was stirred for 90 min. R-sec-butylthiourea (Ex. 2) (267 mg), was added, and the solution was heated to 75 °C for 8 h. Cone. NH OH was added to adjust the pH to 10 followed by the addition of EtOH (15 mL). Water (15 mL) was added and the slurry stirred for 16 h, filtered, and washed with water to give Example 3 as a light brown solid (0.48 g, 69% yield, 98% purity). MS 347.1; HPLC 2.59.
  • Example 4 is prepared following the methods of Example 3 but using the appropriate acryl benzamide and Example 1.
  • an X-ray diffraction pattern may be obtained with a measurement error that is dependent upon the measurement conditions employed.
  • intensities in a X-ray diffraction pattern may fluctuate depending upon measurement conditions employed. It should be further understood that relative intensities may also vary depending upon experimental conditions and, accordingly, the exact order of intensity should not be taken into account.
  • a measurement error of diffraction angle for a conventional X-ray diffraction pattern is typically about 5% or less, and such degree of measurement error should be taken into account as pertaining to the aforementioned diffraction angles.
  • crystal forms of the instant invention are not limited to the crystal forms that provide X-ray diffraction patterns completely identical to the X-ray diffraction patterns depicted in the accompanying Figures disclosed herein. Any crystal forms that provide X- ray diffraction patterns substantially identical to those disclosed in the accompanying Figures fall within the scope of the present invention.
  • the ability to ascertain substantial identities of X-ray diffraction patterns is within the purview of one of ordinary skill in the art.
  • the structures were solved by direct methods and refined on the basis of observed reflections using either the SDP (SDP, Structure Determination Package,Enraf- ⁇ onius, Bohemia NY 11716 Scattering factors, including /and f, in the SDP software were taken from the" International Tables for Crystallography", Kynoch Press, Birmingham, England, 1974; Nol TV, Tables 2.2A and 2.3.1) software package with minor local modifications or the crystallographic package, MAXUS (maXus solution and refinement software suite: S. Mackay, C.J. Gilmore, C. Edwards, M. Tremayne, ⁇ . Stewart, K. Shankland. maXus: a computer program for the solution and refinement of crystal structures from diffraction data).
  • SDP Structure Determination Package,Enraf- ⁇ onius, Bohemia NY 11716 Scattering factors, including /and f, in the SDP software were taken from the" International Tables for Crystallography", Kynoch Press, Birmingham, England, 1974; Nol
  • the DSC instrument used to test the crystalline forms was a TA Instruments® model Q1000.
  • the DSC cell/sample chamber was purged with 100 ml/min of ultra- high purity nitrogen gas.
  • the instrument was calibrated with high purity indium.
  • the accuracy of the measured sample temperature with this method is within about +/- 1°C, and the heat of fusion can be measured within a relative error of about +/-5%.
  • the sample was placed into an open aluminum DSC pan and measured against an empty reference pan. At least 2 mg of sample powder was placed into the bottom of the pan and lightly tapped down to ensure good contact with the pan.
  • the weight of the sample was measured accurately and recorded to a hundredth of a milligram.
  • the instrument was programmed to heat at 10°C per minute in the temperature range between 25 and 350°C.
  • the heat flow which was normalized by a sample weight, was plotted versus the measured sample temperature. The data were reported in units of watts/gram ("W/g"). The plot was made with the endothermic peaks pointing down. The endothermic melt peak was evaluated for extrapolated onset temperature, peak temperature, and heat of fusion in this analysis.
  • the TGA instrument used to test the crystalline forms was a TAHistruments® model Q500. Samples of at least 10 milligrams were analyzed at a heating rate of 10°C per minute in the temperature range between 25°C and about 350°C.
  • Polish filter by transfer the compound of formula (TV) solution at 75 °C through the preheated filter and into the receiver.
  • Seed crystals are not essential to obtaining monohydrate, but provide better control of the crystallization.
  • the monohydrate can be obtained by: 1) An aqueous solution of the acetate salt of compound TV was seeded with monohydrate and heated at 80 °C to give bulk monohydrate.
  • the XRPD is also characterized by the following list comprising 2 ⁇ values selected from the group consisting of: 4.6+ 0.2, 11.2 ⁇ 0.2, 13.8 ⁇ 0.2, 15.2 ⁇ 0.2, 17.9 ⁇ 0.2, 19. l ⁇ 0.2, 19.6+ 0.2, 23.2+ 0.2, 23.6 ⁇ 0.2.
  • the XRPD is also characterized by the list of 2 ⁇ values selected from the group consisting of: 18.0 ⁇ 0.2, 18.4 ⁇ 0.2, 19.2 ⁇ 0.2, 19.6 ⁇ 0.2, 21.2 ⁇ 0.2, 24.5+ 0.2, 25.9+ 0.2, and 28.0+ 0.2.
  • Density (calculated) (g/cm 3 ) 1.354
  • Z' number of drug molecules per asymmetric unit.
  • Nm N(unit cell) / (Z drug molecules per cell).
  • Density (calculated) (g/cm 3 ) 1.300 wherein the compound is at a temperature of about -50°C. ,
  • the simulated XRPD was calculated from the refined atomic parameters at room temperature.
  • the monohydrate of the compound of formula (TV) is represented by the DSC as shown in Figure 2.
  • the DSC is characterized by a broad peak between approximately 95°C and 130°C. This peak is broad and variable and corresponds to the loss of one water of hydration as seen in the TGA graph.
  • the DSC also has a characteristic peak at approximately 287°C which corresponds to the melt of the dehydrated form of the compound of formula (IN).
  • the weight loss corresponds to a loss of one water of hydration from the compound of Formula (IN).
  • the monohydrate may also be prepared by crystallizing from alcoholic solvents, such as methanol, ethanol, propanol, i-propanol, butanol, pentanol, and water.
  • alcoholic solvents such as methanol, ethanol, propanol, i-propanol, butanol, pentanol, and water.
  • EXAMPLE 9 Preparation of: crystalline n-butanol solvate ofN-(2-chloro-6-methylphenyl)-2-(6-(4-(3- hydrQxyethyl)piperazin-l-yl)-2-methylpyrimidin-4-ylamino)thiazole-5-carboxamide (IV)
  • the crystalline butanol solvate of the compound of formula (IN) is prepared by dissolving compound (TV) in 1 -butanol at reflux (116-118 °C) at a concentration of approximately lg/25 mL of solvent. Upon cooling, the butanol solvate crystallizes out of solution. Filter, wash with butanol, and dry.
  • the resulting wet cake was returned to the 100-mL reactor, and charged with 56 mL (12 mL/g) of 200 proof ethanol. At 80 °C an additional 25 mL of ethanol was added. To this mixture was added 10 mL of water resulting in rapid dissolution. Heat was removed and crystallization was observed at 75 - 77 °C. The crystal slurry was further cooled to 20 °C and filtered. The wet cake was washed once with 10 mL of 1:1 ethanol : water and once with 10 mL of n-heptane. The wet cake contained 1.0% water by KF and 8.10% volatiles by LOD.
  • ethanol solvate of the compound of formula (IN) may be represented by the XRPD as shown in Figure 4 or by a representative sampling of peaks.
  • Representative peaks for the crystalline ethanol solvate are 2 ⁇ values of : 5.8 ⁇ 0.2, 11.3 ⁇ 0.2, 15.8 ⁇ 0.2, 17.2 ⁇ 0.2, 19.5 ⁇
  • the title neat form may be prepared by heating the monohydrate form of the compound of formula (IV) above the dehydration temperature.
  • T1H1-7 of the compound of formula (IN) may be represented by the XRPD as shown in Figure 6 or by a representative sampling of peaks.
  • Representative peaks for the crystalline neat form (T1H1-7)) are 2 ⁇ values of : 8.0 ⁇ 0.2, 9.7 ⁇ 0.2, 11.2 ⁇ 0.2, 13.3 ⁇ 0.2, 17.5 ⁇ 0.2, 18.9 ⁇ 0.2, 21.0 ⁇ 0.2, 22.0 ⁇ 0.2.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Thiazole And Isothizaole Compounds (AREA)
  • Low-Molecular Organic Synthesis Reactions Using Catalysts (AREA)

Abstract

The invention relates to processes for preparing compounds having the formula (I) and crystalline forms thereof, wherein Ar is aryl or heteroaryl, L is an optional alkylene linker, and R2, R3, R4, and R5, are as defined in the specification herein, which compounds are useful as kinase inhibitors, in particular, inhibitors of protein tyrosine kinase and p38 kinase.

