WO2016090257A1 - Salts and crystalline forms of 6-acetyl-8-cyclopentyl-5-methyl-2((5-(piperazin-1-yl)pyridin-2-yl)amino)pyrido[2,3-d] pyrimidin-7(8h)-one (palbociclib) - Google Patents

Salts and crystalline forms of 6-acetyl-8-cyclopentyl-5-methyl-2((5-(piperazin-1-yl)pyridin-2-yl)amino)pyrido[2,3-d] pyrimidin-7(8h)-one (palbociclib) Download PDF

Info

Publication number
WO2016090257A1
WO2016090257A1 PCT/US2015/064010 US2015064010W WO2016090257A1 WO 2016090257 A1 WO2016090257 A1 WO 2016090257A1 US 2015064010 W US2015064010 W US 2015064010W WO 2016090257 A1 WO2016090257 A1 WO 2016090257A1
Authority
WO
WIPO (PCT)
Prior art keywords
palbociclib
isethionate
crystalline form
xrpd
crystalline
Prior art date
Application number
PCT/US2015/064010
Other languages
French (fr)
Inventor
Minhua Chen
Yanfeng Zhang
Chaohui YANG
Xiaoyu Zhang
Jiaoyang LI
Peng Wang
Li PIXU
Original Assignee
Crystal Pharmatech Inc.
Crystal Pharmatech Co., Ltd.
Suzhou Pengxu Pharmatech Co., Ltd
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
Application filed by Crystal Pharmatech Inc., Crystal Pharmatech Co., Ltd., Suzhou Pengxu Pharmatech Co., Ltd filed Critical Crystal Pharmatech Inc.
Publication of WO2016090257A1 publication Critical patent/WO2016090257A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This invention relates to new crystalline forms of palbociclib and its salts, and pharmaceutical compositions, methods of preparation, and method of uses thereof.
  • CDK 4/6 inhibitors are novel agents that have shown promising results in the treatment of breast cancer.
  • CDK4 and CDK6 are proteins that are part of a cell cycle regulatory pathway that also includes pi 6, cyclin D, and the retinoblastoma (Rb) protein.
  • CDK4/6 inhibitors bind to CDK4 and CDK6, preventing phosphorylation of the Rb protein. This halts cell cycle progression and induces Gl cell cycle arrest.
  • Malignant cells frequently acquire mutations in the CDK4/6 pathway, either activating mutations of CDK4/6 or mutations in CDK4/6 regulatory mechanisms, thereby conferring a growth advantage. By inhibiting CDK4/6, tumor cells are unable to exploit this pathway for cell proliferation.
  • Palbociclib is a CDK4/6 inhibitor approved by USFDA on 3 rd Feb 2015 for the treatment of ER-positive breast cancer.
  • Palbociclib has a structure of formula (I), with a chemical name as 6-acetyl-8-cyclopentyl-5-methyl-2-((5-(piperazin-l-yl)pyridin-2- yl)amino)pyrido 2,3-d]pyrimidin-7(8H)-one.
  • crystalline forms of palbociclib freebase have been reported in WO2014128588, designated as Form A and Form B, which have very poor solubility and, therefore, low bioavailability.
  • palbociclib salts and their polymorphic forms have also been reported in US7863278.
  • the mono-HCl salt is slightly hygroscopic and has poor crystalline habit.
  • the present inventors surprisingly discovered new crystalline forms of palbociclib and its salts, which have desired pharmacological properties useful for pharmaceutical development and can be prepared readily in environmentally friendly solvent systems.
  • the present invention provides crystalline forms of palbociclib, designated as Forms I and II, respectively.
  • the present invention provides processes for preparation of Forms I and II of palbociclib.
  • the present invention provides pharmaceutical salts of palbociclib, particularly isethionate, sulfate, phosphate, acetate, L-lactate, maleate, fumarate, citrate, succinate, L-tartrate, hippurate, glutarate, adipate, glycolate, esylate and diesylate.
  • the present invention provides processes for preparation of any of the palbociclib salts disclosed herein.
  • the present invention provides solid pharmaceutical compositions comprising any of crystalline forms I and II of palbociclib, crystalline forms of palbociclib salts, or any of combinations thereof.
  • FIG. 1 shows an X-ray powder diffraction (XRPD) pattern of crystalline Form I of palbociclib.
  • FIG. 2 shows a differential scanning calorimetry (DSC) thermogram of crystalline
  • FIG. 3 shows a thermogravimetric analysis (TGA) thermogram of crystalline Form I of palbociclib.
  • FIG. 4 shows an X-ray powder diffraction (XRPD) pattern of crystalline Form II of palbociclib.
  • FIG. 5 shows an X-ray powder diffraction (XRPD) pattern of palbociclib sulfate
  • FIG. 6 shows an X-ray powder diffraction (XRPD) pattern of palbociclib phosphate Form A.
  • FIG. 7 shows an X-ray powder diffraction (XRPD) pattern of palbociclib acetate Form A.
  • FIG. 8 shows a differential scanning calorimetry (DSC) thermogram of palbociclib acetate Form A.
  • FIG. 9 shows an X-ray powder diffraction (XRPD) pattern of palbociclib acetate Form B.
  • FIG. 10 shows a differential scanning calorimetry (DSC) thermogram of palbociclib acetate Form B.
  • FIG. 11 shows an X-ray powder diffraction (XRPD) pattern of palbociclib L-lactate Form A.
  • FIG. 12 shows a differential scanning calorimetry (DSC) thermogram of palbociclib L-lactate Form A.
  • FIG. 13 shows an X-ray powder diffraction (XRPD) pattern of palbociclib maleate Form A.
  • FIG. 14 shows a differential scanning calorimetry (DSC) thermogram of palbociclib maleate Form A.
  • FIG. 15 shows a thermogravimetric analysis (TGA) thermogram of palbociclib maleate Form A.
  • FIG. 16 shows an X-ray powder diffraction (XRPD) pattern of palbociclib maleate Form B.
  • FIG. 17 shows a differential scanning calorimetry (DSC) thermogram of palbociclib maleate Form B.
  • FIG. 18 shows an X-ray powder diffraction (XRPD) pattern of palbociclib fumarate Form A.
  • FIG. 19 shows a differential scanning calorimetry (DSC) thermogram of palbociclib fumarate Form A.
  • FIG. 20 shows an X-ray powder diffraction (XRPD) pattern of palbociclib fumarate Form B.
  • FIG. 21 shows a differential scanning calorimetry (DSC) thermogram of palbociclib fumarate Form B.
  • FIG. 22 shows an X-ray powder diffraction (XRPD) pattern of palbociclib citrate Form A.
  • FIG. 23 shows a differential scanning calorimetry (DSC) thermogram of palbociclib citrate Form A.
  • FIG. 24 shows an X-ray powder diffraction (XRPD) pattern of palbociclib citrate Form B.
  • FIG. 25 shows a differential scanning calorimetry (DSC) thermogram of palbociclib citrate Form B.
  • FIG. 26 shows an X-ray powder diffraction (XRPD) pattern of palbociclib succinate
  • FIG. 27 shows a differential scanning calorimetry (DSC) thermogram of palbociclib succinate Form A.
  • FIG. 28 shows an X-ray powder diffraction (XRPD) pattern of palbociclib succinate Form B.
  • FIG. 29 shows a differential scanning calorimetry (DSC) thermogram of palbociclib succinate Form B.
  • FIG. 30 shows an X-ray powder diffraction (XRPD) pattern of palbociclib succinate Form C.
  • FIG. 31 shows a differential scanning calorimetry (DSC) thermogram of palbociclib succinate Form C.
  • FIG. 32 shows an X-ray powder diffraction (XRPD) pattern of palbociclib L-tartrate Form A.
  • FIG. 33 shows a differential scanning calorimetry (DSC) thermogram of palbociclib L-tartrate Form A.
  • FIG. 34 shows a thermogravimetric analysis (TGA) thermogram of palbociclib L- tartrate Form A.
  • FIG. 35 shows an X-ray powder diffraction (XRPD) pattern of palbociclib L-tartrate Form B.
  • FIG. 36 shows an X-ray powder diffraction (XRPD) pattern of palbociclib glutarate Form A.
  • FIG. 37 shows a differential scanning calorimetry (DSC) thermogram of palbociclib glutarate Form A.
  • FIG. 38 shows an X-ray powder diffraction (XRPD) pattern of palbociclib glutarate Form B.
  • FIG. 39 shows a differential scanning calorimetry (DSC) thermogram of palbociclib glutarate Form B.
  • FIG. 40 shows a thermogravimetric analysis (TGA) thermogram of palbociclib glutarate Form B.
  • FIG. 41 shows an X-ray powder diffraction (XRPD) pattern of palbociclib adipate Form A.
  • FIG. 42 shows a differential scanning calorimetry (DSC) thermogram of adipate
  • FIG. 43 shows a thermogravimetric analysis (TGA) thermogram of palbociclib adipate Form A.
  • FIG. 44 shows an X-ray powder diffraction (XRPD) pattern of palbociclib adipate Form B.
  • FIG. 45 shows an X-ray powder diffraction (XRPD) pattern of palbociclib glycolate Form A.
  • FIG. 46 shows a differential scanning calorimetry (DSC) thermogram of glycolate Form A.
  • FIG. 47 shows a thermogravimetric analysis (TGA) thermogram of palbociclib glycolate Form A.