Description

PROCESS FOR PREPARING 2-AMINOTHIAZOLE-5-AROMATIC CARBOXAMIDES AS KINASE INHIBITORS
FIELD OF THE INVENTION The present invention relates to processes for preparing 2-aminothiazole-5- aromatic carboxamides which are useful as kinase inhibitors, such as inhibitors of protein tyrosine kinase and p38 kinase, intermediates and crystalline forms thereof.
BACKGROUND OF THE INVENTION Aminothiazole-aromatic amides of formula I
Figure imgf000003_0001
wherein Ar is aryl or heteroaryl, L is an optional alkylene linker, and R2, R3, Ri, and R5, are as defined in the specification herein, are useful as kinase inhibitors, in particular, inhibitors of protein tyrosine kinase and p38 kinase. They are expected to be useful in the treatment of protein tyrosine kinase-associated disorders such as immunologic and oncological disorders [see, US Pat. No. 6,596,746 (the '746 patent), assigned to the present assignee and incorporated herein by reference], and p38 kinase-associated conditions such as inflammatory and immune conditions, as described in US patent application Serial No. 10/773,790, filed February 6, 2004, claiming priority to US Provisional application Serial No. 60/445,410, filed February 6, 2003 (hereinafter the '410 application), both of which are also assigned to the present assignee and incorporated herein by reference.
The compound of formula (IN), 'Ν-(2-Chloro-6-methylphenyl)-2-[[6-[4-(2- hydroxyethyl)-l-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamide, is an inhibitor of SRC/ABL and is useful in the treatment of oncological diseases.
Figure imgf000004_0001
(IV). Other approaches to preparing 2-aminothiazole-5-carboxamides are described in the '746 patent and in the '410 application. The '746 patent describes a process involving treatment of chlorothiazole with n-BuLi followed by reaction with phenyl isocyanates to give chlorothiazole-benzamides, which are further elaborated to aminothiazole-benzamide final products after protection, chloro-to-amino substitution, and deprotection, e.g.,
Figure imgf000004_0002
Deprot.
The '410 application describes a multi-step process involving first, converting N-unsubstituted aminothiazole carboxylic acid methyl or ethyl esters to bromothiazole carboxylic acid esters via diazotization with tert-butyl nitrite and subsequent CuBr2 treatment, e.g.,
Figure imgf000004_0003
then, hydrolyzing the resulting bromothiazole esters to the corresponding carboxylic acids and converting the acids to the corresponding acyl chlorides, e.g.,
Figure imgf000004_0004
then finally, coupling the acyl chlorides with anilines to afford bromothiazole- benzamide intermediates which were further elaborated to aminothiazole-benzamide final products, e.g.,
Figure imgf000005_0001
Other approaches for making 2-aminothiazole-5-carboxamides include coupling of 2-aminothiazole-5-carboxylic acids with amines using various coupling conditions such as DCC [Roberts et al, J. Med. Chem. (1972), 15, at p. 1310], and DPPA [Marsham et al, J. Med. Chem. (1991), 34, at p. 1594)].
The above methods present drawbacks with respect to the production of side products, the use of expensive coupling reagents, less than desirable yields, and the need for multiple reaction steps to achieve the 2-aminothiazole-5-carboxamide compounds.
Reaction of N,N-dimethyl-N'-(aminothiocarbonyl)-formamidines with α- haloketones and esters to give 5-carbonyl-2-aminothiazoles has been reported. See Lin, Y. et al, J. Heterocycl Chem. (1979), 16, at 1377; Hartmann, H. et al, J. Chem. Soc. Perkin Trans. (2000), 1, at 4316; Noack, A. et al; Tetrahedron (2002), 58, at 2137; Noack, A.; et al. Angew. Chem. (2001), 7 3,'at 3097; and Kantlehner, W. et al, J. Prakt. Chetn./Chem.-Ztg. (1996), 338, at 403. Reaction of β-ethoxy acrylates and thioureas to prepare 2-aminothiazole-5-carboxylates also has been reported. See Zhao, R., et al, Tetrahedron Lett. (2001), 42, at 2101. However, electrophilic bromination of acrylanilide and crotonanilide has been known to undergo both aromatic bromination and addition to the ,β-unsaturated carbon-carbon double bonds. See Autenrieth, Chem. Ber. (1905), 38, at 2550; Eremeev et al, Chem. Heterocycl. Compd. Engl Transl. (1984), 20, at 1102.
New and efficient processes for preparing 2-aminothiazole-5-carboxamides are desired.
SUMMARY OF THE INVENTION
This invention is related to processes for the preparation of 2-aminothiazole-5- aromatic amides having the formula (I),
Figure imgf000006_0001
wherein L, Ar, R , R3, R , R5, and m are as defined below, comprising reacting a compound having the formula (H),
Figure imgf000006_0002
wherein Q is the group -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, and Ar, L, R2, R3, and m are as defined below, with a halogenating reagent in the presencp of water followed by a thiourea compound having the formula (HI),
Figure imgf000006_0003
wherein, 1^ and R5 are as defined below, to provide the compound of formula (I),
Figure imgf000006_0004
wherein,
Ar is the same in formulae (I) and (IT) and is aryl or heteroaryl;
L is the same in formulae (I) and (U) and is optionally-substituted alkylene;
R2 is the same in formulae (I) and (II), and is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo; R3 is the same in formulae (I) and (U), and is selected from hydrogen, halogen, cyano, haloalkyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aryl, heteroaryl, cycloalkyl, and heterocyclo; 1^ is (i) the same in each of formulae (I) and (IH), and (ii) is independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo, or alternatively, R4 is taken together with R5, to form heteroaryl or heterocyclo; R5 is (i) the same in each of formulae (I) and (III), and (ii) is independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo, or alternatively, R5 is taken together with R4, to form heteroaryl or heterocyclo; and m is 0 or 1.
Applicants have surprisingly discovered said process for converting β-(P*)oxy acryl aromatic amides and thioureas to 2-aminothiazole derivatives, wherein the aromatic amides are not subject to further halogenation producing other side products. Aminothiazole-aromatic amides, particularly, 2-aminothiazole-5-benzamides, can thus be efficiently prepared with this process in high yield.
In another aspect, the present invention is directed to crystalline forms of the compound of formula (IV).
BRIEF DESCRIPTION OF THE DRAWINGS The invention is illustrated by reference to the accompanying drawings described below.
Figure 1 shows a simulated (bottom) (calculated from atomic coordinates generated at room temperature) and experimental (top) pXRD patterns for crystalline monohydrate of the compound of formula (IV). Figure 2 shows a DSC and TGA of the of the monohydrate crystalline form of the compound of Formula (IV). Figure 3 shows a simulated (bottom) (from atomic parameters refined at room temperature) and experimental (top) pXRD patterns for crystalline butanol solvate of the compound of formula (IN).
Figure 4 shows a simulated (bottom) (from atomic parameters refined at - 40°C) and experimental (top) pXRD patterns for crystalline ethanol solvate of the compound of formula (IN).
Figure 5 shows a simulated (bottom) (from atomic parameters refined at room temperature) and experimental (top) pXRD patterns for crystalline neat form (Ν-6) of the compound of formula (IN). Figure 6 shows a simulated (bottom) (from atomic parameters refined at room temperature) and experimental (top) pXRD patterns for crystalline neat form (T1H1-7) of the compound of formula (IN).
DETAILED DESCRIPTION OF THE INVENTION
Abbreviations
For ease of reference, the following abbreviations may be used herein:
Ph = phenyl
Bz = benzyl t-Bu = tertiary butyl
Me = methyl
Et = ethyl
Pr = propyl
Iso-P = isopropyl MeOH = methanol
EtOH = ethanol
EtOAc = ethyl acetate
Boc = tert-butyloxycarbonyl
CBZ = carbobenzyloxy or carbobenzoxy or benzyloxycarbonyl DMF = dimethyl formamide
DMF-DMA = N,N-dimethylformamide dimethyl acetal
DMSO = dimethyl sulf oxide
DPPA = diphenylphosphoryl azide
DPPF = l,l'-bis(diphenylphosphino)ferrocene HATU = O-benzotriazol-1-ylO Ν,Ν,Ν\Ν'-tetramethyluronium hexafluorphosphate
LDA = lithium di-isopropyl amide
TEA = triethylamine
TFA = trifluoroacetic acid
THF = tetrahydrofuran KOH = potassium hydroxide
K CO = potassium carbonate
POCl3 =phosphorous oxychloride
EDC or EDCI = 3-ethyl-3'-(dimethylamino)propyl- carbodiimide DIPEA = diisopropylethylamine
HOBt= 1-hydroxybenzotriazole hydrate
NBS = N-bromosuccinamide
NMP = N-methyl-2-pyrrolidinone
NaH = sodium hydride NaOH = sodium hydroxide '
Na2S2O3 = sodium thiosulfate
Pd = palladium
Pd-C or Pd/C = palladium on carbon min = minute(s) L = liter mL = milliliter μL = microliter g = gram(s) mg = milligram(s) mol = moles mmol = millimole(s) meq = milliequivalent
RT or rt = room temperature
RBF = round bottom flask ret. t. = HPLC retention time (minutes) sat or sat'd = saturated aq. = aqueous
TLC = thin layer chromatography
HPLC = high performance liquid chromatography LC/MS = high performance liquid chromatography/mass spectrometry
MS = mass spectrometry
NMR = nuclear magnetic resonance mp = melting point
DSC = differential scanning calorimetry TGA = thermogravimetric analysis
XRPD = x-ray powder diffraction pattern pXRD = x-ray powder diffraction pattern
Definitions The following are definitions of terms used in this specification and appended claims. The initial definition provided for a group or term herein applies to that group or term throughout the specification and claims, individually or as part of another group, unless otherwise indicated. The term "alkyl" as used herein by itself or as part of another group refers to straight and branched chain saturated hydrocarbons, containing 1 to 20 carbons, 1 to
10 carbons, or 1 to 8 carbons, such as methyl, ethyl, propyl, isopropyl, butyl, t-butyl, isobutyl, pentyl, hexyl, isohexyl, heptyl, 4,4-dimethylpentyl, octyl, 2,2,4-trimethyl- pentyl, nonyl, decyl, undecyl, dodecyl, the various branched chain isomers thereof, and the like. Lower alkyl groups, that is, alkyl groups of 1 to 4 carbon atoms.
The term "substituted alkyl" refers to an alkyl group substituted with one or more substituents (for example 1 to 4 substituents, or 1 to 2 substituents) at any available point of attachment. Exemplary substituents may be selected from one or more (or 1 to 3) of the following groups:
(i) halogen (e.g., a single halo substituent or multiple halo substitutents forming, in the latter case, groups such as a perfluoroalkyl group or an alkyl group bearing Cl3 or CF3), haloalkoxy, cyano, nitro, oxo (=O), -ORa, -SRa, -S(=O)Re, -
S(=O)2Re, -S(=O)3H, -P(=O)2-Re, -S(=O)2ORe, -P(=O)2ORe, -U!-NRbRc, -U N(Rd)-U2-NRbRc, -Uι-NRd-U2-Rb, -NRbP(=O)2Re, -P(=O)2NRbRc, -C(=O)ORe, -
C(=O)Ra, -OC(=O)Ra, -NRdP(=O)2NRbRCj -RbP(=O)2Re, -Uj-aryl, -U heteroaryl,
-Ui-cycloalkyl, -Ui-heterocyclo, -Uι-arylene-Re, -Uι-heteroarylene-Re, -Ui- cycloalkylene-Re, and/or -Ut-heterocyclene-Re, wherein, in group (i), (ii) -Ui- and -U2- are each independently a single bond, -U3-S(O)t-U4-, -
U3-C(O)-U4-, -U3-C(S)-U4-, -U3-O-U4-, -tf-S-U4-, -U3-O-C(O)-U4-, -U3-C(O)-
O-U4-, or -U3-C(=NRg)-U4-; wherein,
(iii) U3 and U4 are each independently a single bond, alkylene, alkenylene, or alkynylene; wherein, in group (i),
(iv) Ra, Rb , Rc Rd, and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclo, or heteroaryl, each of which is unsubstituted or substituted with one to four groups Rf, except Re is not hydrogen; or Rb and Rc may be taken together to form a 3- to 8-membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one to four groups listed below for Rfjor Rb and Rc together with the nitrogen atom to which they are attached may combine to form a group -N=C
RgRh where Rg and Rh are each independently hydrogen, alkyl, or alkyl substituted with a group Rf ; and; ' wherein, (v) Rf is at each occurrence independently selected from alkyl, halogen, cyano, hydroxy, -O(alkyl), SH, -S(alkyl), amino, alkylamino, haloalkyl, haloalkoxy, or a lower alkyl substituted with one to two of halogen, cyano, hydroxy, -O(alkyl), SH, -S(alkyl), amino, alkylamino, haloalkyl, and/or haloalkoxy, and wherein,
(vi) t is θ, 1 or 2.
The term "alkenyl" as used herein by itself or as part of another group refers to straight or branched chain radicals of 2 to 20 carbons, alternatively 2 to 12 carbons, and/or 1 to 8 carbons in the normal chain, which include one to six double bonds in the normal chain, such as vinyl, 2-propenyl, 3-butenyl, 2-butenyl, 4-pentenyl, 3- pentenyl, 2-hexenyl, 3-hexenyl, 2-heptenyl, 3-heptenyl, 4-heptenyl, 3-octenyl, 3- nonenyl, 4-decenyl, 3-undecenyl, 4-dodecenyl, 4,8,12-tetradecatrienyl, and the like. A substituted alkenyl refers to an alkenyl having one or more substituents (for example 1 to 3 substituents, or 1 to 2 substituents), selected from those defined above for substituted alkyl.
The term "alkynyl" as used herein by itself or as part of another group refers to straight or branched chain hydrocarbon groups having 2 to 12 carbon atoms, alternatively 2 to 4 carbon atoms, and at least one triple carbon to carbon bond, such as ethynyl, 2-propynyl, 3-butynyl, 2-butynyl, 4-pentynyl, 3-pentynyl, 2-hexynyl, 3- hexynyl, 2-heptynyl, 3-heptynyl, 4-heptynyl, 3-octynyl, 3-nonynyl, 4-decynyl,3- undecynyl, 4-dodecynyl and the like. A substituted alkynyl refers to an alkynyl having one or more substituents (for example 1 to 4 substituents, or 1 to 2 substituents), selected from those defined above for substituted alkyl.
When the term "alkyl" is used as a suffix with another group, such as in (aryl)alkyl or arylalkyl, this conjunction is meant to refer to a substituted alkyl group wherein at least one of the substituents is the specifically named group in the conjunction. For example, (aryl)alkyl refers to a substituted alkyl group as defined above wherein at least one of the alkyl substituents is an aryl, such as benzyl. However, in groups designated -O(alkyl) and -S(alkyl), it should be understood that the points of attachment in these instances are to the oxygen and sulfur atoms, respectively. Where alkyl groups as defined are divalent, i.e., with two single bonds for attachment to two other groups, they are termed "alkylene" groups. Similarly, where alkenyl groups as defined above and alkynyl groups as defined above, respectively, are divalent radicals having single bonds for attachment to two other groups, they are termed "alkenylene groups" and "alkynylene groups" respectively. Examples of alkylene, alkenylene and alkynylen'e groups include:
CH= CH CH2 ? — CH2CH= CH; ? — C=^=C CH2-
CH3 I CH2 — CH2C = CCH2 — , — -C= CH — CH2 — >
(CH2 ) 29 (CH2)3 f (CH2)4-
>
CH3
— (CH2)2 C CH2CH29 CH2CH— ? ^— CΉ2CΉCΉ — ?
1 1 CH3 ci-3 C2HS
— CH2 — CH — CH — CH2 , — CH2 — CH— CH2 CH ,
CH3 CH3 CH3 CH3
CH3
CH CH2CH ( an(i the like Alkylene groups may be optionally independently substituted as valence allows with one or more groups as defined for substituted alkyl groups. Thus, for example, a substituted alkylene group would
OCH3 f
I (CH2)3-C include CH CH2CH2 anc\ F J and so forth.
The term "cycloalkyl" as used herein by itself or as part of another group refers to optionally-substituted saturated and partially unsaturated (containing 1 or 2 double bonds) cyclic hydrocarbon groups containing 1 to 3 rings, including monocyclicalkyl, bicyclicalkyl and tricyclicalkyl, containing a total of 3 to 20 carbons forming the rings, or 3 to 7 carbons, forming the ring. The further rings of multi-ring cycloalkyls may be either fused, bridged and/or joined through one or more spiro unions. Exemplary cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, cyclododecyl, cyclopentenyl, cycloheptenyl, cyclooctenyl, cyclohexadienyl, cycloheptadienyl,
Figure imgf000013_0001
Each reference to a cycloalkyl is intended to include both substituted and unsubstituted cycloalkyl groups as defined immediately below, unless reference is made to a particular selection of substituents to be made for the cycloalkyl (e.g., wherein cycloalkyl is substituted with one or more groups Rf.) When no particular selection is recited, the optional substituents for the cycloalkyl groups may be selected from the following:
(i) halogen (e.g., a single halo substituent or multiple halo substitutents forming, in the latter case, groups such as a perfluoroalkyl group or an alkyl group bearing Cl3 or CF3), haloalkoxy, cyano, nitro, oxo (=O), -ORa, -SRa, -S(=O)Re, -S(=O)2Re, -S(=O)3H, -P(=O)2-Re, -S(=O)2ORe, -P(=O)2ORe, -U!-NRbRc, -Ui- N(Rd)-U2-NRbRc, -Uι-NRd-U2-Rb, -NRbP(=O)2Re, -P(=O)2NRbRc, -C(=O)ORe, -C(=O)Ra, -OC(=O)Ra, -NRdP(=O)2NRbRc, -RbP(=O)2Re, and/or -U Re, and/or (ii) -Ui-alkyl, -Ui-alkenyl, or -Ui-alkynyl wherein the alkyl, alkenyl, and alkynyl are substituted with one or more (or 1 to 3) groups recited in (i), wherein, in groups (i) and (ii), (iii) -Uι- and -U2- are each independently a single bond, -U3-S(O)t-U4-, - U3-C(O)-U4-, -U3-C(S)-U4-, -U3-O-U4-, -U3-S-U4-, -U3-O-C(O)-U -,-U3-C(O)- O-U4-, or-U3-C(=NRg)-U4-; wherein, in group (iii), (iv) U3 and U4 are each independently a single bond, alkylene, alkenylene, or alkynylene; wherein,
(v) Ra, R , Rc Rd, and Re are each independently hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heterocyclo, or heteroaryl, each of which is unsubstituted or substituted with one or more groups Rf, except R» is not hydrogen; or Rb and Rc may be taken together to form a 3- to 8-membered saturated or unsaturated ring together with the atoms to which they are attached, which ring is unsubstituted or substituted with one or more groups listed below for Rf, or Rb and Rc together with the nitrogen atom to which they are attached may combine to form a group -N=C RgRh, where Rg and Rh are each independently hydrogen, alkyl, or alkyl substituted with a group Rf ; and; wherein,
(vi) Rf is at each occurrence independently selected from alkyl, halogen, cyano, hydroxy, -O(alkyl), SH, -S (alkyl), amino, alkylamino, haloalkyl, haloalkoxy, or a lower alkyl substituted with one to two of halogen, cyano, hydroxy, -O(alkyl), SH, -S(alkyl), amino, alkylamino, haloalkyl, and/or haloalkoxy, and wherein,
(vii) t is 0, 1 or 2.
When the suffix "ene" is used in conjunction with a cyclic group, this is intended to mean the cyclic group as defined herein having two single bonds as points of attachment to other groups. Thus, for example, the term "cycloalkylene" as employed herein refers to a "cycloalkyl" group as defined above which is a linking group such as
Figure imgf000015_0001
and the like.
The term "alkoxy" refers to an alkyl or substituted alkyl group as defined above bonded through an oxygen atom (-O-), i.e., the group -ORi5 wherein R; is alkyl or substituted alkyl.
The term "alkylthio" refers to an alkyl or substituted alkyl group as defined above bonded through a sulfur atom (-S-), i.e., the group -SRi5 wherein R4 is alkyl or substituted alkyl.
The term "acyl" refers to a carbonyl group linked to a radical such as, but not limited to, alkyl, alkenyl, alkynyl, aryl, carbocyclyl, heterocyclyl, more particularly, the group C(=O)Rj, wherein Rj can be selected from alkyl, alkenyl, substituted alkyl, or substituted alkenyl, as defined herein.
O
II
The term "alkoxycarbonyl" refers to a carboxy group ( C— O ) linked to an alkyl radical (i.e., to form CO2Rj), wherein Rj is as defined above for acyl. When the
O II designation "CO2" is used herein, this is intended to refer to the group C— O . The term "alkylamino" refers to amino groups wherein one or both of the hydrogen atoms is replaced with an alkyl group, i.e., NRkRi, wherein one of R and Ri is hydrogen and the other is alkyl, or both Rk and Rj are alkyl.
The term "halo" or "halogen" refers to chloro, bromo, fluoro and iodo. The term "haloalkyl" means a substituted alkyl having one or more halo substituents. For example, "haloalkyl" includes mono, bi, and trifluoromethyl.
The term "haloalkoxy" means an alkoxy group having one or more halo substituents. For example, "haloalkoxy" includes OCF3.
The terms "ar" or "aryl" as used herein by itself or as part of another group refer to optionally-substituted aromatic homocyclic (i.e., hydrocarbon) monocyclic, bicyclic or tricyclic aromatic groups containing 6 to 14 carbons in the ring portion [such as phenyl, biphenyl, naphthyl (including 1-naphthyl and 2-naphthyl) and antracenyl], and may optionally include one to three additional rings (either cycloalkyl, heterocyclo or heteroaryl) fused thereto. Examples include:
Figure imgf000016_0001
Figure imgf000016_0002
Figure imgf000016_0003
Figure imgf000016_0004
and the like.
Each reference to an aryl is intended to include both substituted and unsubstituted aryl groups as defined herein, unless reference is made to a particular selection of substituents to be made for the aryl (e.g., as when aryl is substituted with one or more groups Rf, above). When no particular selection is recited, the optional substituents for the aryl groups may be selected from those recited above, as valence allows, for cycloalkyl groups. The term "heteroaryl" as used herein by itself or as part of another group refers to optionally-substituted monocyclic and bicyclic aromatic rings containing from 5 to 10 atoms, which includes 1 to 4 hetero atoms such as nitrogen, oxygen or sulfur, and such rings fused to an aryl, cycloalkyl, heteroaryl or heterocyclo ring, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. Examples of heteroaryl groups include pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, indolyl, benzothiazolyl, benzodioxolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuranyl, chromonyl, coumarinyl, benzopyranyl, cinnolinyl, quinoxalinyl, indazolyl, pyrrolopyridyl, furopyridyl, dihydroisoindolyl, tetrahydroquinolinyl, carbazolyl, benzidolyl, phenanthrollinyl, acridinyl, phenanthridinyl, xanthenyl
Figure imgf000017_0001
and the like.