  • FIG. 48 shows an X-ray powder diffraction (XRPD) pattern of palbociclib diesylate Form A.
  • FIG. 49 shows an X-ray powder diffraction (XRPD) pattern of palbociclib diesylate Form B.
  • FIG. 50 shows an X-ray powder diffraction (XRPD) pattern of palbociclib diesylate Form C.
  • FIG. 51 shows an X-ray powder diffraction (XRPD) pattern of palbociclib hippurate Form A.
  • FIG. 52 shows a differential scanning calorimetry (DSC) thermogram of palbociclib hippurate Form A.
  • FIG. 53 shows an X-ray powder diffraction (XRPD) pattern of palbociclib hippurate
  • FIG. 54 shows a differential scanning calorimetry (DSC) thermogram of hippurate Form B.
  • FIG. 55 shows an X-ray powder diffraction (XRPD) pattern of palbociclib esylate Form A.
  • FIG. 56 shows a differential scanning calorimetry (DSC) thermogram of palbociclib esylate Form A.
  • FIG. 57 shows an X-ray powder diffraction (XRPD) pattern of palbociclib esylate Form B.
  • FIG. 58 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate
  • FIG. 59 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form a.
  • FIG. 60 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate Form ⁇ .
  • FIG. 61 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate Form ⁇ .
  • FIG. 62 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form ⁇ .
  • FIG. 63 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate
  • FIG. 64 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form ⁇ .
  • FIG. 65 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate Form ⁇ .
  • FIG. 66 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form ⁇ .
  • FIG. 67 shows comparison of the XRPD pattern of palbociclib crystalline Form I after being stored at 25°C/60%RH for 225 days with its initial XPRD pattern, the initial XRPD pattern shown above, and the XRPD pattern after being stored at 25°C/60%RH for 225 days shown below.
  • DSC differential scanning calorimetry
  • FIG. 68 shows comparison of the XRPD pattern of palbociclib crystalline Form I after being stored at 40°C/75%RH for 225 days with its initial XPRD pattern, the initial XRPD pattern shown above, and the XRPD pattern after being stored at 40°C/75%RH for 225 days shown below.
  • FIG. 69 shows the XRPD pattern of palbociclib crystalline Form I converted from Form A in dichloromethane at 50 °C.
  • FIG. 70 shows the XRPD pattern of palbociclib crystalline Form I converted from Form A in dichloromethane/methanol/water mixture at room temperature.
  • the present invention is based on a surprising discovery that palbociclib can exist in different crystalline forms and salts, and these forms and salts can be prepared readily from environmentally friendly solvent systems using convenient methods.
  • the present invention provides a crystalline form of palbociclib, designated as Form I.
  • the crystalline Form I is characterized by an X-ray powder diffraction pattern comprising the following 2 ⁇ values measured using CuKa radiation: 5.1° ⁇ 0.2°, 9.8° ⁇ 0.2°, and 19.0° ⁇ 0.2°.
  • the crystalline Form I is characterized by an X-ray powder diffraction pattern further comprising the following 2 ⁇ values measured using CuKa radiation: 11.5° ⁇ 0.2°, 9.5° ⁇ 0.2°, and 22.2° ⁇ 0.2°.
  • the crystalline Form I is characterized by an X-ray powder diffraction pattern further comprising the following 2 ⁇ values measured using CuKa radiation: 7.9° ⁇ 0.2°, 12.2° ⁇ 0.2°, 14.8° ⁇ 0.2°, and 16.3° ⁇ 0.2°.
  • the crystalline Form I is characterized by an X-ray powder diffraction pattern comprising the following 2 ⁇ values measured using CuKa radiation: 5.1° ⁇ 0.2°, 7.9° ⁇ 0.2°, 9.5° ⁇ 0.2°, 9.8° ⁇ 0.2°, 11.5° ⁇ 0.2°, 12.2° ⁇ 0.2°, 14.8° ⁇ 0.2°, 16.3° ⁇ 0.2°, 19.0° ⁇ 0.2°, and 22.2° ⁇ 0.2°.
  • the crystalline Form I has an X-ray powder diffraction pattern substantially as shown in FIG. 1.
  • the crystalline Form I has a differential scanning calorimetry thermogram substantially as shown in FIG. 2, which exhibits an endothermic peak temperature of about 260.5 °C.
  • the crystalline Form I has a thermal gravimetric analysis thermogram as shown in FIG. 3, which exhibits about 0.8% weight loss when heated up to 220 °C.
  • Form I is unsolvated. In some embodiment, Form I is anhydrous.
  • the present invention provides a crystalline form of palbociclib, designated as Form II.
  • the crystalline Form II is characterized by an X-ray powder diffraction pattern comprising the following 2 ⁇ values measured using CuKa radiation: 9.4° ⁇ 0.2°, 12.6° ⁇ 0.2°, and 17.5° ⁇ 0.2°.
  • the X-ray powder diffraction pattern of crystalline Form II further comprises the following 2 ⁇ values measured using CuKa radiation: 7.3° ⁇ 0.2°, 10.9° ⁇ 0.2°, and 16.4° ⁇ 0.2°.
  • the X-ray powder diffraction pattern of crystalline Form II further comprises the following 2 ⁇ values measured using CuKa radiation: 11.7 o ⁇ 0.2°, 19.3° ⁇ 0.2°, 22.5° ⁇ 0.2°, and 23.7° ⁇ 0.2°.
  • the X-ray powder diffraction pattern of crystalline Form II comprises the following 2 ⁇ values measured using CuKa radiation: 7.3° ⁇ 0.2°, 9.4° ⁇ 0.2°, 10.9° ⁇ 0.2°, 11.7° ⁇ 0.2°, 12.6° ⁇ 0.2°, 16.4° ⁇ 0.2°, 17.5° ⁇ 0.2°, 19.3° ⁇ 0.2°, 22.5° ⁇ 0.2°, and 23.7° ⁇ 0.2°.
  • the crystalline Form II has an X-ray powder diffraction pattern substantially as depicted in FIG. 4.
  • the present invention provides processes for preparation of palbociclib Forms I and II, which comprises crystallizing palbociclib in an aid of one or two crystallization solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ester solvents, ether solvents, aromatic solvents, nitrile solvents, haloalkane and water via a crystallization method selected from slurry, evaporation, cooling, anti-solvent addition, with or without seeding.
  • crystallization solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ester solvents, ether solvents, aromatic solvents, nitrile solvents, haloalkane and water
  • a crystallization method selected from slurry, evaporation, cooling, anti-solvent addition, with or without seeding.
  • the solvent is dichloromethane
  • the crystallization method is slurry and Form I is obtained.
  • the solvent is a mixture of ethanol/water
  • the crystallization method is slow evaporation of the solvents
  • Form II is obtained.
  • the mixture of ethanol/water is in 6: 1 (v/v) ratio.
  • the present invention provides novel pharmaceutical salts of palbociclib, particularly isethionate, sulfate, phosphate, acetate, L-lactate, maleate, fumarate, citrate, succinate, L-tartrate, hippurate, glutarate, adipate, glycolate, esylate and diesylate.
  • the present invention provides crystalline forms of the
  • pharmaceutical salts of palbociclib particularly isethionate Forms ⁇ , ⁇ , ⁇ , ⁇ , and ⁇ , sulfate Form A, phosphate Form A, acetate Forms A and B, L-lactate Form A, maleate Forms A and B, fumarate Forms A and B, citrate Forms A and B, succinate Forms A, B, and C, L- tartrate Forms A and B, glutarate Forms A and B, adipate Forms A and B, glycolate Form A, diesylate Forms A, B, and C, hippurate Forms A and B, and esylate Forms A and B.
  • the crystalline forms of palbociclib salts are characterized by an X- ray powder diffraction pattern comprising the following 2 ⁇ values:
  • succinate Form A 9.2, 18.7, 4.2, 24.6, 19.3, 21.3, 13.9, 10.6, 8.3 succinate Form B 7.0, 17.2, 4.8, 4.2, 8.0, 21.1, 10.5, 13.9, 18.5 succinate Form C 4.2, 12.8, 18.9, 10.8, 16.8, 8.5, 12.3, 9.4, 21.4
  • L-tartrate Form B 10.9, 3.1, 12.0, 19.3, 17.2, 4.0, 9.0, 11.5, 21.9 glutarate Form A 17.3, 13.0, 9.5, 19.8, 9.8, 7.7, 15.2, 26.3, 14.8 glutarate Form B 6.9, 16.9, 10.0, 10.1, 13.9, 20.4, 11.7, 4.9, 7.7 adipate Form A 4.3, 9.2, 13.0, 19.5, 14.9, 19.4, 17.6, 18.4, 11.4 adipate Form B 11.7, 3.9, 10.9, 7.8, 19.5, 21.8, 16.3, 13.9, 8.5 glycolate Form A 10.8, 9.2, 13.8, 4.6, 19.5, 18.4, 22.9, 21.7, 14.4 diesylate Form A 6.0, 9.0, 15.0, 18.2, 21.1, 23.8
  • diesylate Form B 6.8, 3.4, 17.1, 16.8, 13.6, 20.5, 24.1, 11.9, 10.1 diesylate Form C 5.8, 6.8, 8.8, 22.2, 7.3, 11.7, 13.6, 16.5, 11.0 hippurate Form A 5.2, 5.4, 10.8, 10.1, 8.4, 20.9, 15.2, 15.8, 12.9 hippurate Form B 6.0, 4.2, 4.8, 7.5, 18.3, 10.7, 12.4, 9.7, 15.8 esylate Form A 6.4, 4.3, 9.8, 15.0, 12.9, 16.8, 17.2, 25.9, 8.6 esylate Form B 9.0, 8.5, 19.3, 5.0, 10.1, 11.0, 4.2, 22.1, 12.6 isethionate Form a 17.9, 18.2, 22.9,9.6, 14.9, 26.8,10.0,12.5, 19.1 isethionate Form ⁇ 8.0, 10.3, 18.6,10.1, 15.9, 18.3,9.2, 11.9,18.0 isethionate Form ⁇ 15.8, 22.6, 23.8,10.7, 20
  • the present invention provides processes for preparation of palbociclib salts, which comprises the step of reacting the freebase of palbociclib with the corresponding acid in the presence of suitable solvent or mixture of solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ether solvents, ester solvents and water.
  • the present invention provides processes for preparation of crystalline forms of palbociclib isethionate, which comprises reacting palbociclib freebase with 2-hydroethanolsulfonic acid or crystallizing palbociclib isethionate in one or two crystallization solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ester solvents, ether solvents, aromatic solvents, nitrile solvents, haloalkane and water through a crystallization method selected from slurrying, evaporating solvents, cooling, adding anti-solvent(s), with or without seeding.
  • crystallization solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ester solvents, ether solvents, aromatic solvents, nitrile solvents, haloalkane and water through a crystallization method selected from slurrying, evaporating solvents, cooling, adding anti-solvent(s), with or without seeding.
  • the solvent is acetonitrile
  • the crystallization method is reacting palbociclib freebase with 2-hydroethanolsulfonic acid, and Form a is obtained.
  • the solvent is ethanol
  • the crystallization method is slow evaporation of solvent(s)
  • Form ⁇ is obtained.
  • the solvent is ⁇ , ⁇ -Dimethylformamide
  • the crystallization method is slurrying
  • Form ⁇ is obtained.
  • the solvent is a mixture ethanol/water
  • the crystallization method is cooling
  • Form ⁇ is obtained.
  • mixture ethanol/water is in 19: 1 (v/v) ratio.
  • the solvent is a mixture of 1 ,4-dioxane/water
  • the crystallization method is cooling
  • Form ⁇ is obtained.
  • the mixture of ethanol/water is in 133:21 (v/v) ratio.
  • the present invention provides solid pharmaceutical compositions, comprising, as an active ingredient, any one or combination of the crystalline forms of palbociclib and/or crystalline forms of palbociclib salts described herein.
  • the present invention provides a method of treating or delaying the progression or onset of a disease or disorder in connection with activity of cyclin- dependent kinase (CDK) 4/6 inhibitor, comprising administering to a subject in need thereof a therapeutically effective amount of palbociclib selected from the group consisting of any one or combination of the crystalline forms of palbociclib and/or crystalline forms of palbociclib salts described herein.
  • CDK cyclin- dependent kinase
  • said disease or disorder is selected from the group consisting of breast cancer, ovary cancer, cervix cancer, prostate cancer, testis cancer, esophagus cancer, stomach cancer, skin cancer, and lung cancer.
  • the present invention provides use of palbociclib selected from the group consisting of any one or combination of the crystalline forms of palbociclib and/or crystalline forms of palbociclib salts described herein in the manufacture of a medicament for treating or delaying the progression or onset of a disease or disorder in connection with activity of cyclin-dependent kinase (CDK) 4/6 inhibitor.
  • said disease or disorder is selected from the group consisting of breast cancer, ovary cancer, cervix cancer, prostate cancer, testis cancer, esophagus cancer, stomach cancer, skin cancer, and lung cancer.
  • alcohol refers to Ci-C 6 alkyl alcohol, preferably C 1 -C 4 alkyl alcohol, for example, in some embodiments preferably, methanol, ethanol, isopropanol, or the like.
  • ketone refers to C 3 -C7 alkanone, having a formula RCOR', wherein R and R' are each independently C 1 -C 4 alkyl, for example, in some embodiments preferably, acetone, butanone, 2-pentanone, 3-pentanone, methyl isobutyl ketone (MIBK), or the like.
  • R and R' are each independently C 1 -C 4 alkyl, for example, in some embodiments preferably, acetone, butanone, 2-pentanone, 3-pentanone, methyl isobutyl ketone (MIBK), or the like.
  • esters refers to a lower alkyl aliphatic acid ester having a formula RCOOR', wherein R and R' are each independently C 1 -C 4 alkyls, for example, in some embodiments preferably, ethyl acetate, ethyl propionate, methyl acetate, propyl acetate, ispropyl acetate, or the like.
  • ether refers to a lower alkyl ether or cyclic ether (each alkyl having 1 to 4 carbon atoms), including but not limited to diethyl ether, di-isopropyl ether, ethyl propyl ether, methyl t-butyl ether (MTBE), tetrahydrofuran (THF), 1,4-dioxane, or the like.
  • aromatic hydrocarbon refers to benzene optionally substituted by 1 to 3 methyl or ethyl groups, for example, in some embodiments preferably, toluene, 1,2-xylene, 1,4-xylene, 1 ,3-xylene, cumene, ethylbenzene, or the like.
  • halogenated hydrocarbon refers to Ci-C 6 alkane substituted by one to six, preferably one to four, F and/or CI atoms, for example, in some embodiments preferably, dichloromethane, chloroform, 1,1,1-trifluoroethane, or the like.
  • nitrile refers to C 2 -C 4 alkyl nitrile, i.e., CH 3 CN, CH 3 CH 2 CN, CH 3 CH 2 CH 2 CN, or CH 3 CH(CN)CH 3 , preferably CH 3 CN.
  • treatment refers to the management and care of a patient for the purpose of combating the disease, condition or disorder.
  • therapeutically effective amount refers to an amount of a drug or a therapeutic agent that will elicit the desired biological and/or medical response of a tissue, system or an animal (including man) that is being sought by a researcher or clinician.
  • subject or “patient” refers to a mammalian animal.
  • mammal or “mammalian animal” includes, but is not limited to, humans, dogs, cats, horses, pigs, cows, monkeys, rabbits and mice. The preferred mammals are humans.
  • administering means applying a compound of the invention, or a pharmaceutically acceptable salt, pro-drug or composition thereof, to a subject in need of treatment.
  • the administration of the composition of the present invention in order to practice the present methods of therapy is carried out by administering a therapeutically effective amount of the compounds in the composition to a subject in need of such treatment or prophylaxis.
  • the need for a prophylactic administration according to the methods of the present invention is determined via the use of well-known risk factors.
  • the effective amount of an individual compound is determined, in the final analysis, by the physician in charge of the case, but depends on factors such as the exact disease to be treated, the severity of the disease and other diseases or conditions from which the patient suffers, the chosen route of administration, other drugs and treatments which the patient may concomitantly require, and other factors in the physician's judgment.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
  • the term “about” when applied to a parameter, such as amount, temperature, time, or the like, it indicates that the parameter can usually vary by ⁇ 10%, preferably within ⁇ 5%, and more preferably within ⁇ 2%. However, in the case of a melting or onset temperature of a crystalline form as measured by in a DSC thermogram, the term “about” may indicate that the melting or onset temperature can usually vary within ⁇ 2°C, regardless of the absolute value of the melting or onset temperature, as a person skilled in the art would understand it. As would be understood by a person skilled in the art, when a parameter is not critical, a number is often given only for illustration purpose, instead of being limiting.
  • XRPD was performed with Panalytical Empyrean XRPD on a Si single crystal holder. The 2 ⁇ position was calibrated against Panalytical 640 Si powder standard. Details of XRPD method used in the experiments are listed below.
  • characteristic peaks are defined as a subset of representative (prominent) peaks used to differentiate one crystalline polymorph or form from another crystalline polymorph or form. Characteristic peaks may be determined by evaluating which representative peaks, if any, are present in one crystalline polymorph of a compound but not in all other known crystalline polymorphs of that compound. However, not all crystalline polymorphs of a compound would necessarily have at least one characteristic peak. As a person of ordinary skill in the art would understand, in certain situations, the overall diffraction pattern should be used to determine whether a crystal form exists as described or claimed.
  • Heating rate 10 °C per minute.
  • Heating rate 10 °C per minute.
  • Purge gas nitrogen.
  • the XRPD pattern, DSC thermogram, and TGA thermogram of Form I obtained from this example are displayed in FIGs. 1, 2, and 3, respectively.
  • the thermal gravimetric analysis of the sample showed weight loss of only about 0.8% when heated to 220 °C, indicating that the sample is not a solvate, but anhydrous.
  • the XRPD pattern of Form II obtained from this example is displayed in FIG. 4.
  • the XRPD pattern of sulfate Form A obtained from this example are displayed in FIG. 5.
  • the XRPD pattern of phosphate Form A obtained from this example is displayed in FIG. 6.
  • the XRPD pattern and DSC thermogram of fumarate Form B obtained from this example are displayed in FIGs. 20 and 21.
  • the XRPD pattern of L-tartrate Form B obtained from this example is displayed in FIG. 35.
  • the XRPD pattern and DSC thermogram of glutarate Form A obtained from this example are displayed in FIGs. 36 and 37.
  • the XRPD pattern, DSC thermogram and TGA thermogram of glutarate Form B obtained from this example are displayed in FIGs. 38, 39 and 40.