Each reference to a heteroaryl is intended to include both substituted and unsubstituted heteroaryl groups as defined herein, unless reference is made to a particular selection of substituents to be made for the heteroaryl (e.g., as when heteroaryl is substituted with one or more groups Rf, above). When no particular selection is recited, the optional substituents for the heteroaryl groups may be selected from those recited above, as valence allows, for cycloalkyl groups.
The terms "heterocyclic" or "heterocyclo" as used herein by itself or as part of another group refer to non-aromatic, optionally substituted, fully saturated or partially unsaturated cyclic groups (for example, 3 to 13 member monocyclic, 7 to 17 member bicyclic, or 10 to 20 member tricyclic ring systems, or containing a total of 3 to 10 ring atoms) which have at least one heteroatom in at least one carbon atom-containing ring. Each ring of the heterocyclic group containing a heteroatom may have 1, 2, 3 or 4 heteroatoms selected from nitrogen atoms, oxygen atoms and/or sulfur atoms, where the nitrogen and sulfur heteroatoms may optionally be oxidized and the nitrogen heteroatoms may optionally be quaternized. The heterocyclic group may be attached at any heteroatom or carbon atom of the ring or ring system, where valence allows. The rings of multi-ring heterocycles may be fused, bridged and/or joined through one or more spiro unions.
Exemplary heterocyclic groups include oxetanyl, imidazolinyl , oxazolidinyl, isoxazolinyl, thiazolidinyl, isothiazolidinyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, -2-oxopiperidinyl, 2-oxopyrrolodinyl, 2-oxoazepinyl, azepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiamorpholinyl, thiamorpholinyl sulfoxide, thiamorpholinyl sulfone, 1,3-dioxolane and tetrahydro-l,l-dioxothienyl,
Figure imgf000018_0001
and the like, which optionally may be substituted. Each reference to a heterocyclo is intended to include both substituted and unsubstituted heterocyclo groups as defined herein, unless reference is made to a particular selection of substituents to be made for the heterocyclo (e.g., as when heterocyclo is substituted with one or more groups Rf, above). When no particular selection is recited, the optional substituents for the heterocyclo groups may be selected from those recited above, as valence allows, for cycloalkyl groups. The term "ring" encompasses homocyclic (i.e., as used herein, all the ring atoms are carbon) or "heterocyclic" (i.e., as used herein, the ring atoms include carbon and one to four heteroatoms selected from N, O and /or S, also referred to as heterocyclo), where, as used herein, each of which (homocyclic or heterocyclic) may be saturated or partially or completely unsaturated.
Unless otherwise indicated, when reference is made to a specifically-named aryl (e.g., phenyl), cycloalkyl (e.g., cyclohexyl), heterocyclo (e.g., pyrrolidinyl) or heteroaryl (e.g., imidazolyl), unless otherwise specifically indicated, the reference is intended to include rings having 0 to 3, or 0 to 2, substituents selected from those recited above for the aryl, cycloalkyl, heterocyclo and/or heteroaryl groups, as appropriate.
The term "heteroatoms" shall include oxygen, sulfur and nitrogen. The term "carbocyclic" means a saturated or unsaturated monocyclic or bicyclic ring in which all atoms of all rings are carbon. Thus, the term includes cycloalkyl and aryl rings. The carbocyclic ring may be substituted in which case the substituents are selected from those recited above for cycloalkyl and aryl groups.
When the term "unsaturated" is used herein to refer to a ring or group, unless otherwise specified, the ring or group may be fully unsaturated or partially unsaturated. "Base" when used herein includes metal oxides, hydroxides or alkoxides, hydrides, or compounds such as ammonia, that accept protons in water or solvent. Thus, exemplary bases include, but are not limited to, alkali metal hydroxides and alkoxides (i.e., MOR, wherein M is an alkali metal such as potassium, lithium, or sodium, and R is hydrogen or alkyl, as defined above, or where R is straight or branched chain -5 alkyl, thus including, without limitation, potassium hydroxide, potassium tert-butoxide, potassium tert-pentoxide, sodium hydroxide, sodium tert- butoxide, lithium hydroxide, etc.); other hydroxides such as magnesium hydroxide (Mg(OH)2) or calcium hydroxide (Ca(OH)2); alkali metal hydrides (i.e., MH, wherein M is as defined above, thus including, without limitation, sodium hydride and lithium hydride); alkylated disilazides, such as, for example, potassium hexamethyldisilazide and lithium hexamethyldisilazide; carbonates such as potassium carbonate (K2CO3), sodium carbonate (Na2CO3), potassium bicarbonate (KHCO3), and sodium bicarbonate (NaHCO3), alkyl ammonium hydroxides such as n-tetrabutyl ammonium hydroxide (TBAH);and so forth. The term "coupling reagent" as used herein refers to a reagent used to couple a carboxylic acid and an amine or an aniline to form an amide bond. It may include a coupling additive, such as CDI, HOBt, HOAt, HODhbt, HOSu, or NEPIS, used in combination with another coupling reagent to speed up coupling process and inhibit side reactions. Particular peptide-coupling reagents may include CDI, DCC, EDC, BBC, BDMP, BOMI, HATU, HAPyU, HBTU, TAPipU, AOP, BDP, BOP, PyAOP, PyBOP, TDBTU, TNTU, TPTU, TSTU, BEMT, BOP-C1, BroP, BTFFH, CIP, EDPBT, Dpp-Cl, EEDQ, FDPP, HOTT-PF6, TOTT-BF4, PyBrop, PyClop, and TFFH. See "Peptide Coupling Reagents: Names, Acronyms and References," Albany Molecular Research, Inc., Technical Reports, Vol. 4, No. 1, incorporated herein by reference.
The terms "halogenating agent" or "halogenating reagent" mean an agent or agents capable of halogenating compounds of formula (H) herein. Halogenating reagents include inorganic and organic halogenating reagents. Examples of inorganic halogenating reagents include chlorine, bromine, iodine, fluorine, and sodium hypochlorite. Organic halogenting reagents include N-chlorosuccinimide (NCS), N- bromosuccinimide (NBS), N-iodosuccinimide (NIS), l,3-dichloro-5,5- dimethylhydantoin, l,3-dibromo-5,5-dimethylhydantoin, and l,3-diiodo-5,5- dimethylhydantoin.
"High yield" as used herein means a yield of greater than 80%, greater than 85%, than 90%, or than 95%.
"Leaving group" means groups having the capability of being displaced upon reaction with a nucleophile including I, Br, CI, R10SO O- (wherein R10 is alkyl, substituted alkyl, aryl, or heteroaryl, as defined herein), and weak bases, such as, for example, HSO4-. Examples of leaving groups include I, Br, CI, and ions of methyl sulfate, mesylate (methane sulfonate), trifluromethanesulfonate, and tosylate (p- toluenesulf onate) .
In compounds of formula (IT) herein, the group Q is -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, i.e., Q has the capability of being displaced upon reaction with a nucleophile. Accordingly, the group P* may be selected from alkyl, -SO2O Rio, -SO2Rιo, -C(=O)Rn and -Si(R12 )3, wherein R10 is defined as above in the definition of "leaving group," Rn is alkyl, aryl or heteroaryl, and Rι2is selected from alkyl and aryl.
"Suitable solvent" as used herein is intended to refer to a single solvent as well as mixtures of solvents. Solvents may be selected, as appropriate for a given reaction step, from, for example, aprotic polar solvents such as DMF, DMA, DMSO, dimethylpropyleneurea, N-methylpyrrolidone (NMP), and hexamethylphosphoric triamide; ether solvents such as diethyl ether, THF, 1,4-dioxane, methyl t-butyl ether, dimethoxymethane, and ethylene glycol dimethyl ether; alcohol solvents such as MeOH, EtOH, and isopropanol; and halogen-containing solvents such as methylene chloride, chloroform, carbon tetrachloride, and 1,2-dichloroethane. Mixtures of solvents may also include biphasic mixtures.
The term "slurry" as used herein is intended to mean a saturated solution of the compound of Formula (IV) and an additional amount of the compound of Formula (IV) to give a heterogeneous solution of the compound of Formula (IV) and a solvent. The present invention describes crystalline forms of the compound of formula (IN) in substantially pure form. As used herein, "substantially pure" means a compound having a purity greater than 90 percent, including 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, and 100 percent. As one example, a crystalline form of the compound of the formula (IN) can be substantially pure in having a purity greater than 90 percent, where the remaining less than 10 percent of material comprises other form(s) of the compound of the formula (IV), and/or reaction and/or processing impurities arising from its preparation. A crystalline form of the compound of the formula (IV) in substantially pure form may therefore be employed in pharmaceutical compositions to which other desired components are added, for example, excipients, carriers, or active chemical entities of different molecular structure.
When dissolved, crystalline forms of the compound of formula (IN) loses its crystalline structure, and is therefore referred to as a solution of the compound of formula (IN). All forms of the present invention, however, may be used for the preparation of liquid formulations in which the drug is dissolved or suspended. In addition, the crystalline forms of the compound of formula (IV) may be incorporated into solid formulations.
A therapeutically effective'amount of the crystalline forms of the compound of formula (IN) is combined with a pharmaceutically acceptable carrier to produce the pharmaceutical compositions of this invention. By "therapeutically effective amount" it is meant an amount that, when administered alone or an amount when administered with an additional therapeutic agent, is effective to prevent, suppress or ameliorate the disease or condition or the progression of the disease or condition.
GENERAL METHODS
This invention is related to a process for the preparation of 2-aminothiazolyl - 5-aromatic amides which are useful as inhibitors of kinases, particularly protein tyrosine kinase and p38 kinase. The process involves halogenation of β-(P*)oxy-α,β- unsaturated carboxyl aromatic amides (II) (wherein P* is as defined herein), such as β-(alkyl)oxy-α,β-unsaturated carboxyl benzamides, and reaction with thioureas (Iff) to give 2-aminothiazole-5-aromatic amides of formula (I). Desired substituents on the 2- amino group and/or the 5-aromatic group can be attached either before or after the aminothiozole formation. For example, in one embodiment, the compound of formula (I) is prepared via reaction of a thiourea wherein I is hydrogen, and the R4 hydrogen atom is then elaborated to more functionalized groups such as, in one embodiment, substituted pyrimidines. In another embodiment, the compound of formula (I) is prepared via reaction of a thiourea wherein R4 is a pyrimidinyl, and the pyrimidinyl optionally is further elaborated with additional substituents, as desired. The process provides an efficient route for preparing 2-aminothiazolyl -5- aromatic amides, essentially in one step and in high yield, without use of expensive coupling reagents or catalysts. Surprisingly, with this process halogenation followed by reaction with thiourea to form the aminothizole is achieved without an undesired aromatic halogenation. One embodiment of the invention is represented in Scheme 1. Scheme 1
Figure imgf000023_0001
- In Scheme 1, Ar is aryl or heteroaryl, more preferably aryl, even more preferably optionally-substituted phenyl. Most preferred is the process involving compounds wherein Ar is phenyl substituted with one to three of alkyl, halogen, -C(=O)NR8, and or NR8C(=O), wherein R8 is alkyl, cycloalkyl, or heteroaryl, more preferably wherein R8 is cyclopropyl or methyl, and even more preferably wherein Ar is selected from 2-chloro-6-methylphenyl, N-cyclopropyl-1 -methyl -benzamide, and N, 1-dimethyl-benzamide. The inventive process may be carried out where a linker group L is present, as in formula I, but advantageously the Ar group is directly attached to the carboxylamide nitrogen atom, as in formula (la).
As noted, the desired substituents may be attached to the group Ar either before or after the halogenation and cyclization process. Likewise, the thiourea compounds (IH) may be prepared, prior to the cyclization, having desired groups R and R5, corresponding to the groups on the desired final product, or alternatively, the desired groups may be attached to the amino-thiazolyl after cyclization. For example, thiourea compounds (Iff) may be prepared and used in the reaction wherein and R5 are both hydrogen, or R4 and R5 are other groups, different from those of the final desired product, and then, after formation of the aminothiazole (I) or (la), the groups R4 and R5 are elaborated to the substituents of the final desired product. All such alternative embodiments and variations thereof are contemplated as within the scope of the present invention.
In intermediates of formula (IT) and (Ha), herein, preferably the group P* may be selected from alkyl, -SO2O R10, -SO20, -C(=O)Rn and -Si(Rι2 )3, as defined above, but preferably P* is an alkyl, more preferably a lower alkyl, i.e., methyl, ethyl, n-propyl, isoP, or a straight or branched butyl. Preferably the group R is hydrogen or lower alkyl, more preferably hydrogen, and R3 is preferably hydrogen. For compounds (ff), β-alkyloxy- ,β-unsaturated carboxyl benzamides are thus preferred, including β-substituted and β-unsubstituted β-alkyloxy- ,β-unsaturated carboxyl benzamides, with the latter more preferred, wherein the phenyl group of the benzamide is optionally substituted as recited above for Ar in formula (la). Also preferred β-unsubstituted β-alkyloxy-oc,β-unsaturated carboxyl benzamides are β- ethoxy aciyl benzamides, again, wherein the phenyl group of the benzamide is optionally substituted as recited above for Ar. Intermediates (II) and (ffa) can be prepared upon reaction of the corresponding anilines, NHR2-Ar, with alkoxyacryloyl compounds. Methods for making β-ethoxy acryl benzamides are also described, for example, in Ashwell, M. A. et al, J. Bioorg. Med. Chem. Lett. (2001), 24, at 3123; and Yoshizaki, S., et al. Chem. Pharm. Bull. (1980), 28, at 3441, incorporated herein by reference.
The halogenating agent(s) used in the process may be any agent or agents as defined herein capable of halogenating compounds (ff), as previously defined herein. Preferred agents include NBS and the N-halohydantoins. Thiourea compounds (Iff) include unsubstituted thioureas, N-monosubstituted thioureas, and N,N-disubstituted thioureas. The steps of halogenation and cyclization are carried out in a suitable solvent which may include one or more solvents such as hydrocarbons, ethers, esters, amides and ketones with ethers, with dioxane preferred.
Another embodiment of the invention is illustrated in Scheme 2. Scheme 2
Figure imgf000025_0001
Figure imgf000025_0002
As can be seen, in Scheme 2, the β-(P*)oxy-acryl benzamides (lib), wherein R2 and R3 are hydrogen, and P* is as previously defined herein, preferably a lower alkyl, are halogenated with a halogenating agent, such as NBS, in a suitable solvent, in the presence of water, then cyclized with unsubstituted thiourea (Ilia). The resulting 2-(unsubstituted)amino-thiazole-5-aromatic amide (lb) is reacted with a pyrimidine compound 4, wherein R and R' are hydrogen or optional substituents, more preferably hydrogen or lower alkyl, and X and Y are both leaving groups, as defined herein, to produce compounds lc. Leaving groups X and Y are preferably I, Br, CI, or RιoSO2O- (wherein Rio is alkyl, substituted alkyl, aryl, or heteroaryl, as defined herein), more preferably X and Y are selected from I, Br, CI, methyl sulfate, mesylate, trifluoromethanesulfonate, and tosylate, even more preferably from CI and Br. Thus, pyrimidines 4 include bis-halogen and sulfonyloxy substituted pyrimidines with the former such as bis-chloro substituted pyrimidines preferred. Advantageously, this step is carried out in the presence of a base, wherein the bases may include alkali hydride and alkoxides with the latter such as sodium t-butoxide preferred. Suitable solvent(s) include solvents such as hydrocarbons, ethers, esters, amides, ketones and alcohols, or mixtures of the above solvents, with ether such as THF preferred.
Compound (lc) can then be reacted with amine NHR20R2ι (5), to provide compounds of formula (Id). For example, R2o and R2ι can both be hydrogen, or R20 and R2ι can be independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R20 and R2i can be taken together to form a heterocyclo. Preferably, R2o and R2ι are taken together so that NHR20R ι forms an optionally-substituted piperazine, more preferably a piperazine N'- substituted with substituted alkyl, more preferably hydroxyethyl. Advantageously, this step is carried out in the presence of a base, including inorganic and organic bases, with organic bases such as tertiary amines preferred. Suitable solvent(s) include solvents such as hydrocarbons, halogenated hydrocarbons, ethers, esters, amides, ketones, lactams and alcohols, and mixtures of the above solvents, with alcohols such as n-butanol as one nonlimiting example, and DMF (dimethylformamide), DMA (dimethylacetamide) and NMP (N-methylpyrrolidine) as other examples. The compounds of formula (Id) thus formed may optionally be further elaborated as desired and/or purified and crystallized.
An alternative approach is illustrated in Scheme 3, wherein a mono-substituted thiourea compound (Iffb) is used.
Scheme 3
Figure imgf000026_0001
As can be seen, in Scheme 3, the β-(P*)oxy-acryl benzamides (lib), as in Scheme 2, are halogenated with a halogenating agent, then further reacted with a monosubstituted thiourea (Bib) having attached thereto a functional pyrimidine group, wherein R, R' and Y are as in Scheme 2, to provide intermediate 2-substituted- aminothiazole-aromatic amides of formula (lc). The compounds of formula (lc) may optionally then be reacted with amines NHR2oR2ι (5), to provide compounds of formula (Id), and/or optionally further elaborated as desired, and/or purified and crystallized.
FURTHER EMBODIMENTS
In one embodiment, the process comprises preparing a compound of the formula (le),
Figure imgf000027_0001
wherein Zi and Z5 are selected from hydrogen, alkyl, halogen, hydroxy, and alkoxy;
Z2, Z3 and Z4 are selected from hydrogen, alkyl, halogen, hydroxy, alkoxy, C(=O)NR8, and/or NR8C(=O), wherein R8 is alkyl, cycloalkyl, or heteroaryl; comprising reacting a compound having the formula,
Figure imgf000027_0002
wherein Q is the group -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, and Zi, Z , Z3, Z , and Z5 are as defined above, with a halogenating reagent followed in the presence of water by a thiourea compound having the formula,
Figure imgf000028_0001
to provide the compound having the formula (le),
Figure imgf000028_0002
In the above process, in one embodiment, R* is hydrogen, whereby the process provides a compound having the formula (If),
Figure imgf000028_0003
In another embodiment, R4 may be a group having the formula,
Figure imgf000028_0004
wherein R15 and Rι6 are as defined herein, whereby said process provides a compound having the formula (Hi),
Figure imgf000028_0005
wherein R15, Rι6, Zi, Z2, Z3, Z4, Z5, R2o and R2ι are as defined herein. In yet another embodiment, R4 is a group having the formula,
Figure imgf000029_0001
wherein Y, R15 and Rι6 are as defined herein, wherein said process provides a compound having the formula (H),
Figure imgf000029_0002
In yet another embodiment, R is a group having the formula,
"' 1 or ""V1
H3C HaC
In another embodiment of the above process, e.g., when is hydrogen to provide compounds (If), the process may further comprise reacting the compound of the formula
Figure imgf000029_0003
with a pyrimidine compound having the formula,
Figure imgf000029_0004
4a, wherein X and Y are leaving groups, and R15 and Rι6 are independently selected from hydrogen, alkyl and substituted alkyl, to provide a compound having the formula,
Figure imgf000030_0001
wherein Y, Ri5, Rι6, Zi, Z2, Z3, Z4, and Z5 are as defined above.
In another embodiment of the above process, e.g., when R4 is hydrogen to provide compounds (If), the process may further comprise reacting the compound of the formula
Figure imgf000030_0002
with a pyrimidine compound having the formula,
Figure imgf000030_0003
4a, (for example reacting with a base or by metal catalysis) wherein X and Y are leaving groups, and R15 and Rι6 are independently selected from hydrogen, alkyl and substituted alkyl, to provide a compound having the formula,
Figure imgf000030_0004
wherein Y, Rι5, Rι6, Zls Z2, Z3, Z4, and Z5 are as defined above. Compounds (Ig) may optionally further be reacted with an amine having the formula'NHR2oR2i, wherein R2o and R2ι are independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R2o and R2ι can be taken together to form a heterocyclo, to provide a compound having the formula (Hi),
Figure imgf000031_0001
wherein Rι5, Rι6, Zi, Z2, Z3, Z , Z5, R2o and R2i are as defined above.
In one embodiment, the amine NHR20R2ι is piperazine in turn optionally substituted with hydroxy(alkyl), more preferably hydroxyethyl.
In one embodiment, the amine NHR20R21 is
Figure imgf000031_0002
In another embodiment, when 4. is hydrogen to provide compounds (If), the process may further comprise reacting the compound of the formula
Figure imgf000031_0003
with a pyrimidine compound having the formula,
Figure imgf000031_0004
wherein R15, Rι6 , R2o and R2ι are defined as above, to provide a compound having the formula (Hi),
Figure imgf000031_0005
Other variations of the above processes are also contemplated as within the scope of the invention, including processes involving further elaboration of the 2- amino-thiazole-5-aromatic amides.
Hi one embodiment, the present invention provides a crystalline monohydrate of the compound of formula (IV)
Figure imgf000033_0001
(IV).
In another embodiment, the monohydrate form is in substantially pure form.
In another embodiment, the monohydrate form is in substantially pure form, wherein substantially pure is greater than 90 percent pure.
In another embodiment, the monohydrate form of the compound of Formula
(IV) is characterized by an x-ray powder diffraction pattern substantially in accordance with that shown in Figure 1.
Hi another embodiment, the monohydrate form of the compound of Formula (TV) is characterized by differential scanning calorimetry thermogram and a thermogravimetric anaylsis substantially in accordance with that shown in Figure 2.
Hi another embodiment, the monohydrate form of the compound of Formula
(IV) is characterized by an x-ray powder diffraction pattern (CuKα λ=1.5418A at a temperature of about 23 °C) comprising four or more 2Θ values (alternatively, comprising five or more, six or more, or comprising 2Θ values) selected from the group consisting of: 18.0± 0.2, 18.4± 0.2, 19.2± 0.2, 19.6+ 0.2, 21.2+ 0.2, 24.5± 0.2,
25.9± 0.2, and 28.0± 0.2.
Hi another embodiment, the monohydrate form of the compound of Formula
(IV) is characterized by an x-ray powder diffraction pattern (CuKα λ=1.5418A at a temperature of about 23 °C) comprising four or more 2Θ values (alternatively, comprising five or more, six or more, or comprising 2Θ values) selected from the group consisting of: 4.6± 0.2, 11.2± 0.2, 13.8+ 0.2, 15.2± 0.2, 17.9± 0.2, 19.1+ 0.2,
19.6+ 0.2, 23.2± 0.2, 23.6± 0.2. hi another embodiment, the monohydrate form of the compound of Formula (IV) is characterized by unit cell parameters approximately equal to the following:
Cell dimensions: a(A) = 13.862(1); b(A)= 9.286(1); c(A) = 38.143(2);
Volume = 4910(1) A3 Space group Pbca
Molecules/unit cell 8
Density (calculated) (g/cm3) 1.300 wherein the compound is at a temperature of about -50°C. Hi another embodiment, the monohydrate form of the compound of Formula
(IV) there is one water molecule per molecule of formula (IV).
In another embodiment, the present invention provides a crystalline butanol solvate of the compound of formula (IV)
Figure imgf000034_0001
(IV).
In another embodiment, the butanol solvate form of the compound of Formula (IV) is characterized by unit cell parameters approximately equal to the following: Cell dimensions: a(A) = 22.8102(6); b(A)= 8.4691(3); c(A) = 15.1436(5); Volume = 2910.5(2) A3 Space group P2ι/a Molecules/unit cell 4 Density (calculated) (g/cm3) 1.283.
Hi another embodiment, the crystalline butanol solvate of the compound of Formula (IV) is characterized by an x-ray powder diffraction pattern (CuKα λ=1.5418A at a temperature of about 23°C) comprising four or more 2Θ values (alternatively, comprising five or more, six or more, or comprising 2Θ values) selected from the group consisting of: 5.9 ± 0.2, 12.0 ± 0.2, 13.0 ± 0.2, 17.7 ± 0.2, 24.1 ± 0.2, and 24.6 ± 0.2.
Hi another embodiment, the present invention is directed to the crystalline ethanol solvate of the compound of formula (IV). In another embodiment, the crystalline ethanol solvate of the compound of Formula (TV) is characterized by an x-ray powder diffraction pattern (CuKα λ=1.5418A at a temperature of about 23 °C) comprising four or more 2Θ values (alternatively, comprising five or more, six or more, or comprising 2Θ values) selected from the group consisting of: 5.8 ± 0.2, 11.3 ± 0.2, 15.8 ± 0.2, 17.2 ± 0.2, 19.5 ± 0.2, 24.1 + 0.2, 25.3 ± 0.2, and 26.2 ± 0.2.
Hi another embodiment, the present invention is directed to the crystalline neat form of the compound of formula (TV).
In another embodiment, the crystalline neat form of the compound of Formula (TV) is characterized by an x-ray powder diffraction pattern (CuKα λ=1.5418A at a temperature of about 23 °C) comprising four or more 2Θ values (alternatively, comprising five or more, six or more, or comprising 2Θ values) selected from the group consisting of: 6.8 ± 0.2, 11.1 ± 0.2, 12.3 ± 0.2, 13.2 ± 0.2, 13.7 ± 0.2, 16.7 ± 0.2, 21.0 ± 0.2,' 24.3 ± 0.2, and 24.8 ± 0.2. Hi another embodiment, the present invention describes a pharmaceutical composition comprising a therapeutically effective amount of at least one of the crystalline forms of the compound of Formula (TV) and a pharmaceutically acceptable carrier.
H another embodiment, the present invention describes a method for the treatment of cancer which comprises administering to a host in need of such treatment a therapeutically effective amount of at least one of the crystalline forms of the compound of Formula (TV).
In another embodiment, the present invention describes a method of treating oncological disorders which comprises administering to a host in need of such treatment a therapeutically effective amount of at least one of the crystalline forms of the compound of Formula (IN), wherein the disorders are selected from chronic myelogenous leukemia (CML), gastrointestinal stromal tumor (GIST), small cell lung cancer (SCLC), non-small cell lung cancer (ΝSCLC), ovarian cancer, melanoma, mastocytosis, germ cell tumors, acute myelogenous leukemia (AML), pediatric sarcomas, breast cancer, colorectal cancer, pancreatic cancer, and prostate cancer. Hi another embodiment, the present invention is directed to a use of the at least one of the crystalline forms of the compound of Formula (IN), in the preparation of a medicament for the treatment of oncological disorders, such as those described herein. Hi another embodiment, the present invention is directed to a method of treating of oncological disorders, as described herein, which are resistant or intolerant to Gleevec ® (STI-571), comprising administering to a host in need of such treatment a therapeutically effective amount of the compound of Formula (IN) or at least one of the crystalline forms of the compound of Formula (TV).
This invention also encompasses all combinations of alternative aspects of the invention noted herein. It is understood that any and all embodiments of the present invention may be taken in conjunction with any other embodiment to describe additional embodiments of the present invention. Furthermore, any elements of an embodiment are meant to be combined with any and all other elements from any of the embodiments to describe additional embodiments.
UTILITY The compounds of formula (I) prepared according to the inventive process herein inhibit protein tyrosine kinases, especially Src-family kinases such as Lck, Fyn, Lyn, Src, Yes, Hck, Fgr and Blk, and are thus useful in the treatment, including prevention and therapy, of protein tyrosine kinase-associated disorders such as immunologic and oncologic disorders. The compounds of formula (I) also may inhibit receptor tyrosine kinases including HER1 and HER2 and therefore be useful in the treatment of proliferative disorders such as psoriasis and cancer. The ability of these compounds to inhibit HER1 and other receptor kinases will permit their use as anti-angiogenic agents to treat disorders such as cancer and diabetic retinopathy.
"Protein tyrosine kinase-associated disorders" are those disorders which result from aberrant tyrosine kinase activity, and/or which are alleviated by the inhibition of one or more of these enzymes. For example, Lck inhibitors are of value in the treatment of a number of such disorders (for example, the treatment of autoimmune diseases), as Lck inhibition blocks T cell activation. The treatment of T cell mediated diseases, including inhibition of T cell activation and proliferation, is a particularly preferred use for compounds of formula (I) prepared according to the process herein. Use of the compounds of formula (I) in treating protein tyrosine kinase- associated disorders is exemplified by, but is not limited to, treating a range of disorders such as: transplant (such as organ transplant, acute transplant or heterograft or homograft (such as is employed in burn treatment)) rejection; protection from ischemic or reperfusion injury such as ischemic or reperfusion injury incurred during organ transplantation, myocardial infarction, stroke or other causes; transplantation tolerance induction; arthritis (such as rheumatoid arthritis, psoriatic arthritis or osteoarthritis); multiple sclerosis; chronic obstructive pulmonary disease (COPD), such as emphysema; inflammatory bowel disease, including ulcerative colitis and Crohn's disease; lupus (systemic lupus erythematosis); graft vs. host disease; T-cell mediated hypersensitivity diseases, including contact hypersensitivity, delayed-type hypersensitivity, and gluten-sensitive enteropathy (Celiac disease); psoriasis; contact dermatitis (including that due to poison ivy); Hashimoto's thyroiditis; Sjogren's syndrome; Autoimmune Hyperthyroidism, such as Graves' Disease; Addison's disease (autoimmune disease of the adrenal glands); Autoimmune polyglandular disease (also known as autoimmune polyglandular syndrome); autoimmune alopecia; pernicious anemia; vitiligo; autoimmune hypopituatarism; Guillain-Barre syndrome; other autoimmune diseases; cancers, including cancers where Lck or other Src-family kinases such as Src are activated or overexpressed, such as colon carcinoma and thymoma, and cancers where Src-family kinase activity facilitates tumor growth or survival; glomerulonephritis; serum sickness; uticaria; allergic diseases such as respiratory allergies (asthma, hayfever, allergic rhinitis) or skin allergies; scleracierma; mycosis fungoides; acute inflammatory responses (such as acute respiratory distress syndrome and ishchemia/reperfusion injury); dermatomyositis; alopecia areata; chronic actinic dermatitis; eczema; Behcet's disease; Pustulosis palmoplanteris; Pyoderma gangrenum; Sezary's syndrome; atopic dermatitis; systemic schlerosis; and morphea.
The compounds of the present invention are useful for the treatment of cancers such as chronic myelogenous leukemia (CML), gastrointestinal stromal tumor (GIST), small cell lung cancer (SCLC), non-small cell lung cancer (NSCLC), ovarian cancer, melanoma, mastocytosis, germ cell tumors, acute myelogenous leukemia (AML), pediatric sarcomas, breast cancer, colorectal cancer, pancreatic cancer, prostate cancer and others known to be associated with protein tyrosine kinases such as, for example, SRC, BCR-ABL and c-KIT. The compounds of the present invention are also useful in the treatment of cancers that are sensitive to and resistant to chemotherapeutic agents that target BCR-ABL and c-KIT, such as, for example, Gleevec® (STI-571). hi one embodiment of the invention, for example, the compound of the formula (IN) (including, but not limited to the crystalline forms of that compound described herein, such as the crystalline monohydrate) is useful in the treatment of patients resistant or intolerant to Gleevec ® (STI-571) for diseases such as chronic myelogenous leukemias (CML), or other cancers (including other leukemias) as described herein. H another embodiment of the invention a compound of Formulas I is administered in conjunction with at least one anti-neoplastic agent.
As used herein, the phrase "anti-neoplastic agent" or "anti-cancer agent" is synonymous with "chemotherapeutic agent" and/or "anti-proliferative agent" and refers to compounds that prevent cancer, or hyperproliferative cells from multiplying. Anti-proliferative agents prevent cancer cells from multiplying by: (1) interfering with the cell's ability to replicate DΝA and (2) inducing cell death and/or apoptosis in the cancer cells.
Classes of compounds that may be used as anti-proliferative cytotoxic agents and/or anti-proliferative agents include the following: Alkylating agents (including, without limitation, nitrogen mustards, ethyleni ine derivatives, alkyl sulfonates, nitrosoureas and triazenes): Uracil mustard, Chlormethine, Cyclophosphamide (Cytoxan®), ffosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylene-melamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, Dacarbazine, and Temozolomide. Antimetabolites (including, without limitation, folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors): Methotrexate, 5-Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6- Thioguanine, Fludarabine phosphate, Pentostatine, and Gemcitabine.
Natural products and their derivatives (for example, vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins): Ninblastine, Nincristine,
Nindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin,
Idarubicin, Ara-C, paclitaxel (paclitaxel is commercially available as Taxol®), Mithramycin, Deoxyco-formycin, Mitomycin-C, L-Asparaginase, H terferons (especially TFN-a), Etoposide, and Teniposide.
Other anti-proliferative cytotoxic agents and/or anti-proliferative agents are navelbene, CPT-11, anastrazole, letrazole, capecitabine, reloxafine, cyclophosphamide, ifosamide, and droloxafine.
The phrase "radiation therapy" includes, but is not limited to, x-rays or gamma rays which are delivered from either an externally applied source such as a beam or by implantation of small radioactive sources. Radiation therapy may be useful in combination with compounds of the present invention. The following may also be useful when administered in combination with compounds of the present invention.
Microtubule affecting agents interfere with cellular mitosis and are well known in the art for their anti-proliferative cytotoxic activity. Microtubule affecting agents useful in the invention include, but are not limited to, allocolchicine (NSC 406042), Halichondrin B (NSC 609395), colchicine (NSC 757), colchicine derivatives (e.g., NSC 33410), dolastatin 10 (NSC 376128), maytansine (NSC 153858), rhizoxin (NSC 332598), paclitaxel (Taxol®, NSC 125973), Taxol® derivatives (e.g., derivatives (e.g., NSC 608832), thiocolchicine NSC 361792), trityl cysteine (NSC 83265), vinblastine sulfate (NSC 49842), vincristine sulfate (NSC 67574), natural and synthetic epothilones including but not limited to epothilone A, epothilone B, epothilone C, epothilone D, desoxyepothilone A, desoxyepothilone B, [1S- [lR:1:,3R*(E),7R*,10S*,llR*,12R*,16S*]]-7-ll-dihydroxy-8,8,10,12,16- pentamethyl-3 - [ 1 -methyl-2-(2-methyl-4-thiazolyl)ethenyl] -4-aza- 17 oxabicyclo [14.1.0]heptadecane-5,9-dione (disclosed in US Patent 6,262,094, issued July 17, 2001), [lS-[lR*,3R*(E),7R*,10S*,llR*,12R*,16S*]]-3-[2-[2-(aminomethyl)-4- thiazolyl] - 1 -methylethenyl] -7,11 -dihydroxy-8 ,8 , 10,12,16-pentamethyl-4- 17- dioxabicyclo[14.1.0]- heptadecane-5,9-dione (disclosed in USSN 09/506,481 filed on February 17, 2000, and examples 7 and 8 herein), [IS 1R*,3R*(E),7R*,10S*,11R*,12R*, lόS^Jj^αi-dihydro y-S.δ oα^lό-pentamethyl- 3-[l-methyl-2-(2-methyl-4-thiazolyl)ethenyl]-4-aza-17oxabicyclo[14.1.0]- heptadecane-5,9-dione, [lS-[lR*,3R:1:(E),7R*,10S*,HR*,12R*,16S:i:]]-3-[2-[2-
(Aminomethyl)-4-thiazolyl]-l-methylethenyl]-7,ll-dihydroxy-8,8,10,12,16- pentamethyl-4,17-dioxabicyclo[14.1.0]heptadecane-5,9-dione, and derivatives thereof; and other microtubule-disruptor agents. Additional antineoplastic agents include, discodermolide (see Service, (1996) Science, 274:2009) estramustine, nocodazole, MAP4, and the like. Examples of such agents are also described in the scientific and patent literature, see, e.g., Bulinski (1997) J. Cell Sci. 110:3055 3064; Panda (1997) Proc. Natl. Acad. Sci. USA 94:10560-10564; Muhlradt (1997) Cancer Res. 57:3344- 3346; Nicolaou (1997) Nature 387:268-272; Vasquez (1997) Mol Biol. Cell. 8:973- 985; Panda (1996) J. Biol. Chem 271:29807-29812.
In cases where it is desirable to render aberrantly proliferative cells quiescent in conjunction with or prior to treatment with the chemotherapeutic methods of the invention, hormones and steroids (including synthetic analogs): 17a-Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, hlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, Zoladex can also be administered to the patient.
Also suitable for use in the combination chemotherapeutic methods of the invention are antiangiogenics such as matrix metalloproteinase inhibitors, and other VEGF inhibitors, such as anti-VEGF antibodies and small molecules such as ZD6474 ; and SU6668 are also included. Anti- Her2 antibodies from Genetech may also be utilized. A suitable EGFR inhibitor is EKB-569 (an irreversible inhibitor). Also included are Imclone antibody C225 immunospecific for the EGFR, and src inhibitors.
Also suitable for use as an antiproliferative cytostatic agent is Casodex™ which renders androgen-dependent carcinomas non-proliferative. Yet another example of a cytostatic agent is the antiestrogen Tamoxifen which inhibits the proliferation or growth of estrogen dependent breast cancer. Inhibitors of the transduction of cellular proliferative signals are cytostatic agents. Examples are epidermal growth factor inhibitors, Her-2 inhibitors, MEK-1 kinase inhibitors, MAPK kinase inhibitors, PI3 inhibitors, Src kinase inhibitors, and PDGF inhibitors. As mentioned, certain anti-proliferative agents are anti-angiogenic and antivascular agents and, by interrupting blood flow to solid tumors, render cancer cells quiescent by depriving them of nutrition. Castration, which also renders androgen dependent carcinomas non-proliferative, may also be utilized. Starvation by means other than surgical disruption of blood flow is another example of a cytostatic agent. A particular class of antivascular cytostatic agents is the combretastatins. Other exemplary cytostatic agents include MET kinase inhibitors, MAP kinase inhibitors, inhibitors of non-receptor and receptor tyrosine kinases, inhibitors of integrin signaling, and inhibitors of insulin-like growth factor receptors.
Also suitable are anthracyclines (e.g., daunorubicin, doxorubicin), cytarabine (ara-C; Cytosar-U®); 6-thioguanine (Tabloid®), mitoxantrone (Novantrone®) and etoposide (NePesid®),amsacrine (AMSA), and all-trans retinoic acid (ATRA). The compounds of the present invention may be useful in combination with
BCR-ABL inhibitors such as, but not limited to, Gleevec® (imatinib, STI-571) or AMΝ-107, the compound shown below
Figure imgf000041_0001
The compounds of the present invention may be useful in combination with anti-cancer compounds such as fentanyl, doxorubicin, interferon alfa-n3, palonosetron dolasetron anastrozole, exemestane, bevacizumab, bicalutamide, cisplatin, dacarbazine, cytarabine, clonidine, epirubicin, levamisole, toremifene, fulvestrant, letrozole, tamsulosin, gallium nitrate, trastuzumab, altretamine, hydroxycarbamide, ifosfamide, interferon alfacon-1, gefitinib, granisetron, leuprorelin, dronabinol, megestrol, pethidine, promethazine, morphine, vinorelbine, pegfilgrastim, filgrastim, nilutamide, thiethylperazine, leuprorelin, pegaspargase, muromonab-CD3, porfimer sodium, cisplatin, abarelix, capromab, samarium SM153 lexidronam, paclitaxel, docetaxel, etoposide, triptorelin, valrubicin, nofetumomab merpentan technetium 99m Tc, vincristine, capecitabine, strptozocin, and ondansetron. Thus, the present invention provides methods for the treatment of a variety of cancers, including, but not limited to, the following: carcinoma including that of the bladder (including accelerated and metastatic bladder cancer), breast, colon (including colorectal cancer), kidney, liver, lung (including small and non-small cell lung cancer and lung adenocarcinoma), ovary, prostate, testes, genitourinary tract, lymphatic system, rectum, larynx, pancreas (including exocrine pancreatic carcinoma), esophagus, stomach, gall bladder, cervix, thyroid, and skin (including squamous cell carcinoma); hematopoietic tumors of lymphoid lineage including leukemia, acute lymphocytic leukemia, acute lymphoblastic leukemia, B-cell lymphoma, T-cell lymphoma, Hodgkin's lymphoma, non-Hodgkin's lymphoma, hairy cell lymphoma, histiocytic lymphoma, and Burketts lymphoma; hematopoietic tumors of myeloid lineage including acute and chronic myelogenous leukemias, myelodysplastic syndrome, myeloid leukemia, and promyelocytic leukemia; tumors of the central and peripheral nervous system including astrocytoma, neuroblastoma, glioma, and schwannomas; tumors of mesenchymal origin including fibrosarcoma, rhabdomyoscarcoma, and osteosarcoma; and other tumors including melanoma, xenoderma pigmentosum, keratoactanthoma, seminoma, thyroid follicular cancer, and teratocarcinoma. The present invention provides methods for the treatment of a variety of non-cancerous proliferative diseases.
The invention is useful to treat GIST, breast cancer, pancreatic cancer, colon cancer, NSCLC, CML, and ALL, sarcoma, and various pediatric cancers.
The compounds of the present invention are protein tyrosine kinase inhibitors and as such are useful in the treatment of immunological disorders in addition to oncological disorders. U.S. Patent No. 6,596,746 describes the utility of the compound in immunological disorders and is hereby incorporated by reference for the description of the compound in such immunological disorders.
The present invention also encompasses a pharmaceutical composition useful in the treatment of cancer, comprising the administration of a therapeutically effective amount of the combinations of this invention, with or without pharmaceutically acceptable carriers or diluents. The pharmaceutical compositions of this invention comprise an anti-proliferative agent or agents, a formula I compound, and a pharmaceutically acceptable carrier. The methods entail the use of a neoplastic agent in combination with a Formula I compound. The compositions of the present invention may further comprise one or more pharmaceutically acceptable additional ingredient(s) such as alum, stabilizers, antimicrobial agents, buffers, coloring agents, flavoring agents, adjuvants, and the like. The antineoplastic agents, Formula I, compounds and compositions of the present invention may be administered orally or parenterally including the intravenous, intramuscular, intraperitoneal, subcutaneous, rectal and topical routes of administration. The present invention also provides using the compounds obtained with the inventive process to further prepare pharmaceutical compositions capable of treating Src-kinase associated conditions, including the conditions described above. The said compositions may contain other therapeutic agents. Pharmaceutical compositions may be formulated by employing conventional solid or liquid vehicles or diluents, as well as pharmaceutical additives of a type appropriate to the mode of desired administration (e.g., excipients, binders, preservatives, stabilizers, flavors, etc.) according to techniques such as those well known in the art of pharmaceutical formulations.