  • the XRPD pattern, DSC thermogram and TGA thermogram of adipate Form A obtained from this example are displayed in FIGs. 41, 42 and 43.
  • the XRPD pattern of diesylate Form A obtained from this example are displayed in FIG. 48.
  • the XRPD pattern of diesylate Form B obtained from this example are displayed in FIG. 49.
  • the XRPD pattern and DSC thermogram of hippurate Form B obtained from this example are displayed in FIGs. 53 and 54.
  • the XRPD pattern of palbociclib isethionate Form ⁇ obtained from this example is displayed in FIG. 60.
  • the XRPD pattern and DSC thermogram of palbociclib isethionate Form ⁇ obtained from this example are displayed in FIGs. 61 and 62, respectively.
  • DSC thermogram exhibits two endothermic peaks with onset temperature of about 139.5 °C and 275.3 °C
  • the XRPD pattern and DSC thermogram of palbociclib isethionate Form ⁇ obtained from this example are displayed in FIGs. 63 and 64.
  • DSC thermogram exhibits two endothermic peaks with onset temperature of about 47.2 °C and 273.9 °C.
  • the XRPD pattern and DSC thermogram of palbociclib isethionate Form ⁇ obtained from this example is displayed in FIGs. 65 and 66.
  • DSC thermogram exhibits two endothermic peaks with onset temperature of about 186.0 °C and 276.7 °C.
  • Form A (409.9 mg) was added into 20 mL of dichloromethane to suspend in a glass vial, then placed in an oven at 50°C for 7 days stirring (500 r/min), and the solids were isolated (Form I) and analyzed by HPLC. See Table 37.
  • Form A (409.9 mg) (see WO2014128588) was added and suspended in 20 mL of dichloromethane in a glass vial, and the suspension then placed in an oven at 50°C for 7 days while stirring (500 r/min). The solids were isolated and analyzed by XRPD, which shows the formation of crystalline Form I. See FIG. 69.

Abstract

New salts and crystalline Forms of palbociclib, processes for their preparation, pharmaceutical compositions comprising the new salts or crystalline Forms, and use of the new salts and crystalline Forms of palbociclib for treating or delaying diseases or disorders related to activity of cyclin-dependent kinase (CDK) 4/6 are disclosed.

Description

SALTS AND CRYSTALLINE FORMS OF 6-ACETYL-8-CYCLOPENTYL-5- METHYL-2-((5-(PIPERAZIN-l-YL)PYRIDIN-2-YL)AMINO)PYRIDO[2,3- D]PYRIMIDIN-7(8H)-ONE (PALBOCICLIB) CROSS REFERENCE TO RELATED APPLICATIONS
This application claims priority under 35 U.S.C. § 119(e) to Provisional Application No. 62/088,438, filed on December 5, 2014, which is herein incorporated by reference in its entirety.
FIELD OF THE INVENTION
This invention relates to new crystalline forms of palbociclib and its salts, and pharmaceutical compositions, methods of preparation, and method of uses thereof.
BACKGROUND OF THE INVENTION
Cyclin-dependent kinase (CDK) 4/6 inhibitors are novel agents that have shown promising results in the treatment of breast cancer. CDK4 and CDK6 are proteins that are part of a cell cycle regulatory pathway that also includes pi 6, cyclin D, and the retinoblastoma (Rb) protein. CDK4/6 inhibitors bind to CDK4 and CDK6, preventing phosphorylation of the Rb protein. This halts cell cycle progression and induces Gl cell cycle arrest. Malignant cells frequently acquire mutations in the CDK4/6 pathway, either activating mutations of CDK4/6 or mutations in CDK4/6 regulatory mechanisms, thereby conferring a growth advantage. By inhibiting CDK4/6, tumor cells are unable to exploit this pathway for cell proliferation.
Palbociclib is a CDK4/6 inhibitor approved by USFDA on 3rd Feb 2015 for the treatment of ER-positive breast cancer. Palbociclib has a structure of formula (I), with a chemical name as 6-acetyl-8-cyclopentyl-5-methyl-2-((5-(piperazin-l-yl)pyridin-2- yl)amino)pyrido 2,3-d]pyrimidin-7(8H)-one.
Figure imgf000002_0001
Some crystalline forms of palbociclib freebase have been reported in WO2014128588, designated as Form A and Form B, which have very poor solubility and, therefore, low bioavailability. In addition, palbociclib salts and their polymorphic forms have also been reported in US7863278. The mono-HCl salt is slightly hygroscopic and has poor crystalline habit.
Therefore, new crystalline forms or salts of palbociclib, in particular their stable polymorphs with superior pharmacological activities, and convenient methods to prepare them remain a great need.
SUMMARY OF THE INVENTION
The present inventors surprisingly discovered new crystalline forms of palbociclib and its salts, which have desired pharmacological properties useful for pharmaceutical development and can be prepared readily in environmentally friendly solvent systems.
In one aspect, the present invention provides crystalline forms of palbociclib, designated as Forms I and II, respectively.
In another aspect, the present invention provides processes for preparation of Forms I and II of palbociclib.
In another aspect, the present invention provides pharmaceutical salts of palbociclib, particularly isethionate, sulfate, phosphate, acetate, L-lactate, maleate, fumarate, citrate, succinate, L-tartrate, hippurate, glutarate, adipate, glycolate, esylate and diesylate.
In another aspect, the present invention provides processes for preparation of any of the palbociclib salts disclosed herein.
In another aspect, the present invention provides solid pharmaceutical compositions comprising any of crystalline forms I and II of palbociclib, crystalline forms of palbociclib salts, or any of combinations thereof.
Other aspects and embodiments of the present invention will be further illustrated in the following description and examples.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1 shows an X-ray powder diffraction (XRPD) pattern of crystalline Form I of palbociclib.
FIG. 2 shows a differential scanning calorimetry (DSC) thermogram of crystalline
Form I of palbociclib. FIG. 3 shows a thermogravimetric analysis (TGA) thermogram of crystalline Form I of palbociclib.
FIG. 4 shows an X-ray powder diffraction (XRPD) pattern of crystalline Form II of palbociclib.
FIG. 5 shows an X-ray powder diffraction (XRPD) pattern of palbociclib sulfate
Form A.
FIG. 6 shows an X-ray powder diffraction (XRPD) pattern of palbociclib phosphate Form A.
FIG. 7 shows an X-ray powder diffraction (XRPD) pattern of palbociclib acetate Form A.
FIG. 8 shows a differential scanning calorimetry (DSC) thermogram of palbociclib acetate Form A.
FIG. 9 shows an X-ray powder diffraction (XRPD) pattern of palbociclib acetate Form B.
FIG. 10 shows a differential scanning calorimetry (DSC) thermogram of palbociclib acetate Form B.
FIG. 11 shows an X-ray powder diffraction (XRPD) pattern of palbociclib L-lactate Form A.
FIG. 12 shows a differential scanning calorimetry (DSC) thermogram of palbociclib L-lactate Form A.
FIG. 13 shows an X-ray powder diffraction (XRPD) pattern of palbociclib maleate Form A.
FIG. 14 shows a differential scanning calorimetry (DSC) thermogram of palbociclib maleate Form A.
FIG. 15 shows a thermogravimetric analysis (TGA) thermogram of palbociclib maleate Form A.
FIG. 16 shows an X-ray powder diffraction (XRPD) pattern of palbociclib maleate Form B.
FIG. 17 shows a differential scanning calorimetry (DSC) thermogram of palbociclib maleate Form B.
FIG. 18 shows an X-ray powder diffraction (XRPD) pattern of palbociclib fumarate Form A. FIG. 19 shows a differential scanning calorimetry (DSC) thermogram of palbociclib fumarate Form A.
FIG. 20 shows an X-ray powder diffraction (XRPD) pattern of palbociclib fumarate Form B.
FIG. 21 shows a differential scanning calorimetry (DSC) thermogram of palbociclib fumarate Form B.
FIG. 22 shows an X-ray powder diffraction (XRPD) pattern of palbociclib citrate Form A.
FIG. 23 shows a differential scanning calorimetry (DSC) thermogram of palbociclib citrate Form A.
FIG. 24 shows an X-ray powder diffraction (XRPD) pattern of palbociclib citrate Form B.
FIG. 25 shows a differential scanning calorimetry (DSC) thermogram of palbociclib citrate Form B.
FIG. 26 shows an X-ray powder diffraction (XRPD) pattern of palbociclib succinate
Form A.
FIG. 27 shows a differential scanning calorimetry (DSC) thermogram of palbociclib succinate Form A.
FIG. 28 shows an X-ray powder diffraction (XRPD) pattern of palbociclib succinate Form B.
FIG. 29 shows a differential scanning calorimetry (DSC) thermogram of palbociclib succinate Form B.
FIG. 30 shows an X-ray powder diffraction (XRPD) pattern of palbociclib succinate Form C.
FIG. 31 shows a differential scanning calorimetry (DSC) thermogram of palbociclib succinate Form C.
FIG. 32 shows an X-ray powder diffraction (XRPD) pattern of palbociclib L-tartrate Form A.
FIG. 33 shows a differential scanning calorimetry (DSC) thermogram of palbociclib L-tartrate Form A.
FIG. 34 shows a thermogravimetric analysis (TGA) thermogram of palbociclib L- tartrate Form A. FIG. 35 shows an X-ray powder diffraction (XRPD) pattern of palbociclib L-tartrate Form B.