The said pharmaceutical compositions may be administered by any means suitable for the condition to be treated, which may depend on the need for site-specific treatment or quantity of drug to be delivered. Topical administration is generally preferred for skin-related diseases, and systematic treatment preferred for cancerous or pre-cancerous conditions, although other modes of delivery are contemplated. For example, the compounds of formula (1) may be delivered orally, such as in the form of tablets, capsules, granules, powders, or liquid formulations including syrups; topically, such as in the form of solutions, suspensions, gels or ointments; sublingually; bucally; parenterally, such as by subcutaneous, intravenous, intramuscular or intrasternal injection or infusion techniques (e.g., as sterile injectable aq. or non-aq. solutions or suspensions); nasally such as by inhalation spray; topically, such as in the form of a cream or ointment; rectally such as in the form of suppositories; or liposomally.
Dosage unit formulations containing non-toxic, pharmaceutically acceptable vehicles or diluents may be administered. The compounds of formula (I), prepared according to the inventive process, may be administered in a form suitable for immediate release or extended release. Immediate release or extended release may be achieved with suitable pharmaceutical compositions or, particularly in the case of extended release, with devices such as subcutaneous implants or osmotic pumps. Exemplary compositions for topical administration include a topical carrier such as PLASTIBASE® (mineral oil gelled with polyethylene).
Exemplary compositions for oral administration include suspensions which may contain, for example, microcrystalline cellulose for imparting bulk, alginic acid or sodium alginate as a suspending agent, methylcellulose as a viscosity enhancer, and sweeteners or flavoring agents such as those known in the art; and immediate release tablets which may contain, for example, microcrystalline cellulose, dicalcium phosphate, starch, magnesium stearate and/or lactose and/or other excipients, binders, extenders, disintegrants, diluents and lubricants such as those known in the art. The compounds of formula (I) may also be orally delivered by sublingual and/or buccal administration, e.g., with molded, compressed, or freeze-dried tablets. Exemplary compositions may include fast-dissolving diluents such as mannitol, lactose, sucrose, and/or cyclodextrins. Also included in such formulations may be high molecular weight excipients such as celluloses (ANICEL®) or polyethylene glycols (PEG); an excipient to aid mucosal adhesion such as hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), sodium carboxymethyl cellulose (SCMC), and/or maleic anhydride copolymer (e.g., GAΝTREZ®); and agents to control release such as polyacrylic copolymer (e.g., CARBOPOL 934®). Lubricants, glidants, flavors, coloring agents and stabilizers may also be added for ease of fabrication and use. An example of a composition for oral administration is the compound of formula (IN), lactose monohydrate (intra-granular phase), microcrystalline cellulose(intra-granular phase), croscarmellose sodium(intra-granular phase), hydroxypropyl cellulose(intra-granular phase), microcrystalline cellulose (extra- ' granular phase), croscarmellose sodium (extra-granular phase), and magnesium stearate (extragranular phase).
Exemplary compositions for nasal aerosol or inhalation administration include solutions which may contain, for example, benzyl alcohol or other suitable preservatives, absorption promoters to enhance absorption and/or bioavailability, and/or other solubilizing or dispersing agents such as those known in the art.
Exemplary compositions for parenteral administration include injectable solutions or suspensions which may contain, for example, suitable non-toxic, parenterally acceptable diluents or solvents, such as mannitol, 1,3-butanediol, water, Ringer's solution, an isotonic sodium chloride solution, or other suitable dispersing or wetting and suspending agents, including synthetic mono- or diglycerides, and fatty acids, including oleic acid.
Exemplary compositions for rectal administration include suppositories which may contain, for example, suitable non-irritating excipients, such as cocoa butter, synthetic glyceride esters or polyethylene glycols, which are solid at ordinary temperatures but liquefy and/or dissolve in the rectal cavity to release the drug.
The effective amount of a compound of formula (I) may be determined by one of ordinary skill in the art, and includes exemplary dosage amounts for a mammal of from about 0.05 to 100 mg/kg of body weight of active compound per day, which may be administered in a single dose or in the form of individual divided doses, such as from 1 to 4 times per day. It will be understood that the specific dose level and frequency of dosage for any particular subject may be varied and will depend upon a variety of factors, including the activity of the specific compound employed, the metabolic stability and length of action of that compound, the species, age, body weight, general health, sex and diet of the subject, the mode and time of administration, rate of excretion, drug combination, and severity of the particular condition. Preferred subjects for treatment include animals, most preferably mammalian species such as humans, and domestic animals such as dogs, cats, horses, and the like. Thus, when the term "patient" is used herein, this term is intended to include all subjects, most preferably mammalian species, that are affected by mediation of Src kinase levels.
When administered intravenously, the compounds of the present invention, including the crystalline forms of the compounds of formula IN, are administered using the formulations of the invention. In one embodiment , the compounds of the present invention, are administered by IN infusion over a period of from about 10 minutes to about 3 hours, preferably about 30 minutes to about 2 hours, more preferably about 45 minutes to 90 minutes, and most preferably about 1 hour. Typically, the compounds are administered intravenously in a dose of from about 0.5 mg/m2 to 65 mg/m2, preferably about 1 mg/m2 to 50 mg/m2, more preferably about 2.5 mg/m2 to 30 mg/m2, and most preferably about 25 mg/m2. One of ordinary skill in the art would readily know how to convert doses from mg/kg to mg/m2 given either or both the height and or weight of the patient (See, e.g., http://www.fda.gov/cder/cancer/animalframe.htm).
As discussed above, compounds of the present invention, including the crystalline forms of the compounds of formula IN can be administered orally, intravenously, or both. In particular, the methods of the invention encompass dosing protocols such as once a day for 2 to 10 days, preferably every 3 to 9 days, more preferably every 4 to 8 days and most preferably every 5 days. In one embodiment there is a period of 3 days to 5 weeks, alternatively 4 days to 4 weeks, or 5 days to 3 weeks, or 1 week to 2 weeks, in between cycles where there is no treatment. Hi another embodiment the compounds of the present invention, including the crystalline forms of the compounds of formula TV can be administered orally, intravenously, or both, once a day for 3 days, with a period of 1 week to 3 weeks in between cycles where there is no treatment. Hi yet another embodiment the compounds of the present invention, the crystalline forms of the compounds of formula TV, can be administered orally, intravenously, or both, once a day for 5 days, with a period of 1 week to 3 weeks in between cycles where there is no treatment. hi another embodiment the treatment cycle for administration of the compounds of the present invention, the crystalline forms of the compounds of formula IN, is once daily for 5 consecutive days and the period between treatment cycles is from 2 to 10 days, or alternatively one week. In one embodiment, a compound of the present invention, for example, a compound of formula IN, is administered once daily for 5 consecutive days, followed by 2 days when there is no treatment.
The compounds of the present invention, the crystalline forms of the compounds of formula TV, can also be administered orally, intravenously, or both once every 1 to 10 weeks, every 2 to 8 weeks, every 3 to 6 weeks, alternatively every 3 weeks. In another method of the invention, the compounds of the present invention, the crystalline forms of the compounds of formula IN, are administered in a 28 day
' cycle wherein the compounds are intravenously administered on days 1, 7, and 14 and orally administered on day 21. Alternatively, the compounds of the present invention, the crystalUne forms of the compounds of formula IN, are administered in a 28 day cycle wherein the compound of formulae TV are orally administered on day 1 and intravenously administered on days 7, 14, and 28.
According to the methods of the invention, the compounds of the present invention, including compounds of formulae TV, are administered until the patient shows a response, for example, a reduction in tumor size, or until dose limiting toxicity is reached.
Compounds within the scope of formula (I) may be tested for activity as inhibitors of protein kinases using the assays described below, or variations thereof that are within the level ordinary skill in the art.
Cell assays .
(1) Cellular tyrosine phosphorylation
Jurkat T cells are incubated with the test compound and then stimulated by the addition of antibody to CD3 (monoclonal antibody G19-4). Cells are lysed after 4 minutes or at another desired time by the addition of a lysis buffer containing ΝP-40 detergent. Phosphorylation. of proteins is detected by anti-phosphotyrosine immunoblotting. Detection of phosphorylation of specific proteins of interest such as
ZAP-70 is detected by immunoprecipitation with anti-ZAP-70 antibody followed by anti-phosphotyrosine immunoblotting. Such procedures are described in Schieven, G.L., Mittler, R.S., Nadler, S.G., Kirihara, J.M., Bolen, J.B., Kanner, S.B., and
Ledbetter, J.A., "ZAP-70 tyrosine kinase, CD45 and T cell receptor involvement in
UV and H2O2 induced T cell signal transduction", J. Biol Chem., 269, 20718-20726
(1994), and the references incorporated therein. The Lck inhibitors inhibit the tyrosine phosphorylation of cellular proteins induced by anti-CD3 antibodies. For the preparation of G19-4, see Hansen, J.A., Martin, P.J., Beatty, P.G.,
Clark, E.A., and Ledbetter, J.A., "Human T lymphocyte cell surface molecules defined by the workshop monoclonal antibodies," in Leukocyte Typing I, A. Bernard, J. Boumsell, J. Dausett, C. Milstein, and S. Schlossman, eds. (New York: Springer Verlag), p. 195-212 (1984); and Ledbetter, J.A., June, C.H., Rabinovitch, P.S., Grossman, A., Tsu, T.T., and Imboden, J.B., "Signal transduction through CD4 receptors: stimulatory vs. inhibitory activity is regulated by CD4 proximity to the CD3/T cell receptor", Eur. J. Immunol, 18, 525 (1988).
(2) Calcium assay
Lck inhibitors block calcium mobilization in T cells stimulated with anti-CD3 antibodies. Cells are loaded with the calcium indicator dye indo-1, treated with anti- CD3 antibody such as the monoclonal antibody G19-4, and calcium mobilization is measured using flow cytometry by recording changes in the blue/violet indo-1 ratio as described in Schieven, G.L., Mittler, R.S., Nadler, S.G., Kirihara, J.M., Bolen, J.B., Kanner, S.B., and Ledbetter, J.A., "ZAP-70 tyrosine kinase, CD45 and T cell receptor involvement in UV and H2O2 induced T cell signal transduction", /. Biol. Chem., 269, 20718-20726 (1994), and the references incorporated therein.
(3) Proliferation assays
Lck inhibitors inhibit the proliferation of normal human peripheral blood T cells stimulated to grow with anti-CD3 plus anti-CD28 antibodies. A 96 well plate is coated with a monoclonal antibody to CD3 (such as G19-4), the antibody is allowed to bind, and then the plate is washed. The antibody bound to the plate serves to stimulate the cells. Normal human peripheral blood T cells are added to the wells along with test compound plus anti-CD28 antibody to provide co-stimulation. After a desired period of time (e.g., 3 days), the [3H]-thymidine is added to the cells, and after further incubation to allow incorporation of the label into newly synthesized DNA, the cells are harvested and counted in a scintillation counter to measure cell proliferation.
The following examples illustrate the invention but should not be interpreted as a limitation thereon. EXAMPLES
Example 1 Preparation of intermediate:
(S)-l -sec-Butylthiourea
Figure imgf000049_0001
To a solution of S- sec-butyl-amine (7.3 lg, 0.1 mol) in chloroform (80 mL) at 0 °C was slowly added benzoyl isothiocyanate (13.44 mL, 0.1 mol). The mixture was allowed to warm to 10°C and stirred for 10 min. The solvent was then removed under reduced pressure, and the residue was dissolved in MeOH (80 mL). An aqueous solution (10 mL) of NaOH (4g, 0.1 mol) was added to this solution, and the mixture was stirred at 60°C for another 2h. The MeOH was then removed under reduced pressure, and the residue was stirred in water (50 mL). The precipitate was collected by vacuum filtration and dried to provide S-l-sec-butyl-thiourea (12.2 g, 92% yield). mp 133-134°C; 1H NMR (500 MHz, DMSO-D6) δ 7.40 (s, IH), 7.20 (br s, IH), 6.76 (s, IH), 4.04 (s, IH), 1.41 (m, 2H), 1.03 (d, J= 6.1 Hz, 3H), 0.81 (d, J = 7.7 Hz, 3H); 13C NMR (125 MHz, DMSO-D6) δ 182.5, 50.8, 28.8, 19.9, 10.3; LRMS m z 133.2 (M+H); Anal. Calcd for C52N2S: C, 45.41; H, 9.14.; N, 21.18; S, 24.25. Found: C,
45.49; H, 8.88; N, 21.32; S, 24.27.
Example 2
Preparation of intermediate:
(R)-l -sec-Butylthiourea L X s
N NH2
H (R)-l -sec-Butylthiourea was prepared in 92% yield according to the general method outlined for Example 1. mp 133-134°C; 1H NMR(500 MHz, DMSO) δ 0.80(m, 3H, J=7.7), 1.02(d, 3H, J=6.1), 1.41(m, 2H), (3.40, 4.04)(s, IH), 6.76(s, IH), 7.20(s, br, IH), 7.39(d, IH, J=7.2); 13C NMR (500MHz, DMSO) δ: 10.00, 19.56, 28.50, 50.20, 182.00; m/z 133.23 (M+H); Anal. Calcd for C52N2S: C, 45.41; H, 9.14.; N, 21.18; S, 24.25. Found: C, 45.32; H, 9.15; N, 21.14; S, 24.38.
Example 3
Preparation of:
Figure imgf000050_0001
3A.
Figure imgf000050_0002
To a solution of 3-amino-N-methyl-4-methylbenzamide hydrochloride (1.0 g, 5 mmol) in acetone (10 mL) at 0 °C was added pyridine (1.2 mL, 15 mmol) dropwise via syringe. 3-Methoxyacryloyl chloride (0.72 mL. 6.5 mmol) was added and the reaction stirred at room temperature for 1 h. The solution was cooled again to 0°C and IN HCl (1.5 mL) was added dropwise via pipet. The reaction mixture was stirred for 5 min, then water (8.5 mL) was added via an addition funnel. The acetone was removed in vacua and the resulting solution stirred for 4h. Crystallization began within 15 min. After stirring for 4 h, the vessel was cooled in an ice bath for 30 min, filtered, and rinsed with ice cold water (2 x 3 mL) to give compound 3A (0.99 g, 78 % yield) as a white solid. 1H NMR (400 MHz, CDC13) δ 8.95 (s, IH), 8.12 (br s, IH), 7.76 (s, IH), 7.29 (m, 2H), 7.05 (d, J = 7.9 Hz, IH), 5.47 (d, J= 12.3 Hz, IH), 3.48 (s, 3H), 2.54 (d, J = 4.7 Hz, 3H), 2.03 (s, 3H); HPLC rt 2.28 min (Condition A). 3B. Example 3 To a 50 mL RBF containing the above compound 3 A (0.5g, 2.0 mmol) was added THF (2.5 mL) and water (2 mL), followed by NBS (0.40 g, 2.22 mmol), and the solution was stirred for 90 min. R-sec-butylthiourea (Ex. 2) (267 mg), was added, and the solution was heated to 75 °C for 8 h. Cone. NH OH was added to adjust the pH to 10 followed by the addition of EtOH (15 mL). Water (15 mL) was added and the slurry stirred for 16 h, filtered, and washed with water to give Example 3 as a light brown solid (0.48 g, 69% yield, 98% purity). MS 347.1; HPLC 2.59.
Example 4
Preparation of:
Figure imgf000051_0001
Example 4 is prepared following the methods of Example 3 but using the appropriate acryl benzamide and Example 1.
Example 5
Preparation of:
N-(2-chloro-6-methγlphenyl)-2-(6-(4-(3-hydroxyethyl)piperazin-l-yl)-2- methylpyriιnidin-4-ylamino)thiazole-5-carboxamide (The compound of Formula (IV))
Figure imgf000051_0002
(IN)
5A. 1 -(6-Chloro-2-methylpyrimidin-4-yl)thiourea
Me (5A)
To a stirring slurry of 4-amino-5-chloro-2-methylρyrimidine (6.13g, 42.7 mmol) in THF (24 mL) was added ethyl isothiocyanatoformate (7.5 mL, 63.6 mmol), and the mixture heated to reflux. After 5h, another portion of ethyl isothiocyanato formate (1.0 mL, 8.5 mmol) was added and after lOh, a final portion (1.5 mL, 12.7 mmol) was added and the mixture stirred 6h more. The slurry was evaporated under vacuum to remove most of the solvent and heptane (6 mL) added to the residue. The solid was collected by vacuum filtration and washed with heptane (2 x 5 mL) giving 8.01 g (68 % yield) of the intermediate ethyl 6-chloro-2-methylpyrimidin-4- ylcarbamothioylcarbamate.
A solution of ethyl 6-chloro-2-methylpyrimidin-4-ylcarbamothioylcarbamate (275 mg, 1.0 mmol) and IN sodium hydroxide (3.5eq) was heated and stirred at 50°C for 2h. The resulting slurry was cooled to 20-22°C. The solid was collected by vacuum filtration, washed with water, and dried to give 185 mg of ls-(6-chloro-2- methylpyrimidin-4-yl)thiourea (91 % yield). 1H NMR (400MHz, DMSO-d6): 52.51 (S, 3H), 7.05 (s, IH), 9.35 (s,lH), 10.07 (s, IH), 10.91 (s, IH) ; 13C NMR (125MHz, DMSO-d6) δ: 25.25, 104.56, 159.19, 159.33, 167.36, 180.91.
5B. (E)-N-(2-Chloro-6-methylphenyl)-3-ethoxyacrylamide
Figure imgf000052_0001
To a cold stirring solution of 2-chloro-6-methylaniline (59.5 g 0.42 mol) and pyridine (68 ml, 0.63 mol) in THF (600 mL) was added 3-ethoxyacryloyl chloride (84.7 g, 0.63 mol) slowly keeping the temp at 0-5°C. The mixture was then warmed and stirred for 2 h. at 20°C. Hydrochloric acid (IN, 115 mL) was added at 0-10°C. The mixture was diluted with water (310 mL) and the resulting solution was concentrated under vacuum to a thick slurry. The slurry was diluted with toluene (275 mL) and stirred for 15 min. at 20-22°C then 1 h. at 0°C. The solid was collected by vacuum filtration, washed with water (2 x 75 mL) and dried to give 74.1 g (73.6 % yield) of (E)-N-(2-chloro-6-methylρhenyl)-3-ethoxyacrylamide/). 1H NMR (400 Hz, DMSO-d6) δ 1.26 (t, 3H, J= 7 Hz), 2.15 (s, 3H), 3.94 (q, 2H, J= 7 Hz), 5.58 (d, IH, J=12.4 Hz), 7.10-7.27 (m, 2H, J=7.5 Hz), 7.27-7.37 (d, IH, J=7.5 Hz), 7.45(d, IH, J=12.4 Hz), 9.28 (s, IH) ; 13C NMR (100MHz, CDC13) δ: 14.57, 18.96, 67.17, 97.99, 126.80, 127.44, 129.07, 131.32, 132.89, 138.25, 161.09, 165.36.
5C. 2-Amino-N-(2-chloro-6-methylphenyl)thiazole-5-carboxamide
Figure imgf000053_0001
To a mixture of compound 5B (5.00 g, 20.86 mmol) in 1,4-dioxane (27 mL) and water (27 mL) was added NBS (4.08 g, 22.9 mmol) at -10 to 0°C. The slurry was warmed and stirred at 20-22°C for 3h. Thiourea (1.60 g, 21 mmol) was added and the mixture heated to 80°C. After 2h, the resulting solution was cooled to 20-22° and cone, ammonium hydroxide (4.2 mL) was added dropwise. The resulting slurry was concentrated under vacuum to about half volume and cooled to 0-5°C. The solid was collected by vacuum filtration, washed with cold water (10 mL), and dried to give 5.3 g (94.9 % yield) of 2-aπιino-N-(2-chloro-6-methylphenyl)thiazole-5-carboxamide. 1H NMR (400 MHz, DMSO-d6) δ δ 2.19 (s, 3H), 7.09-7.29 (m, 2H, J=7.5), 7.29-7.43 (d, IH, J=7.5), 7.61 (s, 2H), 7.85 (s, IH), 9.63 (s, IH) ; 13C NMR (125MHz, DMSO-d6) δ: 18.18, 120.63, 126.84, 127.90, 128.86, 132.41, 133.63, 138.76, 142.88, 159.45, 172.02.
5D. 2-(6-Chloro-2-methylpyrimidin-4-ylamino)-N-(2-chloro-6- methylphenyl)thiazole-5-carboxamide
Figure imgf000053_0002
To a stirring solution of compound 5C (5.00 g, 18.67 mmol) and 4,6-dichloro- 2-methylpyrimidine (3.65 g 22.4/mmol) in THF (65 mL) was added a 30%"wt. solution of sodium t-butoxide in THF (21.1 g, 65.36 mmol) slowly with cooling to keep the temperature at 10-20°C. The mixture was stirred at room temperature for 1.5 h and cooled to 0-5°C. Hydrochloric acid, 2N (21.5 mL) was added slowly and the mixture stirred 1.75 h at 0-5°C. The solid was collected by vacuum filtration, washed with water (15 mL) and dried to give 6.63 g (86.4 % yield) of compound 5D. 1H NMR (400 MHz, DMSO-d6) δ 2.23 (s, 3H), 2.58 (s, 3H), 6.94 (s, IH), 7.18-7.34, ( m, 2H, J=7.5), 7.34-7.46 (d, IH, , J=7.5), 8.31 (s, IH), 10.02 (s, IH), 12.25 (s, IH).
5E. Example 5
To a mixture of compound 5D (4.00 g, 10.14 mmol) and hydroxyethylpiperazine (6.60 g, 50.69 mmol) in n-butanol (40 mL) was added DIPEA (3.53 mL, 20.26 mmol). The slurry was heated at 118°C for 4.5h, then cooled slowly to room temperature. The solid was collected by vacuum filtration, washed with n- butanol (5 mL), and dried. The product (5.11 g) was dissolved in hot 80% EtOH-H2O (80 mL), and the solution was clarified by filtration. The hot solution was slowly diluted with water (15 mL) and cooled slowly to room temperature. The solid was collected by vacuum filtration, washed with 50% ethanol-water (5 mL) and dried affording 4.27 g (83.2 % yield) of N-(2-chloro-6-methylphenyl)-2-(6-(4-(3- hydroxyethyl)piperazin-l-yl)-2-methylpyrirmdin-4-ylamino)thiazole-5-carboxamide as monohydrate. 1H NMR (400 MHz, DMSO-d6) δ 2.23 (s, 3H), 2.40 (s, 3H), 2.42 (t, 2H, J=6), 2.48 (t, 4H, J=6.3), 3.50 (m, 4H), 3.53 (q, 2H, J=6), 4.45 (t, IH, J=5.3), 6.04 (s, IH), 7.25 (t, IH, J=7.6), 7.27 (dd, IH, J=7.6, 1.7), 7.40 (dd, IH, J=7.6, 1.7), 8.21 (s, IH), 9.87 (s, lH), 11.47.