FIG. 36 shows an X-ray powder diffraction (XRPD) pattern of palbociclib glutarate Form A.
FIG. 37 shows a differential scanning calorimetry (DSC) thermogram of palbociclib glutarate Form A.
FIG. 38 shows an X-ray powder diffraction (XRPD) pattern of palbociclib glutarate Form B.
FIG. 39 shows a differential scanning calorimetry (DSC) thermogram of palbociclib glutarate Form B.
FIG. 40 shows a thermogravimetric analysis (TGA) thermogram of palbociclib glutarate Form B.
FIG. 41 shows an X-ray powder diffraction (XRPD) pattern of palbociclib adipate Form A.
FIG. 42 shows a differential scanning calorimetry (DSC) thermogram of adipate
Form A.
FIG. 43 shows a thermogravimetric analysis (TGA) thermogram of palbociclib adipate Form A.
FIG. 44 shows an X-ray powder diffraction (XRPD) pattern of palbociclib adipate Form B.
FIG. 45 shows an X-ray powder diffraction (XRPD) pattern of palbociclib glycolate Form A.
FIG. 46 shows a differential scanning calorimetry (DSC) thermogram of glycolate Form A.
FIG. 47 shows a thermogravimetric analysis (TGA) thermogram of palbociclib glycolate Form A.
FIG. 48 shows an X-ray powder diffraction (XRPD) pattern of palbociclib diesylate Form A.
FIG. 49 shows an X-ray powder diffraction (XRPD) pattern of palbociclib diesylate Form B.
FIG. 50 shows an X-ray powder diffraction (XRPD) pattern of palbociclib diesylate Form C. FIG. 51 shows an X-ray powder diffraction (XRPD) pattern of palbociclib hippurate Form A.
FIG. 52 shows a differential scanning calorimetry (DSC) thermogram of palbociclib hippurate Form A.
FIG. 53 shows an X-ray powder diffraction (XRPD) pattern of palbociclib hippurate
Form B.
FIG. 54 shows a differential scanning calorimetry (DSC) thermogram of hippurate Form B.
FIG. 55 shows an X-ray powder diffraction (XRPD) pattern of palbociclib esylate Form A.
FIG. 56 shows a differential scanning calorimetry (DSC) thermogram of palbociclib esylate Form A.
FIG. 57 shows an X-ray powder diffraction (XRPD) pattern of palbociclib esylate Form B.
FIG. 58 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate
Form a.
FIG. 59 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form a.
FIG. 60 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate Form β.
FIG. 61 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate Form γ.
FIG. 62 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form γ.
FIG. 63 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate
Form δ.
FIG. 64 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form δ.
FIG. 65 shows an X-ray powder diffraction (XRPD) pattern of palbociclib isethionate Form ε.
FIG. 66 shows a differential scanning calorimetry (DSC) thermogram of palbociclib isethionate Form ε. FIG. 67 shows comparison of the XRPD pattern of palbociclib crystalline Form I after being stored at 25°C/60%RH for 225 days with its initial XPRD pattern, the initial XRPD pattern shown above, and the XRPD pattern after being stored at 25°C/60%RH for 225 days shown below.
FIG. 68 shows comparison of the XRPD pattern of palbociclib crystalline Form I after being stored at 40°C/75%RH for 225 days with its initial XPRD pattern, the initial XRPD pattern shown above, and the XRPD pattern after being stored at 40°C/75%RH for 225 days shown below.
FIG. 69 shows the XRPD pattern of palbociclib crystalline Form I converted from Form A in dichloromethane at 50 °C.
FIG. 70 shows the XRPD pattern of palbociclib crystalline Form I converted from Form A in dichloromethane/methanol/water mixture at room temperature.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is based on a surprising discovery that palbociclib can exist in different crystalline forms and salts, and these forms and salts can be prepared readily from environmentally friendly solvent systems using convenient methods.
In an aspect, the present invention provides a crystalline form of palbociclib, designated as Form I.
In one embodiment, the crystalline Form I is characterized by an X-ray powder diffraction pattern comprising the following 2Θ values measured using CuKa radiation: 5.1°±0.2°, 9.8°±0.2°, and 19.0°±0.2°.
In another embodiment, the crystalline Form I is characterized by an X-ray powder diffraction pattern further comprising the following 2Θ values measured using CuKa radiation: 11.5°±0.2°, 9.5°±0.2°, and 22.2°±0.2°.
In another embodiment, the crystalline Form I is characterized by an X-ray powder diffraction pattern further comprising the following 2Θ values measured using CuKa radiation: 7.9°±0.2°, 12.2°±0.2°, 14.8°±0.2°, and 16.3°±0.2°.
In another embodiment, the crystalline Form I is characterized by an X-ray powder diffraction pattern comprising the following 2Θ values measured using CuKa radiation: 5.1°±0.2°, 7.9°±0.2°, 9.5°±0.2°, 9.8°±0.2°, 11.5°±0.2°, 12.2°±0.2°, 14.8°±0.2°, 16.3°±0.2°, 19.0°±0.2°, and 22.2°±0.2°.
In another embodiment, the crystalline Form I has an X-ray powder diffraction pattern substantially as shown in FIG. 1.
In another embodiment, the crystalline Form I has a differential scanning calorimetry thermogram substantially as shown in FIG. 2, which exhibits an endothermic peak temperature of about 260.5 °C.
In another embodiment, the crystalline Form I has a thermal gravimetric analysis thermogram as shown in FIG. 3, which exhibits about 0.8% weight loss when heated up to 220 °C.
In some embodiment, Form I is unsolvated. In some embodiment, Form I is anhydrous.
In another aspect, the present invention provides a crystalline form of palbociclib, designated as Form II.
In one embodiment, the crystalline Form II is characterized by an X-ray powder diffraction pattern comprising the following 2Θ values measured using CuKa radiation: 9.4°±0.2°, 12.6°±0.2°, and 17.5°±0.2°.
In another embodiment, the X-ray powder diffraction pattern of crystalline Form II further comprises the following 2Θ values measured using CuKa radiation: 7.3°±0.2°, 10.9°±0.2°, and 16.4°±0.2°.
In another embodiment, the X-ray powder diffraction pattern of crystalline Form II further comprises the following 2Θ values measured using CuKa radiation: 11.7o±0.2°, 19.3°±0.2°, 22.5°±0.2°, and 23.7°±0.2°.
In another embodiment, the X-ray powder diffraction pattern of crystalline Form II comprises the following 2Θ values measured using CuKa radiation: 7.3°±0.2°, 9.4°±0.2°, 10.9°±0.2°, 11.7°±0.2°, 12.6°±0.2°, 16.4°±0.2°, 17.5°±0.2°, 19.3°±0.2°, 22.5°±0.2°, and 23.7°±0.2°.
In another embodiment, the crystalline Form II has an X-ray powder diffraction pattern substantially as depicted in FIG. 4.
In another aspect, the present invention provides processes for preparation of palbociclib Forms I and II, which comprises crystallizing palbociclib in an aid of one or two crystallization solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ester solvents, ether solvents, aromatic solvents, nitrile solvents, haloalkane and water via a crystallization method selected from slurry, evaporation, cooling, anti-solvent addition, with or without seeding.
In a preferred embodiment, the solvent is dichloromethane, the crystallization method is slurry and Form I is obtained.
In a preferred embodiment, the solvent is a mixture of ethanol/water, the crystallization method is slow evaporation of the solvents, and Form II is obtained. In a more preferred embodiment, the mixture of ethanol/water is in 6: 1 (v/v) ratio.
In another aspect, the present invention provides novel pharmaceutical salts of palbociclib, particularly isethionate, sulfate, phosphate, acetate, L-lactate, maleate, fumarate, citrate, succinate, L-tartrate, hippurate, glutarate, adipate, glycolate, esylate and diesylate.
In further aspects, the present invention provides crystalline forms of the
pharmaceutical salts of palbociclib, particularly isethionate Forms α, β, γ, δ, and ε, sulfate Form A, phosphate Form A, acetate Forms A and B, L-lactate Form A, maleate Forms A and B, fumarate Forms A and B, citrate Forms A and B, succinate Forms A, B, and C, L- tartrate Forms A and B, glutarate Forms A and B, adipate Forms A and B, glycolate Form A, diesylate Forms A, B, and C, hippurate Forms A and B, and esylate Forms A and B.