EXAMPLE 6 Preparation of:
N-(2-chloro-6-methylphenyl)-2-(6-(4-(3-hydroxyethyl)piperazin-l-yl)-2- methylpyrimidin-4-ylamino)thiazole-5-carboxamide
Figure imgf000055_0001
(IV)
To a slurry of (E)-N-(2-chloro-6-methylphenyl)-3-ethoxyacrylamide 5B (120 mg, 0.50 mmol) in THF (0.75 ml) and water (0.5 mL) was added NBS (98 mg, 0.55 mmol) at 0°C. The mixture was warmed and stirred at 20-22°C for 3h. To this was added l-(6-chloiO-2-methylpyrimidin-4-yl)thiourea 5A (100 mg, 0.49 mmol), and the slurry heated and stirred at reflux for 2h. The slurry was cooled to 20-22°C and the solid collected by vacuum filtration giving 140 mg (71% yield) of 2-(6-chloro-2- methylpyrimidin-4-ylamino)-N-(2-chloro-6-methylphenyl)thiazole-5-carboxamide 5D. 1H NMR (400 MHz, DMSO-d6) δ 2.23 (s, 3H), 2.58 (s, 3H), 6.94 (s, IH), 7.18- 7.34, ( m, 2H, J=7.5), 7.34-7.46 (d, IH, , J=7.5), 8.31 (s, IH), 10.02 (s, IH), 12.25 (s, IH).
Compound 5D was elaborated to N-(2-chloro-6-methylphenyl)-2-(6-(4-(3- hydroxyethyl)piperazin- 1 -yl)-2-methylpyrimidin-4-ylamino)thiazole-5-carboxamide, following Step 5E.
EXAMPLE 7
Preparation of: N-(2-chloro-6-methylphenyl)-2-(6-(4-(3-hydroxyethyl)piperazin-l-yl)-2- methylpyrimidin-4-ylamino)thiazole-5-carboxamide
7A. 2- [4-(6-Chloro-2-methyl-pyrimidin-4-yl)-piperazin-l-yl] -ethanol
Figure imgf000056_0001
(7A) (7B) (7C)
2-Piperazin-l-yl-ethanol (8.2g, 63.1 mmol) was added to a solution of 4,6-dichloro-2- methylpyrimidine (5.2g, 31.9 mmol) in dichloromethane (80 ml) at rt. The mixture was stirred for two hours and triethylamine (0.9 ml) was added. The mixture was stirred at rt for 20h. The resultant solid was filtered. The cake was washed with dichloromethane (20 ml). The filtrate was concentrated to give an oil. This oil was dried under high vacuum for 20h to give a solid. This solid was stirred with heptane (50 ml) at rt for 5h. Filtration gave 7C (8.13g) as a white solid
7B. Example 7
Figure imgf000056_0002
(5C) (IV)
To a 250 ml of round bottom flask were charged compound 5C (1.9g, 7.1 mmol), compound 7C (1.5 g, 5.9 mmol), K2CO3 (16g, 115.7 mmol), Pd (OAc)2 (52 mg, 0.23 mmol) and BINAP (291 mg, 0.46 mmol). The flask was placed under vacuum and flushed with nitrogen. Toluene was added (60 ml). The suspension was heated to 100-110°C and stirred at this temperature for 20h. After cooling to room temperature, the mixture was applied to a silica gel column. The column was first eluted with EtOAC, and then with 10% of MeOH in EtOAC. Finally, the column was washed with 10% 2M ammonia solution in MeOH/90% EtOAC. The fractions which contained the desired product were collected and concentrated to give compound IN as a yellow solid (2.3 g).
ANALYTICAL METHODS
Solid State Nuclear Magnetic Resonance (SSNMR)
All solid-state C-13 NMR measurements were made with a Bruker DSX-400, 400 MHz NMR spectrometer. High resolution spectra were obtained using high-power proton decoupling and the TPPM pulse sequence and ramp amplitude cross- polarization (RAMP-CP) with magic-angle spinning (MAS) at approximately 12 kHz (A.E. Bennett et al, J. Chem. Phys.,1995, 103, 6951),(G. Metz, X. Wu and S.O. Smith, J. Magn. Reson. A,. 1994, 110, 219-227). Approximately 70 mg of sample, packed into a canister-design zirconia rotor was used for each experiment. Chemical shifts (δ) were referenced to external adamantane with the high frequency resonance being set to 38.56 ppm (W.L. Earl and D.L. NanderHart, J. Magn. Reson., 1982, 48, 35-54).
x-Ray Powder Diffraction
One of ordinary skill in the art will appreciate that an X-ray diffraction pattern may be obtained with a measurement error that is dependent upon the measurement conditions employed. Hi particular, it is generally known that intensities in a X-ray diffraction pattern may fluctuate depending upon measurement conditions employed. It should be further understood that relative intensities may also vary depending upon experimental conditions and, accordingly, the exact order of intensity should not be taken into account. Additionally, a measurement error of diffraction angle for a conventional X-ray diffraction pattern is typically about 5% or less, and such degree of measurement error should be taken into account as pertaining to the aforementioned diffraction angles. Consequently, it is to be understood that the crystal forms of the instant invention are not limited to the crystal forms that provide X-ray diffraction patterns completely identical to the X-ray diffraction patterns depicted in the accompanying Figures disclosed herein. Any crystal forms that provide X- ray diffraction patterns substantially identical to those disclosed in the accompanying Figures fall within the scope of the present invention. The ability to ascertain substantial identities of X-ray diffraction patterns is within the purview of one of ordinary skill in the art.
X-Ray powder diffraction data for the crystalline forms of Compound (IN) were obtained using a Bruker GADDS (BRUKER AXS, Inc., 5465 East Cheryl Parkway Madison, WI 53711 USA) (General Area Detector Diffraction System) manual chi platform goniometer. Powder samples were placed in thin walled glass capillaries of 1mm or less in diameter; the capillary was rotated during data collection. The sample-detector distance was 17 cm. The radiation was Cu Kα (45kN 111mA, λ = 1.5418 A). Data were collected for 3<2Θ <35° with a sample exposure time of at least 300 seconds.
Single Crystal X-Ray
All single crystal data were collected on a Bruker-Νonius (BRUKER AXS, Inc., 5465 East Cheryl Parkway Madison, WI 53711 USA) Kappa CCD 2000 system using Cu Kα radiation (λ = 1.5418 A) and were corrected only for the Lorentz- polarization factors. Indexing and processing of the measured intensity data were carried out with the HKL2000 software package (Otwinowski, Z. & Minor, W. (1997) in Macromolecular Gystallography, eds. Carter, W.C. Jr & Sweet, R.M. (Academic, ΝY), Nol. 276, pp.307-326) in the Collect program suite (Data collection and processing user interface: Collect: Data collection software, R. Hooft, Nonius B.N., 1998).
The structures were solved by direct methods and refined on the basis of observed reflections using either the SDP (SDP, Structure Determination Package,Enraf-Νonius, Bohemia NY 11716 Scattering factors, including /and f, in the SDP software were taken from the" International Tables for Crystallography", Kynoch Press, Birmingham, England, 1974; Nol TV, Tables 2.2A and 2.3.1) software package with minor local modifications or the crystallographic package, MAXUS (maXus solution and refinement software suite: S. Mackay, C.J. Gilmore, C. Edwards, M. Tremayne, Ν. Stewart, K. Shankland. maXus: a computer program for the solution and refinement of crystal structures from diffraction data).
The derived atomic parameters (coordinates and temperature factors) were refined through full matrix least-squares. The function minimized in the refinements was ΣW(|F0| - |FC|)2- R is defined as Σ ||F0| - |Fp||/Σ |F0| while Rw = [∑w( |F0| - |FC|)2W |F0|2]1/2 where w is an appropriate weighting function based on errors in the observed intensities. Difference maps were examined at all stages of refinement. Hydrogens were introduced in idealized positions with isotropic temperature factors, but no hydrogen parameters were varied.
The derived atomic parameters (coordinates and temperature factors) were refined through full matrix least-squares. The function minimized in the refinements was ΣW(|F0| - |FC|)2- R is defined as Σ ||F0| - |FC||/Σ |F0| while Rw = [∑w( |F0| - |FC|)2W
|F0|2]1/2 where w is an appropriate weighting function based on errors in the observed intensities. Difference maps were examined at all stages of refinement. Hydrogens were introduced in idealized positions with isotropic temperature factors, but no hydrogen parameters were varied
Differential Scanning Calorimetry
The DSC instrument used to test the crystalline forms was a TA Instruments® model Q1000. The DSC cell/sample chamber was purged with 100 ml/min of ultra- high purity nitrogen gas. The instrument was calibrated with high purity indium. The accuracy of the measured sample temperature with this method is within about +/- 1°C, and the heat of fusion can be measured within a relative error of about +/-5%. The sample was placed into an open aluminum DSC pan and measured against an empty reference pan. At least 2 mg of sample powder was placed into the bottom of the pan and lightly tapped down to ensure good contact with the pan. The weight of the sample was measured accurately and recorded to a hundredth of a milligram. The instrument was programmed to heat at 10°C per minute in the temperature range between 25 and 350°C.
The heat flow, which was normalized by a sample weight, was plotted versus the measured sample temperature. The data were reported in units of watts/gram ("W/g"). The plot was made with the endothermic peaks pointing down. The endothermic melt peak was evaluated for extrapolated onset temperature, peak temperature, and heat of fusion in this analysis.
Thermogravimetric Analysis (TGA)
The TGA instrument used to test the crystalline forms was a TAHistruments® model Q500. Samples of at least 10 milligrams were analyzed at a heating rate of 10°C per minute in the temperature range between 25°C and about 350°C.
EXAMPLE 8
Preparation of: crystalline monohydrate ofN-(2-chloro-6-methylphenyl)-2-(6-(4-(3- hydroxyethyl)piperazin-l-yl)-2-methylpyrimidin-4-ylamino)thiazole-5-carboxamide
(IV)
An example of the crystallization procedure to obtain the crystalline monohydrate form is shown here:
Charge 48- g of the compound of formula (IV).
Charge approximately 1056 mL (22 mL/g) of ethyl alcohol, or other suitable alcohol. Charge approximately 144 mL of water.
Dissolve the suspension by heating to approximately 75 °C.
Optional: Polish filter by transfer the compound of formula (TV) solution at 75 °C through the preheated filter and into the receiver.
Rinse the dissolution reactor and transfer lines with a mixture of 43 mL of ethanol and 5 mL of water.
Heat the contents in the receiver to 75 - 80 °C and maintain 75 - 80 °C to achieve complete dissolution. Charge approximately 384 mL of water at a rate such that the batch temperature is maintained between 75-80 °C.
Cool to 75 °C, and, optionally, charge monohydrate seed crystals. Seed crystals are not essential to obtaining monohydrate, but provide better control of the crystallization.
Cool to 70 °C and maintain 70 °C for ca. 1 h.
Cool from 70 to 5 C over 2 h, and maintain the temperature between 0 at 5 °C for at least 2 h.
Filter the crystal slurry. Wash the filter cake with a mixture of 96 mL of ethanol and 96 mL of water.
Dry the material at < 50 °C under reduced pressure until the water content is 3.4 to 4.1% by KF to afford 41 g (85 M%).
Alternately, the monohydrate can be obtained by: 1) An aqueous solution of the acetate salt of compound TV was seeded with monohydrate and heated at 80 °C to give bulk monohydrate.
2) An aqueous solution of the acetate salt of compound TV was seeded with monohydrate. On standing several days at room temperature, bulk monohydrate had formed.
3) An aqueous suspension of compound TV was seeded with monohydrate and heated at 70 °C for 4 hours to give bulk monohydrate. Hi the absence of seeding, an aqueous slurry of compound IN was unchanged after 82 days at room temperature.
4) A solution of compound IN in a solvent such as ΝMP or DMA was treated with water until the solution became cloudy and was held at 75-85 °C for several hours. Monohydrate was isolated after cooling and filtering. 5) A solution of compound TV in ethanol, butanol, and water was heated. Seeds of monohydrate were added to the hot solution and then cooled. Monohydrate was isolated upon cooling and filtration.
One of ordinary skill in the art will appreciate that the monohydrate of the compound of formula (IV) may be represented by the XRPD as shown in Figure 1 or by a representative sampling of peaks as shown in Table 1.
Representative peaks taken from the XRPD of the monohydrate of the compound of formula (TV) are shown in Table 1.
Table 1. 2-Theta d(A) Height
17.994 4.9257 915
18.440 4.8075 338 19.153 4.6301 644
19.599 4.5258 361
21.252 4.1774 148
24.462 3.6359 250
25.901 3.4371 133 28.052 3.1782 153
The XRPD is also characterized by the following list comprising 2Θ values selected from the group consisting of: 4.6+ 0.2, 11.2± 0.2, 13.8± 0.2, 15.2± 0.2, 17.9± 0.2, 19. l± 0.2, 19.6+ 0.2, 23.2+ 0.2, 23.6± 0.2. The XRPD is also characterized by the list of 2Θ values selected from the group consisting of: 18.0± 0.2, 18.4± 0.2, 19.2± 0.2, 19.6± 0.2, 21.2± 0.2, 24.5+ 0.2, 25.9+ 0.2, and 28.0+ 0.2.
Single crystal x-ray data was obtained at room temperature (+25°C). The molecular structure was confirmed as a monohydrate form of the compound of Formula (TV). The following unit cell parameters were obtained for the monohydrate of the compound of formula (IN) from the x-ray analysis at 25°C: a(A) = 13.8632(7); b(A)= 9.3307(3); c(A) = 38.390(2); N(A3) 4965.9(4); Z' -= 1; Nm = 621
Space group Pbca
Molecules/unit cell 8
Density (calculated) (g/cm3) 1.354 Wherein Z' = number of drug molecules per asymmetric unit. Nm = N(unit cell) / (Z drug molecules per cell).
Single crystal x-ray data was also obtained at -50°C. The monohydrate form of the compound of Formula (IN) is characterized by unit cell parameters approximately equal to the following: Cell dimensions: a(A) = 13.862(1); b(A)= 9.286(1); c(A) = 38.143(2);
Volume = 4910(1) A3
Space group Pbca Molecules/unit cell 8
Density (calculated) (g/cm3) 1.300 wherein the compound is at a temperature of about -50°C. ,
The simulated XRPD was calculated from the refined atomic parameters at room temperature. The monohydrate of the compound of formula (TV) is represented by the DSC as shown in Figure 2. The DSC is characterized by a broad peak between approximately 95°C and 130°C. This peak is broad and variable and corresponds to the loss of one water of hydration as seen in the TGA graph. The DSC also has a characteristic peak at approximately 287°C which corresponds to the melt of the dehydrated form of the compound of formula (IN).
The TGA for the monohydrate of the compound of Formula (IN) is shown in
Figure 2 along with the DSC. The TGA shows a 3.48% weight loss from 50°C to
175°C. The weight loss corresponds to a loss of one water of hydration from the compound of Formula (IN). The monohydrate may also be prepared by crystallizing from alcoholic solvents, such as methanol, ethanol, propanol, i-propanol, butanol, pentanol, and water. EXAMPLE 9 Preparation of: crystalline n-butanol solvate ofN-(2-chloro-6-methylphenyl)-2-(6-(4-(3- hydrQxyethyl)piperazin-l-yl)-2-methylpyrimidin-4-ylamino)thiazole-5-carboxamide (IV)
The crystalline butanol solvate of the compound of formula (IN) is prepared by dissolving compound (TV) in 1 -butanol at reflux (116-118 °C) at a concentration of approximately lg/25 mL of solvent. Upon cooling, the butanol solvate crystallizes out of solution. Filter, wash with butanol, and dry.
The following unit cell parameters were obtained from the x-ray analysis for the crystalline butanol solvate, obtained at room temperature: a(A) = 22.8102(6); b(A)= 8.4691(3); c(A) = 15.1436(5); N(A3) 2910.5(2); Z' = 1; Nm = 728 Space group P2ι/a
Molecules/unit cell 4 Density (calculated) (g/cm3) 1.283 Wherein Z' = number of drug molecules per asymmetric unit. Nm = V(unit cell) / (Z drug molecules per cell). One of ordinary skill in the art will appreciate that the butanol solvate of the compound of formula (IN) may be represented by the XRPD as shown in Figure 3 or by a representative sampling of peaks. Representative peaks for the crystalline butanol solvate are 20 values of : 5.9 ± 0.2, 12.0 ± 0.2, 13.0 ± 0.2, 17.7 ± 0.2, 24.1 ± 0.2, and 24.6 ± 0.2.
Example 10 Preparation of: crystalline ethanol solvate ofN-(2-chloro-6-methylphenyl)-2-(6-(4-(3- hydroxyethyl)piperazin-l-yl)-2-methylpyrimidin-4-ylamino)thiazole-5-carboxamide (IV)
Figure imgf000065_0001
To a 100-mL round bottom flask was charged 4.00 g (10.1 mmol) of 5D (contained 2.3 Area% 5C) 6.60 g (50.7 mmol) of 7B, 80 mL of n-butanol and 2.61 g (20.2 mmol) of DIPEA. The resulting slurry was heated to 120 °C and maintained at 120 °C for 4.5 h whereby HPLC analysis showed 0.19 relative Area% of residual 5D to compound IN. The homogeneous mixture was cooled to 20 °C and left stirring overnight. The resulting crystals were filtered. The wet cake was washed twice with 10-mL portions of n-butanol to afford a white crystalline product. HPLC analysis showed this material to contain 99.7 Area% compound IN and 0.3 Area% 5C.
The resulting wet cake was returned to the 100-mL reactor, and charged with 56 mL (12 mL/g) of 200 proof ethanol. At 80 °C an additional 25 mL of ethanol was added. To this mixture was added 10 mL of water resulting in rapid dissolution. Heat was removed and crystallization was observed at 75 - 77 °C. The crystal slurry was further cooled to 20 °C and filtered. The wet cake was washed once with 10 mL of 1:1 ethanol : water and once with 10 mL of n-heptane. The wet cake contained 1.0% water by KF and 8.10% volatiles by LOD. The material was dried at 60 °C/30 in Hg for 17 h to afford 3.55 g (70 M%) of material containing only 0.19% water by KF, 99.87 Area% by HPLC. The 1H ΝMR spectrum, however revealed that the ethanol solvate had been formed.
The following unit cell parameters were obtained from the x-ray analysis for the crystalline ethanol solvate (di-ethanolate), obtained at -40°C: a(A) = 22.076(1); b(A)= 8.9612(2); c(A) = 16.8764(3); N(A3) 3031.1(1); Z' = 1; Nm = 758
Space group P2ι/a
Molecules/unit cell 4
Density (calculated) (g/cm3) 1.271 Wherein Z' = number of drug molecules per asymmetric unit. Nm = N(unit cell) / (Z drug molecules per cell).
One of ordinary skill in the art will appreciate that the ethanol solvate of the compound of formula (IN) may be represented by the XRPD as shown in Figure 4 or by a representative sampling of peaks. Representative peaks for the crystalline ethanol solvate are 2Θ values of : 5.8 ± 0.2, 11.3 ± 0.2, 15.8 ± 0.2, 17.2 ± 0.2, 19.5 ±
0.2, 24.1 ± 0.2, 25.3 ± 0.2, and 26.2 ± 0.2.
Example 11 Preparation of: crystalline N-(2-chloro-6-methylphenyl)-2-(6-(4-(3-hydroxyethyl)piperazin-l-yl)-2- methylpyrimidin-4-ylamino)thiazole-5-carboxamide (TV) (Neat form N-6)
To a mixture of compound 5D (175.45 g, 0.445 mol) and hydroxyethylpiperazine (289.67 g, 2.225 mol) in ΝMP (1168 mL) was added DIPEA (155 mL, 0.89 mol). The suspension was heated at 110°C (solution obtained) for 25 min., then cooled to about 90°C. The resulting hot solution was added dropwise into hot (80°C) water (8010) mL, keeping the temperature at about 80°C. The resulting suspension was stirred 15 min at 80°C then cooled slowly to room temperature. The solid was collected by vacuum filtration, washed with water (2x 1600 mL) and dried in vacuo at 55-60°C affording 192.45 g (88.7 % yield) of Ν-(2-chloro-6- methylphenyl)-2-(6-(4-(3-hydroxyethyl)piperazin-l-yl)-2-methylpyrimidin-4- ylamino)thiazole-5-carboxamide. 1H NMR (400 MHz, DMSO-d6): δ 2.24 (s, 3H), 2.41 (s, 3H), 2.43 (t, 2H, J=6), 2.49 (t, 4H, J=6.3), 3.51 (m, 4H), 3.54 (q, 2H, J=6), 4.46 (t, IH, J=5.3), 6.05 (s, IH), 7.26 (t, IH, J=7.6), 7.28 (dd, IH, J=7.6, 1.7), 7.41 (dd, IH, J=7.6, 1.7), 8.23 (s, IH), 9.89 (s, IH), 11.48. KF0.84; DSC: 285.25°C (onset), 286.28°C (max). The following unit cell parameters were obtained from the x-ray analysis for the neat crystalline compound IN, obtained at 23 °C: a(A) = 22.957(1); b(A)= 8.5830(5); c(A) = 13.803(3);
N(A3) = 2521.0(5); Z' = 1; Nm = 630 Space group P2ι/a
Molecules/unit cell 4
Density (calculated) (g/cm3) 1.286 Wherein Z' = number of drug molecules per asymmetric unit. Nm = N(unit cell) / (Z drug molecules per cell). One of ordinary skill in the art will appreciate that the crystalline form of the compound of formula (IV) may be represented by the XRPD as shown in Figure 5 or by a representative sampling of peaks. Representative peaks for the crystalline neat form (Ν-6) are 2Θ values of : 6.8 + 0.2, 11.1 ± 0.2, 12.3 ± 0.2, 13.2 ± 0.2, 13.7 ± 0.2, 16.7 ± 0.2, 21.0 ± 0.2, 24.3 ± 0.2, and 24.8 ± 0.2.
Example 12 Preparation of: crystalline N-(2-chloro-6-methylphenyl)-2-(6-(4-(3-hydroxyethyl)piperazin-l-yl)-2- methylpyrimidin-4-ylamino)thiazole-5-carboxamide (TV) (neat form T1H1-7)
' The title neat form may be prepared by heating the monohydrate form of the compound of formula (IV) above the dehydration temperature.
The following unit cell parameters were obtained from the x-ray analysis for the neat crystalline (T1H1-7) compound TV, obtained at 25 °C: a(A) = 13.4916; b(A)= 9.3992(2); c(A) = 38.817(1);
V(A3) = 4922.4(3); Z' = 1; Nm = 615 Space group Pbca Density (calculated) (g/cm3) 1.317 Wherein Z' = number of drug molecules per asymmetric unit. Nm = N(unit cell) / (Z drug molecules per cell). One of ordinary skill in the art will appreciate that the neat crystalline form
(T1H1-7) of the compound of formula (IN) may be represented by the XRPD as shown in Figure 6 or by a representative sampling of peaks. Representative peaks for the crystalline neat form (T1H1-7)) are 2Θ values of : 8.0 ± 0.2, 9.7 ± 0.2, 11.2 ± 0.2, 13.3 ± 0.2, 17.5 ± 0.2, 18.9 ± 0.2, 21.0 ± 0.2, 22.0 ± 0.2.
Obviously, numerous modifications and variations of the present invention are possible in light of the above teachings. It is therefore to be understood that within the scope of the appended claims, the invention may be practiced otherwise than as specifically described herein.

Claims

CLAIMSWhat we claim is:
1. A process for preparing a compound having the formula (I),
Figure imgf000069_0001
wherein L, Ar, R2, R3, R^ R5, and m are as defined below, comprising reacting a compound having the formula (ff),
Figure imgf000069_0002
wherein Q is the group -O-P*, wherein P* is selected so that, when considered together with the oxygen atom to which P* is attached, Q is a leaving group, and Ar, L, R2, R3, and m are as defined below, with a halogenating reagent followed by a thiourea compound having the formula (Iff),
Figure imgf000069_0003
wherein, R4 and R5 are as defined below, to provide the compound of formula (I),
Figure imgf000069_0004
wherein,
Ar is the same in formulae (1) and (ff) and is aryl or heteroaryl; L is the same in formulae (I) and (ff) and is optionally-substituted alkylene; R2 is the same in formulae (I) and (TT), and is selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo;
R3 is the same in formulae (I) and (TT), and is selected from hydrogen, halogen, cyano, haloalkyl, alkyl, substituted alkyl, alkenyl, substituted alkenyl, aryl, heteroaryl, cycloalkyl, and heterocyclo; Φ is (i) the same in each of formulae (I) and (Iff), and (ii) is independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo, or alternatively, R is taken together with R5, to form heteroaryl or heterocyclo;
R5 (i) is the same in each of formulae (I) and (Iff), and (ii) is independently selected from hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, aryl, heteroaryl, cycloalkyl, and heterocyclo, or alternatively, R5 is taken together with R j to form heteroaryl or heterocyclo; and m is O or 1.
2. The process of claim 1, comprising preparing a compound of the formula (le),
Figure imgf000070_0001
wherein Zi and Z5 are selected from hydrogen, alkyl, halogen; hydroxy, and alkoxy; Z2, Z3 and Z4 are selected from hydrogen, alkyl, halogen; hydroxy, alkoxy, C(=O)NR8, and/or NR8C(=O), wherein R8 is alkyl, cycloalkyl, or heteroaryl; comprising reacting a compound having the formula,
Figure imgf000071_0001
wherein Q is as defined in claim 1, and Zi, Z2, Z3, Z4, and Z5 are as defined above, with a halogenating reagent followed by a thiourea compound having the formula,
Figure imgf000071_0002
to provide the compound having the formula,
Figure imgf000071_0003
The process of claim 2, wherein R4 is hydrogen, to provide the compound having the formula (If),
Figure imgf000071_0004
4. The process of claim 3, further comprising: reacting the compound of the formula
Figure imgf000071_0005
with a pyrimidine compound having the formula,
Figure imgf000072_0001
4a, wherein X and Y are leaving groups, and R15 and Rig are independently selected from hydrogen, alkyl and substituted alkyl, to provide a compound having the formula,
Figure imgf000072_0002
wherein Y, R15, Rι6, Zi, Z , Z3, Z4, and Z5 are as defined in claim 2.
5. The process of claim 4, further comprising reacting the compound having the formula,
Figure imgf000072_0003
with an amine having the formula NHR oR2ι, wherein R20 and R ι are independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R20 and R ι can be taken together to form a heterocyclo, to provide a compound having the formula (Hi),
Figure imgf000072_0004
wherein R15, Rι6, Zi, Z2, Z3, Z , Z5, R2o and R2ι are as defined above.
6. The process of claim 5 wherein the amine NHR2oR2ι is piperazine in turn optionally substituted with hydroxy (alkyl).
7. The process of claim 1, comprising preparing the compound having the formula,
Figure imgf000073_0001
8. The process of claim 3, further comprising: reacting the compound of the formula (If),
Figure imgf000073_0002
wherein Zi, Z2, Z3, Z4, and Z5 are as defined in claim 2, with a pyrimidine compound having the formula,
Figure imgf000073_0003
wherein R15 and Rι6 are independently selected from hydrogen, alkyl and substituted alkyl, and R2o and R2ι are independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R20 and R2ι can be taken together to form a heterocyclo; to provide a compound having the formula (Hi),
Figure imgf000073_0004
wherein R15, Rι6, Zi, Z2, Z3, Z4, Z5, R20 and R21 are as defined above.
9. The process of claim 2, wherein R is a group having the formula,
Figure imgf000074_0001
wherein R15 and Rι are independently selected from hydrogen, alkyl and substituted alkyl, and R2o and R2ι are independently selected from hydrogen, alkyl, substituted alkyl, cycloalkyl, heterocyclo, aryl, and heteroaryl, or R20 and R2ι can be taken together to form a heterocyclo; whereby said process provides a compound having the formula (Hi),
Figure imgf000074_0002
wherein R15, Rι6, Zi, Z2, Z3, Z , Z5, R20 and R2ι are as defined above.
10. The process of claim 2, wherein R4 is a group having the formula,
Figure imgf000074_0003
wherein Y is a leaving group and R15 and Rι6 are independently selected from hydrogen, alkyl and substituted alkyl, wherein said process provides a compound having the formula (H),
Figure imgf000074_0004
wherein Y, R15, Ri6, Zi, Z2, Z3, Z4, and Z5 are as defined above.
11. The process of claim 10, further comprising reacting said compound of formula (H) with an amine having the formula NHR20R21, to provide a compound having the formula (Hi)
Figure imgf000075_0001
wherein R15, Rι6, Zi, Z2, Z3, Z , Z5, R2o and R21 are as defined in claim 10.
12. The process of claim 2, wherein R4 is a group having the formula,
"' 1 or ""V1 HsC "
13. The process of claim 1, in which Ar is optionally-substituted phenyl.
14. The process of claim 1, in which Ar is selected from,
Figure imgf000075_0002
15. The process of claim 1 , in which L is optionally-substituted alkylene and m is 1.
16. The process of claim 1, in which m is 0.
17. The process of claim 1, in which, R2 is hydrogen or lower alkyl; R3 is hydrogen or lower alkyl; and R5 is hydrogen.
18. The process of claim 1 in which the halogenating agent is selected from NBS, l,3-dichloro-5,5-dimethylhydantoin, l,3-dibromo-5,5-dimethylhydantoin, and 1,3- diiodo-5,5-dimethylhydantoin.
19. An intermediate useful in preparing compounds useful as kinase inhibitors, having the formula,
Figure imgf000076_0001
wherein Ris is Ci_4alkyl;
Zi and Z5 are selected from hydrogen, lower alkyl, and halogen; and Z is hydrogen, or -C(=O)NR8, wherein R8 is alkyl, cycloalkyl, or heteroaryl.
20. Crystalline monohydrate of the compound of formula (TV)
Figure imgf000076_0002
(IV).
21. The compound of Claim 20, which is characterized by an x-ray powder diffraction pattern substantially in accordance with that shown in Figure 1.
22. The compound of Claim 20, which is characterized by differential scanning calorimetry thermogram and a thermogravimetric anaylsis substantially in accordance with that shown in Figure 2.
23. The compound of Claim 20, which is characterized by an x-ray powder diffraction pattern (CuKα λ=1.5418A at a temperature of about 23 °C) comprising four or more 2Θ values selected from the group consisting of: 18.0± 0.2, 18.4± 0.2, 19.2± 0.2, 19.6± 0.2, 21.2± 0.2, 24.5± 0.2, 25.9+ 0.2, and 28.0± 0.2.
24. A pharmaceutical composition comprising a therapeutically effective amount of the compound of Claim 20 and a pharmaceutically acceptable carrier.
25. A method for the treatment of cancer which comprises administering to a host in need of such treatment a therapeutically effective amount of a compound of Claim 20.
26. The compound of claim 20, characterized by unit cell parameters approximately equal to the following:
Cell dimensions: a(A) = 13.8632(7); < b(A)= 9.3307(3); c(A) = 38.390(2);
Volume = 4965.9(4) A3
Space group Pbca
Molecules/unit cell 8 Density (calculated) (g/cm3) 1.354.
27. The compound of claim 20, wherein there is one water molecule per molecule of formula (TV).
28. A method of treating oncological disorders which comprises administering to a host in need of such treatment a therapeutically effective amount of a compound of Claim 20, wherein the disorders are selected from chronic myelogenous leukemia (CML), gastrointestinal stromal tumor (GIST), small cell lung cancer (SCLC), non- small cell lung cancer (NSCLC), ovarian cancer, melanoma, mastocytosis, germ cell tumors, acute myelogenous leukemia (AML), pediatric sarcomas, breast cancer, colorectal cancer, pancreatic cancer, and prostate cancer.
29. Crystalline butanol solvate of the compound of formula (IV)
Figure imgf000078_0001
(IV).
30. The compound of claim 29, characterized by unit cell parameters approximately equal to the following:
Cell dimensions: a(A) = 22.8102(6); b(A)= 8.4691(3); c(A) = 15.1436(5); Volume = 2910.5(2) A3 Space group P2ι/a Molecules/unit cell: 4 Density (calculated) (g/cm3): 1.283.
31. A process for preparing crystalline monohydrate of the compound of formula (TV)
Figure imgf000078_0002
(TV) comprising heating and dissolving the compound of formula (TV) in an ethanol/water mixture and crystallizing the monohydrate from the ethanol/water mixture as it cools.
32. The process of claim 31, wherein the butanol solvate of the compound of formula (IN) is dissolved in the ethanol/water mixture.
PCT/US2005/003728 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors WO2005077945A2 (en)

Priority Applications (20)

Application Number Priority Date Filing Date Title
KR1020067015839A KR101310427B1 (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
PL05722772T PL1711481T5 (en) 2004-02-06 2005-02-04 Crystalline monohydrate as kinase inhibitors
ES05722772T ES2337272T5 (en) 2004-02-06 2005-02-04 Crystalline monohydrate as a kinase inhibitor
AU2005212405A AU2005212405B2 (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
DK05722772.0T DK1711481T4 (en) 2004-02-06 2005-02-04 Crystalline monohydrate as kinase inhibitors
CA2555291A CA2555291C (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
JP2006552303A JP5148115B2 (en) 2004-02-06 2005-02-04 Method for producing kinase inhibitor 2-aminothiazole-5-aromatic carboxamide compound
EP05722772.0A EP1711481B2 (en) 2004-02-06 2005-02-04 Crystalline monohydrate as kinase inhibitors
IN6567DEN2014 IN2014DN06567A (en) 2004-02-06 2005-02-04
AT05722772T ATE453630T1 (en) 2004-02-06 2005-02-04 METHOD FOR PRODUCING 2-AMINOTHIAZOLE-5-AROMATIC CARBOXYLIC ACID AMIDES AS KINASE INHIBITORS
DE602005018601T DE602005018601D1 (en) 2004-02-06 2005-02-04 PROCESS FOR THE PREPARATION OF 2-AMINOTHIAZOL-5-ARO
BRPI0507476A BRPI0507476B8 (en) 2004-02-06 2005-02-04 crystalline monohydrate, its pharmaceutical composition and its use
SI200530905T SI1711481T2 (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
NZ548613A NZ548613A (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
CN2005800119166A CN1980909B (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
ZA2006/06242A ZA200606242B (en) 2004-02-06 2006-07-27 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
IL177280A IL177280A (en) 2004-02-06 2006-08-03 Crystalline monohydrate of a 2-aminothiazole-5-aromatic carboxamide, process for preparing same, pharmaceutical composition comprising same and use thereof for preparation of a medicament
NO20063780A NO338049B1 (en) 2004-02-06 2006-08-24 Crystalline monohydrate of 2-aminothiazole-5-aromatic carboxamide, preparation thereof, pharmaceutical composition comprising it and its use in the treatment of cancer
HK06112879.5A HK1091835A1 (en) 2004-02-06 2006-11-23 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
HRP20100166TT HRP20100166T4 (en) 2004-02-06 2010-03-22 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US54249004P 2004-02-06 2004-02-06
US60/542,490 2004-02-06
US62493704P 2004-11-04 2004-11-04
US60/624,937 2004-11-04

Publications (2)

Publication Number Publication Date
WO2005077945A2 true WO2005077945A2 (en) 2005-08-25
WO2005077945A3 WO2005077945A3 (en) 2006-05-18

Family

ID=34864498

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2005/003728 WO2005077945A2 (en) 2004-02-06 2005-02-04 Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors

Country Status (27)

Country Link
EP (1) EP1711481B2 (en)
JP (2) JP5148115B2 (en)
KR (2) KR101310427B1 (en)
AR (1) AR047533A1 (en)
AT (1) ATE453630T1 (en)
AU (1) AU2005212405B2 (en)
BR (1) BRPI0507476B8 (en)
CA (1) CA2555291C (en)
CY (1) CY1109907T1 (en)
DE (1) DE602005018601D1 (en)
DK (1) DK1711481T4 (en)
ES (1) ES2337272T5 (en)
GE (1) GEP20094804B (en)
HK (1) HK1091835A1 (en)
HR (1) HRP20100166T4 (en)
IL (1) IL177280A (en)
IN (1) IN2014DN06567A (en)
NO (1) NO338049B1 (en)
NZ (1) NZ548613A (en)
PE (1) PE20050691A1 (en)
PL (1) PL1711481T5 (en)
PT (1) PT1711481E (en)
RU (1) RU2382039C2 (en)
SI (1) SI1711481T2 (en)
TW (1) TWI338004B (en)
WO (1) WO2005077945A2 (en)
ZA (1) ZA200606242B (en)

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007019210A1 (en) * 2005-08-05 2007-02-15 Bristol-Myers Squibb Company Preparation of 2-amino-thiazole-5-carboxylic-acid derivatives
WO2006121742A3 (en) * 2005-05-05 2007-02-22 Bristol Myers Squibb Co Formulations of a src/abl inhibitor
WO2007035874A1 (en) * 2005-09-21 2007-03-29 Bristol-Myers Squibb Company Oral administration of n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-1,3-thiazole-5-carboxamide and salts thereof
WO2007106879A2 (en) * 2006-03-15 2007-09-20 Bristol-Myers Squibb Company Process for preparing n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamide and related metabolites thereof
WO2008044045A1 (en) 2006-10-12 2008-04-17 Astex Therapeutics Limited Pharmaceutical combinations
WO2008044041A1 (en) 2006-10-12 2008-04-17 Astex Therapeutics Limited Pharmaceutical combinations
US7390810B2 (en) 2003-05-01 2008-06-24 Bristol-Myers Squibb Company Pyrazole-amine compounds useful as kinase inhibitors
WO2008142031A1 (en) 2007-05-18 2008-11-27 Institut Curie P38alpha as a therapeutic target in bladder carcinoma
US7534881B2 (en) 2004-06-30 2009-05-19 Bristol-Myers Squibb Company Method for preparing pyrrolotriazine compounds
US7652146B2 (en) 2004-02-06 2010-01-26 Bristol-Myers Squibb Company Process for preparing 2-aminothiazole-5-carboxamides useful as kinase inhibitors
WO2010062715A2 (en) * 2008-11-03 2010-06-03 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010067374A2 (en) * 2008-12-08 2010-06-17 Hetero Research Foundation Polymorphs of dasatinib
US7973045B2 (en) 2007-10-23 2011-07-05 Teva Pharmaceutical Industries Ltd. Anhydrous form of dasatinib and process for preparation thereof
WO2011095588A1 (en) 2010-02-04 2011-08-11 Ratiopharm Gmbh Pharmaceutical composition comprising n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamid
CN102250084A (en) * 2010-02-08 2011-11-23 南京卡文迪许生物工程技术有限公司 Dasatinib polymorphic substance as well as preparation method and pharmaceutical composition thereof
US8119649B2 (en) 2001-08-10 2012-02-21 Novartis Ag Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
CN102827156A (en) * 2012-09-11 2012-12-19 湖南欧亚生物有限公司 Novel industrial synthetic method of dasatinib
US8426419B2 (en) 2006-09-11 2013-04-23 Curis, Inc. Tyrosine kinase inhibitors containing a zinc binding moiety
WO2013065063A1 (en) 2011-11-03 2013-05-10 Cadila Healthcare Limited Anhydrous form of dasatinib, process for its preparation and its use
CN103319476A (en) * 2013-06-13 2013-09-25 济南德爱医药技术有限公司 Kinase inhibitor
CN103483289A (en) * 2013-09-06 2014-01-01 浙江科源化工有限公司 2-amino-N-(2-chloro-6-methyl phenyl)thiazole-5-carboxamide synthesis method
CN103819469A (en) * 2012-11-16 2014-05-28 重庆医药工业研究院有限责任公司 Crystal form of dasatinib and preparation method for crystal form of dasatinib
WO2014086326A1 (en) 2012-12-06 2014-06-12 Zentiva, K.S. A method for the preparation and purification of new and known polymorphs and solvates of dasatinib
CN103880833A (en) * 2012-12-19 2014-06-25 北京本草天源药物研究院 Novel crystal form of dasatinib monohydrate, and preparation method and pharmaceutical composition thereof
WO2014102759A2 (en) 2012-12-31 2014-07-03 Ranbaxy Laboratories Limited Process for the preparation of dasatinib and its intermediates
CN104341410A (en) * 2013-08-09 2015-02-11 上海科胜药物研发有限公司 New Dasatinib crystal form and preparation method thereof
WO2015090259A1 (en) * 2013-12-19 2015-06-25 Zentiva, K.S. Method of preparing anhydrous polymorphic form n-6 of dasatinib
WO2015107545A1 (en) 2013-12-18 2015-07-23 Dharmesh Mahendrabhai Shah Water soluble salts of dasatinib hydrate
US9115124B1 (en) 2014-03-11 2015-08-25 Cerbios-Pharma Sa Process and intermediates for the preparation of dasatinib
WO2015181573A1 (en) 2014-05-26 2015-12-03 Egis Gyógyszergyár Zrt. Dasatinib salts
EP2861589A4 (en) * 2012-06-15 2016-03-02 Basf Se Multicomponent crystals comprising dasatinib and selected cocrystal formers
CN105503854A (en) * 2015-12-31 2016-04-20 哈药集团技术中心 New crystal form substance of Dasatinib anhydrous substance and preparation method thereof
CN106117195A (en) * 2016-06-09 2016-11-16 青岛辰达生物科技有限公司 A kind of synthetic method for treating leukemic medicine Dasatinib
WO2017002131A1 (en) * 2015-06-29 2017-01-05 Msn Laboratories Private Limited Crystalline forms of n-(2-chloro-6-methy]phenvn-2-[f6-[4-(2-hvdroxvethvl)-l- piperazinvil-2-methvl-4-pvrimidinvllaminol-5-thiazolecarboxamide and their process thereof
WO2017098391A1 (en) * 2015-12-11 2017-06-15 Shilpa Medicare Limited Process for the preparation of dasatinib
WO2017108605A1 (en) 2015-12-22 2017-06-29 Synthon B.V. Pharmaceutical composition comprising amorphous dasatinib
WO2017144109A1 (en) 2016-02-25 2017-08-31 Remedica Ltd Dasatinib formulation
WO2017199014A1 (en) 2016-05-16 2017-11-23 University Of Dundee Treatment of opioid tolerance
US9957232B2 (en) 2006-10-11 2018-05-01 Bayer Healthcare Llc 4-[4-({[4-chloro-3-(trifluoromethyl)phenyl]carbamoyl}amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
IT201700006157A1 (en) * 2017-01-20 2018-07-20 Cerbios Pharma Sa Co-crystals of an antitumor compound
IT201700006145A1 (en) * 2017-01-20 2018-07-20 Cerbios Pharma Sa Co-crystal of an antitumor compound
CN109369638A (en) * 2018-11-21 2019-02-22 山东罗欣药业集团股份有限公司 A kind of preparation process of Dasatinib
CN109503568A (en) * 2018-12-29 2019-03-22 山东罗欣药业集团股份有限公司 A kind of preparation method of Dasatinib
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10464933B2 (en) 2016-02-03 2019-11-05 Dr. Reddy's Laboratories Limited Solid state forms of dasatinib and processes for their preparation
CN111108104A (en) * 2017-07-07 2020-05-05 拜康有限公司 Polymorphic forms of dasatinib
WO2021032129A1 (en) * 2019-08-20 2021-02-25 湖南华纳大药厂股份有限公司 Kinase inhibitor, preparation therefor, pharmaceutical composition thereof and use thereof
US10940149B1 (en) 2018-06-15 2021-03-09 Handa Oncology, Llc Kinase inhibitor salts and compositions thereof
WO2021150981A1 (en) 2020-01-24 2021-07-29 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
WO2021176083A1 (en) 2020-03-06 2021-09-10 Zentiva K.S. Pharmaceutical compositions comprising dasatinib anhydrous and uses thereof
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
CN114634421A (en) * 2022-02-24 2022-06-17 济宁晟泰药业有限公司 Preparation method of dasatinib intermediate
EP4071248A1 (en) 2021-04-07 2022-10-12 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Means and methods for enhancing receptor-targeted gene transfer
US11980619B2 (en) 2022-07-28 2024-05-14 Nanocopoeia, Llc Pharmaceutical compositions and crushable tablets including amorphous solid dispersions of dasatinib and uses

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI338004B (en) * 2004-02-06 2011-03-01 Bristol Myers Squibb Co Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
WO2010129208A1 (en) * 2009-05-05 2010-11-11 Merck Sharp & Dohme Corp. P38 kinase inhibiting agents
CN101812060B (en) * 2010-02-02 2011-08-17 南京卡文迪许生物工程技术有限公司 Simple novel method for preparing high-purity Sprycel, and intermediate compound
JP5589097B2 (en) * 2010-02-08 2014-09-10 南京▲か▼文迪許生物工程技術有限公司 Dasatinib polycrystal, preparation method thereof and drug composition
EP3024831A2 (en) * 2013-07-25 2016-06-01 Basf Se Salts of dasatinib in amorphous form
JP6501773B2 (en) * 2013-07-25 2019-04-17 ビーエーエスエフ ソシエタス・ヨーロピアBasf Se Salt of dasatinib in crystalline form
RU2567537C1 (en) * 2014-04-23 2015-11-10 Олег Ростиславович Михайлов Crystal anhydrous delta modification of n-(2-chlorine-6-methylphenyldiazomethane)-2-[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pirimidinyl]amino]-5-thiazolcarboxamide, method of its production and pharmaceutical composition based on it
CN105474831B (en) * 2015-12-16 2017-11-17 湖南省西瓜甜瓜研究所 A kind of bletilla striata naked seed sterilization method
CN105830582B (en) * 2016-04-15 2018-05-01 成都大学 A kind of the quick of bletilla seed returns native breeding method
JP6597727B2 (en) 2017-05-11 2019-10-30 東洋インキScホールディングス株式会社 Surface protective adhesive and adhesive sheet