In further aspects, the crystalline forms of palbociclib salts are characterized by an X- ray powder diffraction pattern comprising the following 2Θ values:
Figure imgf000010_0001
succinate Form A 9.2, 18.7, 4.2, 24.6, 19.3, 21.3, 13.9, 10.6, 8.3 succinate Form B 7.0, 17.2, 4.8, 4.2, 8.0, 21.1, 10.5, 13.9, 18.5 succinate Form C 4.2, 12.8, 18.9, 10.8, 16.8, 8.5, 12.3, 9.4, 21.4
L-tartrate Form A 4.2, 10.5, 6.8, 4.8, 15.5, 12.6, 17.8, 13.9, 21.2
L-tartrate Form B 10.9, 3.1, 12.0, 19.3, 17.2, 4.0, 9.0, 11.5, 21.9 glutarate Form A 17.3, 13.0, 9.5, 19.8, 9.8, 7.7, 15.2, 26.3, 14.8 glutarate Form B 6.9, 16.9, 10.0, 10.1, 13.9, 20.4, 11.7, 4.9, 7.7 adipate Form A 4.3, 9.2, 13.0, 19.5, 14.9, 19.4, 17.6, 18.4, 11.4 adipate Form B 11.7, 3.9, 10.9, 7.8, 19.5, 21.8, 16.3, 13.9, 8.5 glycolate Form A 10.8, 9.2, 13.8, 4.6, 19.5, 18.4, 22.9, 21.7, 14.4 diesylate Form A 6.0, 9.0, 15.0, 18.2, 21.1, 23.8
diesylate Form B 6.8, 3.4, 17.1, 16.8, 13.6, 20.5, 24.1, 11.9, 10.1 diesylate Form C 5.8, 6.8, 8.8, 22.2, 7.3, 11.7, 13.6, 16.5, 11.0 hippurate Form A 5.2, 5.4, 10.8, 10.1, 8.4, 20.9, 15.2, 15.8, 12.9 hippurate Form B 6.0, 4.2, 4.8, 7.5, 18.3, 10.7, 12.4, 9.7, 15.8 esylate Form A 6.4, 4.3, 9.8, 15.0, 12.9, 16.8, 17.2, 25.9, 8.6 esylate Form B 9.0, 8.5, 19.3, 5.0, 10.1, 11.0, 4.2, 22.1, 12.6 isethionate Form a 17.9, 18.2, 22.9,9.6, 14.9, 26.8,10.0,12.5, 19.1 isethionate Form β 8.0, 10.3, 18.6,10.1, 15.9, 18.3,9.2, 11.9,18.0 isethionate Form γ 15.8, 22.6, 23.8,10.7, 20.8, 22.0,14.6, 25.2, 26.9 isethionate Form δ 7.9, 10.8, 18.6,11.8, 15.8, 18.0,14.3, 17.5, 29.0 isethionate Form ε 3.4, 10.0,18.3,9.1, 18.8, 22.1,4.0, 6.7, 25.5 wherein 2-theta values are each variable within ±0.2°.
In another aspect, the present invention provides processes for preparation of palbociclib salts, which comprises the step of reacting the freebase of palbociclib with the corresponding acid in the presence of suitable solvent or mixture of solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ether solvents, ester solvents and water.
In another aspect, the present invention provides processes for preparation of crystalline forms of palbociclib isethionate, which comprises reacting palbociclib freebase with 2-hydroethanolsulfonic acid or crystallizing palbociclib isethionate in one or two crystallization solvents selected from the group consisting of alcoholic solvents, alkylketone solvents, ester solvents, ether solvents, aromatic solvents, nitrile solvents, haloalkane and water through a crystallization method selected from slurrying, evaporating solvents, cooling, adding anti-solvent(s), with or without seeding.
In one embodiment, the solvent is acetonitrile, the crystallization method is reacting palbociclib freebase with 2-hydroethanolsulfonic acid, and Form a is obtained.
In another embodiment, the solvent is ethanol, the crystallization method is slow evaporation of solvent(s), and Form β is obtained.
In another embodiment, the solvent is Ν,Ν-Dimethylformamide, the crystallization method is slurrying, and Form γ is obtained.
In another embodiment, the solvent is a mixture ethanol/water, the crystallization method is cooling, and Form δ is obtained. In a preferred embodiment, mixture ethanol/water is in 19: 1 (v/v) ratio.
In another embodiment, the solvent is a mixture of 1 ,4-dioxane/water, the crystallization method is cooling, and Form ε is obtained. In a preferred embodiment, the mixture of ethanol/water is in 133:21 (v/v) ratio.
In another aspect, the present invention provides solid pharmaceutical compositions, comprising, as an active ingredient, any one or combination of the crystalline forms of palbociclib and/or crystalline forms of palbociclib salts described herein.
In another aspect, the present invention provides a method of treating or delaying the progression or onset of a disease or disorder in connection with activity of cyclin- dependent kinase (CDK) 4/6 inhibitor, comprising administering to a subject in need thereof a therapeutically effective amount of palbociclib selected from the group consisting of any one or combination of the crystalline forms of palbociclib and/or crystalline forms of palbociclib salts described herein.
In a preferred embodiment, said disease or disorder is selected from the group consisting of breast cancer, ovary cancer, cervix cancer, prostate cancer, testis cancer, esophagus cancer, stomach cancer, skin cancer, and lung cancer.
In another aspect, the present invention provides use of palbociclib selected from the group consisting of any one or combination of the crystalline forms of palbociclib and/or crystalline forms of palbociclib salts described herein in the manufacture of a medicament for treating or delaying the progression or onset of a disease or disorder in connection with activity of cyclin-dependent kinase (CDK) 4/6 inhibitor. In a preferred embodiment, said disease or disorder is selected from the group consisting of breast cancer, ovary cancer, cervix cancer, prostate cancer, testis cancer, esophagus cancer, stomach cancer, skin cancer, and lung cancer.
The terms in the present invention, if not specifically defined, take their ordinary meanings as would be understood by those skilled in the art.The term "alcohol," "alcoholic solvent," or the like, refers to Ci-C6 alkyl alcohol, preferably C1-C4 alkyl alcohol, for example, in some embodiments preferably, methanol, ethanol, isopropanol, or the like.
The term "ketone," "alkylketone," or the like, refers to C3-C7 alkanone, having a formula RCOR', wherein R and R' are each independently C1-C4 alkyl, for example, in some embodiments preferably, acetone, butanone, 2-pentanone, 3-pentanone, methyl isobutyl ketone (MIBK), or the like.
The term "ester," or the like, refers to a lower alkyl aliphatic acid ester having a formula RCOOR', wherein R and R' are each independently C1-C4 alkyls, for example, in some embodiments preferably, ethyl acetate, ethyl propionate, methyl acetate, propyl acetate, ispropyl acetate, or the like.
The term "ether," or the like, refers to a lower alkyl ether or cyclic ether (each alkyl having 1 to 4 carbon atoms), including but not limited to diethyl ether, di-isopropyl ether, ethyl propyl ether, methyl t-butyl ether (MTBE), tetrahydrofuran (THF), 1,4-dioxane, or the like.
The term "aromatic hydrocarbon," or the like, refers to benzene optionally substituted by 1 to 3 methyl or ethyl groups, for example, in some embodiments preferably, toluene, 1,2-xylene, 1,4-xylene, 1 ,3-xylene, cumene, ethylbenzene, or the like.
The term "halogenated hydrocarbon," or the like, refers to Ci-C6 alkane substituted by one to six, preferably one to four, F and/or CI atoms, for example, in some embodiments preferably, dichloromethane, chloroform, 1,1,1-trifluoroethane, or the like.The term "nitrile," "nitrile solvent," or the like, refers to C2-C4 alkyl nitrile, i.e., CH3CN, CH3CH2CN, CH3CH2CH2CN, or CH3CH(CN)CH3, preferably CH3CN.
The term "treatment" or "treating" refers to the management and care of a patient for the purpose of combating the disease, condition or disorder. The term "therapeutically effective amount" refers to an amount of a drug or a therapeutic agent that will elicit the desired biological and/or medical response of a tissue, system or an animal (including man) that is being sought by a researcher or clinician.
The term "subject" or "patient" refers to a mammalian animal.
The term "mammal" or "mammalian animal" includes, but is not limited to, humans, dogs, cats, horses, pigs, cows, monkeys, rabbits and mice. The preferred mammals are humans.
The term "administering" means applying a compound of the invention, or a pharmaceutically acceptable salt, pro-drug or composition thereof, to a subject in need of treatment. The administration of the composition of the present invention in order to practice the present methods of therapy is carried out by administering a therapeutically effective amount of the compounds in the composition to a subject in need of such treatment or prophylaxis. The need for a prophylactic administration according to the methods of the present invention is determined via the use of well-known risk factors. The effective amount of an individual compound is determined, in the final analysis, by the physician in charge of the case, but depends on factors such as the exact disease to be treated, the severity of the disease and other diseases or conditions from which the patient suffers, the chosen route of administration, other drugs and treatments which the patient may concomitantly require, and other factors in the physician's judgment.
The term "pharmaceutically acceptable", as used herein, refers to those compounds, materials, compositions and/or dosage forms, which are, within the scope of sound medical judgment, suitable for contact with the tissues of mammals, especially humans, without excessive toxicity, irritation, allergic response and other problem complications commensurate with a reasonable benefit/risk ratio.
When the term "about" is applied to a parameter, such as amount, temperature, time, or the like, it indicates that the parameter can usually vary by ±10%, preferably within ±5%, and more preferably within ±2%. However, in the case of a melting or onset temperature of a crystalline form as measured by in a DSC thermogram, the term "about" may indicate that the melting or onset temperature can usually vary within ±2°C, regardless of the absolute value of the melting or onset temperature, as a person skilled in the art would understand it. As would be understood by a person skilled in the art, when a parameter is not critical, a number is often given only for illustration purpose, instead of being limiting.
The term "a," "an," or "the," as used herein, represents both singular and plural forms. In general, when either a singular or a plural form of a noun is used, it denotes both singular and plural forms of the noun.
The following non-limiting examples further illustrate certain aspects of the present invention.
EXAMPLES
GENERAL METHODS
X-ray Powder diffraction (XRPD)
XRPD was performed with Panalytical Empyrean XRPD on a Si single crystal holder. The 2Θ position was calibrated against Panalytical 640 Si powder standard. Details of XRPD method used in the experiments are listed below.
Parameters Settings/V alues (Reflection Mode)
Cu, ka,
X-Ray wavelength Kal (A): 1.540598, Ka2 (A): 1.544426
Ka2/Kal intensity ratio: 0.50
X-Ray tube setting 45 kV, 40 mA
Divergence slit Automatic
Scan mode Continuous
Scan speed (°/min) About 10 Persons skilled in the art of X-ray powder diffraction will realize that the relative intensity of peaks can be affected by, for example, grains above 30 microns in size and non-unitary aspect ratios that may affect analysis of samples. The skilled person will also realize that the position of reflections can be affected by the precise height at which the sample sits in the diffractometer and the zero calibration of the diffractometer. The surface planarity of the sample may also have a small effect. Hence, the diffraction pattern data presented are not to be taken as absolute values.
When an XRPD pattern of a crystal form is described as comprising certain "representative" or "characteristic" peaks or 2Θ values, it refers to more prominent peaks, or a subset thereof, in the XRPD pattern. Typically, "characteristic peaks" are defined as a subset of representative (prominent) peaks used to differentiate one crystalline polymorph or form from another crystalline polymorph or form. Characteristic peaks may be determined by evaluating which representative peaks, if any, are present in one crystalline polymorph of a compound but not in all other known crystalline polymorphs of that compound. However, not all crystalline polymorphs of a compound would necessarily have at least one characteristic peak. As a person of ordinary skill in the art would understand, in certain situations, the overall diffraction pattern should be used to determine whether a crystal form exists as described or claimed.
Differential Scanning Calorimetry (DSC)
Analytical Instrument: TA Instruments Q2000 DSC.
Heating rate: 10 °C per minute.
Purge gas: nitrogen
Thermal Gravimetric Analysis (TGA)
Analytical Instrument: TA Instruments Q500 TGA.
Heating rate: 10 °C per minute.
Purge gas: nitrogen.
Example 1. Preparation of palbociclib crystalline Form I
To 12.0 mL of dichloromethane was added 202.3 mg of palbociclib. The mixture was stirred at 90 °C for 24 hours. The solid was isolated and Form I was obtained, which was analyzed by XRPD, DSC, and TGA. The XRPD data of the Form I obtained in this example are listed in Table 1.
The XRPD pattern, DSC thermogram, and TGA thermogram of Form I obtained from this example are displayed in FIGs. 1, 2, and 3, respectively. The thermal gravimetric analysis of the sample showed weight loss of only about 0.8% when heated to 220 °C, indicating that the sample is not a solvate, but anhydrous.
Table 1
Figure imgf000016_0001
9.83 9.00 30.86
10.31 8.58 6.84
11.46 7.72 21.77
12.16 7.28 7.01
13.16 6.73 6.84
14.82 5.98 11.09
15.56 5.70 5.16
16.34 5.43 8.31
17.57 5.05 6.90
19.04 4.66 33.31
19.76 4.49 5.20
22.16 4.01 11.14
Example 2. Preparation of palbociclib crystalline Form II
To 3.5 mL of ethanol/water (6/1, v/v) was added 10.3 mg of palbociclib. The mixture was placed at 80 °C hot-stage plate for 3 hours and filtered by 0.45 um filter, and the clear filtrate was evaporated slowly at 80 °C until precipitation. The solid was isolated and Form II was obtained, which was analyzed by XRPD. The XRPD data of the Form II obtained in this example are listed in Table 2.
The XRPD pattern of Form II obtained from this example is displayed in FIG. 4.
Table 2
Figure imgf000017_0001
16.35 5.42 25.92
17.51 5.06 76.27
19.30 4.60 23.10
22.47 3.96 22.86
23.67 3.76 17.71
25.39 3.51 7.25
31.78 2.82 7.05
Example 3. Preparation of palbociclib sulfate
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 0.6 μΙ_, of sulphuric acid (98 wt%). The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and sulfate Form A was obtained, which was analyzed by XRPD. The XRPD data of palbociclib sulfate Form A obtained in this example are listed in Table 3.
The XRPD pattern of sulfate Form A obtained from this example are displayed in FIG. 5.
Table 3
Figure imgf000018_0001
18.24 4.86 28.66
19.57 4.54 20.94
21.65 4.11 10.62
Example 4. Preparation of palbociclib phosphate
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 0.65 μΐ, of phosphoric acid (85 wt%). The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and phosphate Form A was obtained, which was analyzed by XRPD. The XRPD data of palbociclib phosphate Form A obtained in this example are listed in Table 4.
The XRPD pattern of phosphate Form A obtained from this example is displayed in FIG. 6.
Table 4
Figure imgf000019_0001
Example 5. Preparation of palbociclib acetate Form A
To 20.0 mL of acetonitrile was added 95.9 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 0.67 of acetic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and acetate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib acetate Form A obtained in this example are listed in Table 5.
The XRPD pattern and DSC thermogram of acetate Form A obtained from this example are displayed in FIGs. 7 and 8.
Table 5
Figure imgf000020_0001
Example 6. Preparation of palbociclib acetate Form B
To 20.0 mL of acetone was added 96.1 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 0.67 μΐ, of acetic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and acetate Form B was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib acetate Form B obtained in this example are listed in Table 6.
The XRPD pattern and DSC thermogram of acetate Form B obtained from this example are displayed in FIGs. 9 and 10.
Table 6
Figure imgf000020_0002
10.02 8.83 30.95
11.65 7.60 64.86
13.45 6.58 35.30
15.36 5.77 3.50
16.28 5.44 3.00
17.43 5.09 58.06
18.68 4.75 25.32
19.44 4.57 8.74
20.11 4.42 8.41
21.72 4.09 24.07
22.56 3.94 5.49
23.44 3.80 5.76
27.11 3.29 6.35
Example 7. Preparation of palbociclib L-lactate Form A
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 0.84 μΐ, of lactic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and L-lactate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib L-lactate Form A obtained in this example are listed in Table 7.
The XRPD pattern and DSC thermogram of L-lactate Form A obtained from this example are displayed in FIGs. 11 and 12.
Table 7
Figure imgf000021_0001
16.85 5.26 12.35
19.06 4.66 31.87
21.76 4.08 9.70
Example 8. Preparation of palbociclib maleate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.31 mg of maleic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and maleate Form A was obtained, which was analyzed by XRPD, DSC and TGA. The XRPD data of palbociclib maleate Form A obtained in this example are listed in Table 8.
The XRPD pattern, DSC thermogram and TGA thermogram of maleate Form A obtained from this example is displayed in FIGs. 13, 14 and 15.
Table 8
Figure imgf000022_0001
Example 9. Preparation of palbociclib maleate Form B
To 20.0 mL of acetonitrile was added 95.9 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.35 mg of maleic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and maleate Form B was obtained, which was analyzed by XRPD. The XRPD data of palbociclib maleate Form B obtained in this example are listed in Table 9.
The XRPD pattern and DSC thermogram of maleate Form B obtained from this example is displayed in FIGs. 16 and 17.
Table 9
Figure imgf000023_0001
Example 10. Preparation of palbociclib fumarate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.30 of fumaric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and fumarate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib fumarate Form A obtained in this example are listed in Table 10.
The XRPD pattern and DSC thermogram of fumarate Form A obtained from this example are displayed in FIGs. 18 and 19. Table 10
Figure imgf000024_0001
Example 11. Preparation of palbociclib fumarate Form B
To 20.0 mL of acetonitrile was added 95.9 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.32 of fumaric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and fumarate Form B was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib fumarate Form B obtained in this example are listed in Table 1 1.
The XRPD pattern and DSC thermogram of fumarate Form B obtained from this example are displayed in FIGs. 20 and 21.
Table 11
Figure imgf000024_0002
12.54 7.06 33.86
15.89 5.58 100.00
19.59 4.53 43.09
25.13 3.54 12.91
26.99 3.30 5.07
Example 12. Preparation of palbociclib citrate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 2.17 of citric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and citrate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib citrate Form A obtained in this example are listed in Table 12.
The XRPD pattern and DSC thermogram of citrate Form A obtained from this example are displayed in FIGs. 22 and 23.
Table 12
Figure imgf000025_0001
20.76 4.28 18.96
21.14 4.20 6.13
Example 13. Preparation of palbociclib citrate Form B
To 20.0 mL of tetrahydrofuran/water (19: 1, v/v) was added 95.3 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 2.20 of citric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and citrate Form B was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib citrate Form B obtained in this example are listed in Table 13.
The XRPD pattern and DSC thermogram of citrate Form B obtained from this example are displayed in FIGs. 24 and 25.
Table 13
Figure imgf000026_0001
Example 14. Preparation of palbociclib succinate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.31 mg of succinic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and succinate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib succinate Form A obtained in this example are listed in Table 14. The XRPD pattern and DSC thermogram of succinate Form A obtained from this example are displayed in FIGs. 26 and 27.
Table 14
Figure imgf000027_0001
Example 15. Preparation of palbociclib succinate Form B
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.28 mg of succinic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and succinate Form B was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib succinate Form B obtained in this example are listed in Table 15.
The XRPD pattern and DSC thermogram of succinate Form B obtained from this example are displayed in FIGs. 28 and 29. Table 15
Figure imgf000028_0001
Example 16. Preparation of palbociclib succinate Form C
To 20.0 mL of acetonitrile was added 95.9 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.35 mg of succinic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and succinate Form C was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib succinate Form C obtained in this example are listed in Table 16.
The XRPD pattern and DSC thermogram of succinate Form C obtained from this example are displayed in FIGs. 30 and 31.