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3547917A (en) * 1966-12-07 1970-12-15 Uniroyal Inc 2-amino-4-methylthiazole-5-carboxamides
EP0639574A1 (en) * 1993-08-16 1995-02-22 Lucky Ltd. 2-aminothiazolecarboxamide derivatives, processes for their preparation and their use for controlling phytopathogenic organisms
US6596746B1 (en) * 1999-04-15 2003-07-22 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
WO2004071440A2 (en) * 2003-02-06 2004-08-26 Bristol-Myers Squibb Company Thiazolyl-based compounds useful as kinase inhibitors
WO2005072826A2 (en) * 2004-01-21 2005-08-11 Emory University Compositions and use of tyrosine kinase inhibitors to treat pathogenic infection

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI338004B (en) * 2004-02-06 2011-03-01 Bristol Myers Squibb Co Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3547917A (en) * 1966-12-07 1970-12-15 Uniroyal Inc 2-amino-4-methylthiazole-5-carboxamides
EP0639574A1 (en) * 1993-08-16 1995-02-22 Lucky Ltd. 2-aminothiazolecarboxamide derivatives, processes for their preparation and their use for controlling phytopathogenic organisms
US6596746B1 (en) * 1999-04-15 2003-07-22 Bristol-Myers Squibb Company Cyclic protein tyrosine kinase inhibitors
WO2004071440A2 (en) * 2003-02-06 2004-08-26 Bristol-Myers Squibb Company Thiazolyl-based compounds useful as kinase inhibitors
WO2005072826A2 (en) * 2004-01-21 2005-08-11 Emory University Compositions and use of tyrosine kinase inhibitors to treat pathogenic infection

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
SHAH NEIL P ET AL: "Overriding imatinib resistance with a novel ABL kinase inhibitor." SCIENCE. 16 JUL 2004, vol. 305, no. 5682, 16 July 2004 (2004-07-16), pages 399-401, XP002361026 ISSN: 1095-9203 *
ZHAO R ET AL: "A new facile synthesis of 2-aminothiazole-5-carboxylates" TETRAHEDRON LETTERS, ELSEVIER SCIENCE PUBLISHERS, AMSTERDAM, NL, vol. 42, no. 11, 11 March 2001 (2001-03-11), pages 2101-2102, XP004229211 ISSN: 0040-4039 *

Cited By (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8268837B2 (en) 2001-08-10 2012-09-18 Novartis Ag Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
US8119649B2 (en) 2001-08-10 2012-02-21 Novartis Ag Use of c-Src inhibitors alone or in combination with STI571 for the treatment of leukaemia
US7390810B2 (en) 2003-05-01 2008-06-24 Bristol-Myers Squibb Company Pyrazole-amine compounds useful as kinase inhibitors
US7414056B2 (en) 2003-05-01 2008-08-19 Bristol-Myers Squibb Company Pyrazole-amide compounds useful as kinase inhibitors
US7652146B2 (en) 2004-02-06 2010-01-26 Bristol-Myers Squibb Company Process for preparing 2-aminothiazole-5-carboxamides useful as kinase inhibitors
US7534881B2 (en) 2004-06-30 2009-05-19 Bristol-Myers Squibb Company Method for preparing pyrrolotriazine compounds
WO2006121742A3 (en) * 2005-05-05 2007-02-22 Bristol Myers Squibb Co Formulations of a src/abl inhibitor
EP1885339B1 (en) 2005-05-05 2015-07-29 Bristol-Myers Squibb Holdings Ireland Formulations of a src/abl inhibitor
NO343939B1 (en) * 2005-05-05 2019-07-15 Bristol Myers Squibb Holdings Ireland Formulations of an src / abl inhibitor
WO2007019210A1 (en) * 2005-08-05 2007-02-15 Bristol-Myers Squibb Company Preparation of 2-amino-thiazole-5-carboxylic-acid derivatives
US7932386B2 (en) 2005-08-05 2011-04-26 Bristol-Myers Squibb Company Preparation of 2-amino-thiazole-5-carboxylic-acid derivatives
WO2007035874A1 (en) * 2005-09-21 2007-03-29 Bristol-Myers Squibb Company Oral administration of n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-1,3-thiazole-5-carboxamide and salts thereof
WO2007106879A3 (en) * 2006-03-15 2007-11-01 Bristol Myers Squibb Co Process for preparing n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamide and related metabolites thereof
WO2007106879A2 (en) * 2006-03-15 2007-09-20 Bristol-Myers Squibb Company Process for preparing n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamide and related metabolites thereof
US8426419B2 (en) 2006-09-11 2013-04-23 Curis, Inc. Tyrosine kinase inhibitors containing a zinc binding moiety
US9957232B2 (en) 2006-10-11 2018-05-01 Bayer Healthcare Llc 4-[4-({[4-chloro-3-(trifluoromethyl)phenyl]carbamoyl}amino)-3-fluorophenoxy]-N-methylpyridine-2-carboxamide monohydrate
WO2008044041A1 (en) 2006-10-12 2008-04-17 Astex Therapeutics Limited Pharmaceutical combinations
WO2008044045A1 (en) 2006-10-12 2008-04-17 Astex Therapeutics Limited Pharmaceutical combinations
WO2008142031A1 (en) 2007-05-18 2008-11-27 Institut Curie P38alpha as a therapeutic target in bladder carcinoma
EP2508523A2 (en) 2007-10-23 2012-10-10 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
US8067423B2 (en) 2007-10-23 2011-11-29 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib isopropyl alcohol and process for preparation thereof
US7973045B2 (en) 2007-10-23 2011-07-05 Teva Pharmaceutical Industries Ltd. Anhydrous form of dasatinib and process for preparation thereof
EP2537847A1 (en) 2007-10-23 2012-12-26 Teva Pharmaceutical Industries, Ltd. Polymorphs of dasatinib and process for preparation thereof
EP2508523B2 (en) 2007-10-23 2019-04-17 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010062715A3 (en) * 2008-11-03 2010-11-18 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010062715A2 (en) * 2008-11-03 2010-06-03 Teva Pharmaceutical Industries Ltd. Polymorphs of dasatinib and process for preparation thereof
WO2010067374A3 (en) * 2008-12-08 2011-05-26 Hetero Research Foundation Polymorphs of dasatinib
WO2010067374A2 (en) * 2008-12-08 2010-06-17 Hetero Research Foundation Polymorphs of dasatinib
EP2359813A1 (en) 2010-02-04 2011-08-24 Ratiopharm GmbH Pharmaceutical composition comprising N-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamid
WO2011095588A1 (en) 2010-02-04 2011-08-11 Ratiopharm Gmbh Pharmaceutical composition comprising n-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl]-2-methyl-4-pyrimidinyl]amino]-5-thiazolecarboxamid
CN102250084A (en) * 2010-02-08 2011-11-23 南京卡文迪许生物工程技术有限公司 Dasatinib polymorphic substance as well as preparation method and pharmaceutical composition thereof
WO2013065063A1 (en) 2011-11-03 2013-05-10 Cadila Healthcare Limited Anhydrous form of dasatinib, process for its preparation and its use
US9340536B2 (en) 2012-06-15 2016-05-17 Basf Se Multicomponent crystals comprising dasatinib and selected co-crystal formers
EP2861589A4 (en) * 2012-06-15 2016-03-02 Basf Se Multicomponent crystals comprising dasatinib and selected cocrystal formers
RU2650524C2 (en) * 2012-06-15 2018-04-16 Басф Се Multicomponent crystals comprising dasatinib and selected cocrystal formers
CN102827156A (en) * 2012-09-11 2012-12-19 湖南欧亚生物有限公司 Novel industrial synthetic method of dasatinib
CN103819469A (en) * 2012-11-16 2014-05-28 重庆医药工业研究院有限责任公司 Crystal form of dasatinib and preparation method for crystal form of dasatinib
WO2014086326A1 (en) 2012-12-06 2014-06-12 Zentiva, K.S. A method for the preparation and purification of new and known polymorphs and solvates of dasatinib
CN103880833A (en) * 2012-12-19 2014-06-25 北京本草天源药物研究院 Novel crystal form of dasatinib monohydrate, and preparation method and pharmaceutical composition thereof
CN103880833B (en) * 2012-12-19 2018-04-06 北京本草天源药物研究院 New crystalline form of Dasatinib monohydrate and preparation method thereof and pharmaceutical composition
WO2014102759A2 (en) 2012-12-31 2014-07-03 Ranbaxy Laboratories Limited Process for the preparation of dasatinib and its intermediates
US9365526B2 (en) 2012-12-31 2016-06-14 Sun Pharmaceutical Industries Limited Process for the preparation of dasatinib and its intermediates
CN103319476B (en) * 2013-06-13 2015-12-02 济南德爱医药技术有限公司 A kind of kinase inhibitor
CN103319476A (en) * 2013-06-13 2013-09-25 济南德爱医药技术有限公司 Kinase inhibitor
CN104341410A (en) * 2013-08-09 2015-02-11 上海科胜药物研发有限公司 New Dasatinib crystal form and preparation method thereof
CN103483289B (en) * 2013-09-06 2016-01-27 浙江科源化工有限公司 A kind of synthetic method of 2-amino-N-(the chloro-6-aminomethyl phenyl of 2-) thiazole-5-methane amide
CN103483289A (en) * 2013-09-06 2014-01-01 浙江科源化工有限公司 2-amino-N-(2-chloro-6-methyl phenyl)thiazole-5-carboxamide synthesis method
WO2015107545A1 (en) 2013-12-18 2015-07-23 Dharmesh Mahendrabhai Shah Water soluble salts of dasatinib hydrate
WO2015090259A1 (en) * 2013-12-19 2015-06-25 Zentiva, K.S. Method of preparing anhydrous polymorphic form n-6 of dasatinib
CZ306732B6 (en) * 2013-12-19 2017-05-31 Zentiva, K.S. A method of preparation of the anhydrous polymorphic form of N-6 Dasatinib
US9115124B1 (en) 2014-03-11 2015-08-25 Cerbios-Pharma Sa Process and intermediates for the preparation of dasatinib
WO2015181573A1 (en) 2014-05-26 2015-12-03 Egis Gyógyszergyár Zrt. Dasatinib salts
US10023566B2 (en) 2014-05-26 2018-07-17 Egis Gyogyszergyar Zrt Dasatinib salts
EA030142B1 (en) * 2014-05-26 2018-06-29 Эгиш Дьёдьсердьяр Зрт. Dasatinib salts
WO2017002131A1 (en) * 2015-06-29 2017-01-05 Msn Laboratories Private Limited Crystalline forms of n-(2-chloro-6-methy]phenvn-2-[f6-[4-(2-hvdroxvethvl)-l- piperazinvil-2-methvl-4-pvrimidinvllaminol-5-thiazolecarboxamide and their process thereof
US10301302B2 (en) 2015-06-29 2019-05-28 Msn Laboratories Private Limited Crystalline forms of N-(2-chloro-6-methy]phenvn-2-[F6-[4-(2-hvdroxvethvl)-L-piperazin-vil-2-methvil-4-pvrimidinvllaminol-5-thiazolecarboxamide and their process thereof
WO2017098391A1 (en) * 2015-12-11 2017-06-15 Shilpa Medicare Limited Process for the preparation of dasatinib
WO2017108605A1 (en) 2015-12-22 2017-06-29 Synthon B.V. Pharmaceutical composition comprising amorphous dasatinib
CN105503854A (en) * 2015-12-31 2016-04-20 哈药集团技术中心 New crystal form substance of Dasatinib anhydrous substance and preparation method thereof
US10464933B2 (en) 2016-02-03 2019-11-05 Dr. Reddy's Laboratories Limited Solid state forms of dasatinib and processes for their preparation
WO2017144109A1 (en) 2016-02-25 2017-08-31 Remedica Ltd Dasatinib formulation
WO2017199014A1 (en) 2016-05-16 2017-11-23 University Of Dundee Treatment of opioid tolerance
CN106117195A (en) * 2016-06-09 2016-11-16 青岛辰达生物科技有限公司 A kind of synthetic method for treating leukemic medicine Dasatinib
WO2018134189A1 (en) 2017-01-20 2018-07-26 Cerbios-Pharma Sa Co-crystal of an antitumoral compound
WO2018134190A1 (en) * 2017-01-20 2018-07-26 Cerbios-Pharma Sa Co-crystals of an antitumoral compound
IT201700006145A1 (en) * 2017-01-20 2018-07-20 Cerbios Pharma Sa Co-crystal of an antitumor compound
IT201700006157A1 (en) * 2017-01-20 2018-07-20 Cerbios Pharma Sa Co-crystals of an antitumor compound
EP3649126A4 (en) * 2017-07-07 2021-04-07 Biocon Limited Polymorphic forms of dasatinib
CN111108104A (en) * 2017-07-07 2020-05-05 拜康有限公司 Polymorphic forms of dasatinib
US11479770B2 (en) 2017-10-05 2022-10-25 Fulcrum Therapeutics, Inc. Use of p38 inhibitors to reduce expression of DUX4
US10342786B2 (en) 2017-10-05 2019-07-09 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US10537560B2 (en) 2017-10-05 2020-01-21 Fulcrum Therapeutics. Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11291659B2 (en) 2017-10-05 2022-04-05 Fulcrum Therapeutics, Inc. P38 kinase inhibitors reduce DUX4 and downstream gene expression for the treatment of FSHD
US11052088B2 (en) 2018-06-15 2021-07-06 Handa Oncology, Llc Kinase inhibitor salts, and compositions thereof
US11007195B2 (en) 2018-06-15 2021-05-18 Handa Oncology, Llc Kinase inhibitor salts, and compositions thereof
US10940149B1 (en) 2018-06-15 2021-03-09 Handa Oncology, Llc Kinase inhibitor salts and compositions thereof
US11160805B2 (en) 2018-06-15 2021-11-02 Handa Onocology, Llc Kinase inhibitor salts and compositions thereof
CN109369638A (en) * 2018-11-21 2019-02-22 山东罗欣药业集团股份有限公司 A kind of preparation process of Dasatinib
CN109503568A (en) * 2018-12-29 2019-03-22 山东罗欣药业集团股份有限公司 A kind of preparation method of Dasatinib
WO2021032129A1 (en) * 2019-08-20 2021-02-25 湖南华纳大药厂股份有限公司 Kinase inhibitor, preparation therefor, pharmaceutical composition thereof and use thereof
WO2021150981A1 (en) 2020-01-24 2021-07-29 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11202778B2 (en) 2020-01-24 2021-12-21 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11298356B1 (en) 2020-01-24 2022-04-12 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11324745B2 (en) 2020-01-24 2022-05-10 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11633398B2 (en) 2020-01-24 2023-04-25 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
US11413290B2 (en) 2020-01-24 2022-08-16 Nanocopoeia, Llc Amorphous solid dispersions of dasatinib and uses thereof
WO2021176083A1 (en) 2020-03-06 2021-09-10 Zentiva K.S. Pharmaceutical compositions comprising dasatinib anhydrous and uses thereof
WO2022214588A1 (en) 2021-04-07 2022-10-13 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Means and methods for enhancing receptor-targeted gene transfer
EP4071248A1 (en) 2021-04-07 2022-10-12 Deutsches Krebsforschungszentrum Stiftung des öffentlichen Rechts Means and methods for enhancing receptor-targeted gene transfer
CN114634421A (en) * 2022-02-24 2022-06-17 济宁晟泰药业有限公司 Preparation method of dasatinib intermediate
US11980619B2 (en) 2022-07-28 2024-05-14 Nanocopoeia, Llc Pharmaceutical compositions and crushable tablets including amorphous solid dispersions of dasatinib and uses

Also Published As

Publication number Publication date
BRPI0507476A (en) 2007-07-17
ATE453630T1 (en) 2010-01-15
DE602005018601D1 (en) 2010-02-11
RU2006131591A (en) 2008-03-20
DK1711481T3 (en) 2010-04-19
IL177280A0 (en) 2006-12-10
TWI338004B (en) 2011-03-01
KR20060124692A (en) 2006-12-05
IL177280A (en) 2013-09-30
EP1711481B2 (en) 2013-05-22
NO338049B1 (en) 2016-07-25
RU2382039C2 (en) 2010-02-20
KR20120097424A (en) 2012-09-03
ES2337272T5 (en) 2013-09-30
JP2012188446A (en) 2012-10-04
DK1711481T4 (en) 2013-08-26
NZ548613A (en) 2010-05-28
AU2005212405A1 (en) 2005-08-25
KR101310427B1 (en) 2013-09-24
PE20050691A1 (en) 2005-09-25
PL1711481T5 (en) 2013-10-31
EP1711481A2 (en) 2006-10-18
CY1109907T1 (en) 2014-09-10
BRPI0507476B1 (en) 2020-12-08
PT1711481E (en) 2010-02-25
JP2007521340A (en) 2007-08-02
CA2555291C (en) 2017-01-17
NO20063780L (en) 2006-10-27
HRP20100166T1 (en) 2010-05-31
AR047533A1 (en) 2006-01-25
BRPI0507476B8 (en) 2021-05-25
SI1711481T1 (en) 2010-03-31
HK1091835A1 (en) 2007-01-26
HRP20100166T4 (en) 2013-09-30
PL1711481T3 (en) 2010-05-31
IN2014DN06567A (en) 2015-07-10
AU2005212405B2 (en) 2010-08-12
JP5148115B2 (en) 2013-02-20
ES2337272T3 (en) 2010-04-22
EP1711481B1 (en) 2009-12-30
ZA200606242B (en) 2008-01-08
WO2005077945A3 (en) 2006-05-18
CA2555291A1 (en) 2005-08-25
GEP20094804B (en) 2009-10-26
SI1711481T2 (en) 2013-10-30
TW200530205A (en) 2005-09-16

Similar Documents

Publication Publication Date Title
US8680103B2 (en) Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
CA2555291C (en) Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
US20050215795A1 (en) Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
CN1980909B (en) Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
EP1928844B1 (en) Bis-aryl amide compounds and methods of use
US11820756B2 (en) Bradykinin B2 receptor antagonists
JP7157764B2 (en) Novel piperidine-2,6-dione derivative and use thereof
JP2002542193A (en) Cyclic protein tyrosine kinase inhibitor
WO2005076990A2 (en) Process for preparing 2-aminothiazole-5-carboxamides useful as kinase inhibitors
WO2022117051A1 (en) Macrocyclic compound, preparation method therefor and use thereof
KR20080031038A (en) NOVEL HETEROCYCLIC NF-kappa;B INHIBITORS
EP1953148B1 (en) Heterocyclic amide compound and use thereof
MXPA06008683A (en) Process for preparing 2-aminothiazole-5-aromatic carboxamides as kinase inhibitors
CN115484954A (en) Potent and selective irreversible inhibitors of IRAK1
CZ20001717A3 (en) Benzothiazole inhibitors of protein-tyrosine kinase

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 548613

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 12006501426

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 4309/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2006/06242

Country of ref document: ZA

Ref document number: 200606242

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: PA/a/2006/008683

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2005722772

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 177280

Country of ref document: IL

Ref document number: 06076384

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 2555291

Country of ref document: CA

Ref document number: 1020067015839

Country of ref document: KR

Ref document number: 2005212405

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2006552303

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Ref document number: DE

ENP Entry into the national phase

Ref document number: 2005212405

Country of ref document: AU

Date of ref document: 20050204

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005212405

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 9607

Country of ref document: GE

WWE Wipo information: entry into national phase

Ref document number: 2006131591

Country of ref document: RU

Ref document number: 1200601461

Country of ref document: VN

WWE Wipo information: entry into national phase

Ref document number: 200580011916.6

Country of ref document: CN

WWP Wipo information: published in national office

Ref document number: 2005722772

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1020067015839

Country of ref document: KR

ENP Entry into the national phase

Ref document number: PI0507476

Country of ref document: BR