Table 16
Figure imgf000028_0002
12.82 6.91 46.52
14.14 6.27 10.92
16.84 5.26 26.74
18.86 4.71 34.80
21.42 4.15 12.50
23.30 3.82 2.93
Example 17. Preparation of palbociclib L-tartarate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.71 mg of L-tartaric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and L- tartrate Form A was obtained, which was analyzed by XRPD, DSC and TGA. The XRPD data of palbociclib L-tartrate Form A obtained in this example are listed in Table 17.
The XRPD pattern, DSC thermogram and TGA thermogram of L-tartrate Form A obtained from this example are displayed in FIGs. 32, 33 and 34.
Table 17
Figure imgf000029_0001
Example 18. Preparation of palbociclib L-tartrate Form B
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.78 mg of L-tartaric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and L- tartrate Form B was obtained, which was analyzed by XRPD. The XRPD data of palbociclib L-tartrate Form B obtained in this example are listed in Table 18.
The XRPD pattern of L-tartrate Form B obtained from this example is displayed in FIG. 35.
Table 18
Figure imgf000030_0001
Example 19. Preparation of palbociclib glutarate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 2.08 mg of glutaric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and glutarate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib glutarate Form A obtained in this example are listed in Table 19.
The XRPD pattern and DSC thermogram of glutarate Form A obtained from this example are displayed in FIGs. 36 and 37.
Table 19
Figure imgf000030_0002
9.83 9.00 33.56
13.02 6.80 74.46
14.78 5.99 2.32
15.25 5.81 4.94
17.29 5.13 100.00
19.81 4.48 41.93
22.43 3.96 1.71
26.28 3.39 4.13
Example 20. Preparation of palbociclib glutarate Form B
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 2.11 mg of glutaric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and glutarate Form B was obtained, which was analyzed by XRPD, DSC and TGA. The XRPD data of palbociclib glutarate Form B obtained in this example are listed in Table 20.
The XRPD pattern, DSC thermogram and TGA thermogram of glutarate Form B obtained from this example are displayed in FIGs. 38, 39 and 40.
Table 20
Figure imgf000031_0001
18.86 4.71 4.06
20.10 4.42 5.31
20.43 4.35 13.59
Example 21. Preparation of palbociclib adipate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.47 mg of adipic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and adipate Form A was obtained, which was analyzed by XRPD, DSC and TGA. The XRPD data of palbociclib adipate Form A obtained in this example are listed in Table 21.
The XRPD pattern, DSC thermogram and TGA thermogram of adipate Form A obtained from this example are displayed in FIGs. 41, 42 and 43.
Table 21
Figure imgf000032_0001
22.90 3.88 4.61
23.69 3.76 1.46
24.37 3.65 2.32
25.82 3.45 4.45
26.26 3.39 1.66
27.87 3.20 1.10
Example 22. Preparation of palbociclib adipate Form B
To 20.0 mL of acetonitrile was added 95.9 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.49 mg of adipic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and adipate Form B was obtained, which was analyzed by XRPD. The XRPD data of palbociclib adipate Form B obtained in this example are listed in Table 22.
The XRPD pattern of adipate Form B obtained from this example is displayed in FIG.
44.
Table 22
Figure imgf000033_0001
Example 23. Preparation of palbociclib glycolate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 2.08 mg of glycolic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and glycolate Form A was obtained, which was analyzed by XRPD, DSC and TGA. The XRPD data of palbociclib glycolate Form A obtained in this example are listed in Table 23.
The XRPD pattern, DSC thermogram and TGA thermogram of glycolate Form A obtained from this example are displayed in FIGs. 45, 46 and 47.
Table 23
Figure imgf000034_0001
Example 24. Preparation of palbociclib diesylate Form A
To 20.0 mL of methanol was added 95.5 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.31 mg of 1 ,2-ethanedisulfonic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and diesylate Form A was obtained, which was analyzed by XRPD. The XRPD data of palbociclib diesylate Form A obtained in this example are listed in Table 24.
The XRPD pattern of diesylate Form A obtained from this example are displayed in FIG. 48.
Table 24
Figure imgf000034_0002
18.16 4.88 1.84
21.10 4.21 0.73
23.83 3.73 0.57
Example 25. Preparation of palbociclib diesylate Form B
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.30 mg of 1,2-ethanedisulfonic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and diesylate Form B was obtained, which was analyzed by XRPD. The XRPD data of palbociclib diesylate Form B obtained in this example are listed in Table 25.
The XRPD pattern of diesylate Form B obtained from this example are displayed in FIG. 49.
Table 25
Figure imgf000035_0001
Example 26. Preparation of palbociclib diesylate Form C
To 20.0 mL of tetrahydrofuran/water (19: 1, v/v) was added 95.3 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.29 mg of 1,2-ethanedisulfonic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and diesylate Form C was obtained, which was analyzed by XRPD. The XRPD data of palbociclib diesylate Form C obtained in this example are listed in Table 26. The XRPD pattern of diesylate Form C obtained from this example are displayed in FIG. 50.
Table 26
Figure imgf000036_0001
Example 27. Preparation of palbociclib hippurate Form A
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.53 mg of hippuric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and hippurate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib hippurate Form A obtained in this example are listed in Table 27.
The XRPD pattern and DSC thermogram of hippurate Form A obtained from this example are displayed in FIGs. 51 and 52.
Table 27
Figure imgf000036_0002
15.17 5.84 7.50
15.76 5.62 5.79
20.90 4.25 7.95
21.60 4.11 2.35
Example 28. Preparation of palbociclib hippurate Form B
To 20.0 mL of acetonitrile was added 95.9 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.58 mg of hippuric acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and hippurate Form B was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib hippurate Form B obtained in this example are listed in Table 28.
The XRPD pattern and DSC thermogram of hippurate Form B obtained from this example are displayed in FIGs. 53 and 54.
Table 28
Figure imgf000037_0001
Example 29. Preparation of palbociclib esylate Form A
To 20.0 mL of isopropanol was added 95.6 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.15 mg of ethanesulfonic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and esylate Form A was obtained, which was analyzed by XRPD and DSC. The XRPD data of palbociclib esylate Form A obtained in this example are listed in Table 29.
The XRPD pattern and DSC thermogram of esylate Form A obtained from this example are displayed in FIGs. 55 and 56.
Table 29
Figure imgf000038_0001
Example 30. Preparation of palbociclib esylate Form B
To 20.0 mL of ethyl acetate was added 94.8 mg of palbociclib. 1.0 mL of the solution was added to a glass vial, followed by the addition of 1.10 of ethanesulfonic acid. The mixture was stirred under ambient conditions for 24 hours. The solid was isolated and esylate Form B was obtained, which was analyzed by XRPD. The XRPD data of palbociclib esylate Form B obtained in this example are listed in Table 30.
The XRPD pattern of esylate Form B obtained from this example are displayed in FIG. 57. Table 30
Figure imgf000039_0001
Example 31. Preparation of palbociclib isethionate Form a
To 15.0 mL of acetonitrile was added 1.0 g of palbociclib freebase. 300 μΐ, of 2- hydroethanolsulfonic acid (80wt%) was added to 3 mL of acetonitrile. Mix the two solutions by adding the acid solution dropwise. Stir the suspension at 800 rpm overnight, then centrifuge it and isethionate Forma was produced, which was analyzed by XRPD and DSC. The XRPD data of the palbociclib isethionate Form a obtained in this example are listed in Table 31. The XRPD pattern and DSC thermogram of palbociclib isethionate Form a obtained from this example are displayed in FIGs. 58 and 59, respectively. DSC thermogram exhibits two endothermic peaks with onset temperature of about 62.0 °C and 264.2 °C.
Table 31
Figure imgf000040_0001
27.84 3.20 6.53
28.24 3.16 3.21
29.23 3.06 8.68
29.71 3.01 7.48
Example 32. Preparation of palbociclib isethionate Form β
To 3.5 mL of ethanol was added 6.3 mg of palbociclib isethionate. The mixture was filtered by 0.45 μιη filter after stirred at 50 °C for 2 hours. The filtrate was slowly evaporated at RT until precipitation, and palbociclib isethionate Form β was obtained, which was analyzed by XRPD. The XRPD data of the palbociclib isethionate Form β obtained in this example are listed in Table 32.
The XRPD pattern of palbociclib isethionate Form β obtained from this example is displayed in FIG. 60.
Table 32
Figure imgf000041_0001
18.32 4.84 31.50
18.62 4.76 59.03
19.35 4.59 12.76
19.76 4.49 13.69
20.25 4.39 11.35
21.47 4.14 10.13
22.21 4.00 16.80
23.26 3.82 11.34
24.68 3.61 8.48
25.64 3.48 9.21
26.92 3.31 3.27
27.55 3.24 4.64
30.90 2.89 2.40
32.95 2.72 3.95
Example 33. Preparation of palbociclib isethionate Form γ
To 0.2 mL of Ν,Ν-Dimethylformamide was added 11.9 mg of palbociclib isethionate. The mixture was stirred under ambient conditions overnight. The suspension was isolated by centrifugation and palbociclib isethionate Form γ was obtained, which was analyzed by XRPD and DSC. The XRPD data of the palbociclib isethionate Form γ obtained in this example are listed in Table 33.
The XRPD pattern and DSC thermogram of palbociclib isethionate Form γ obtained from this example are displayed in FIGs. 61 and 62, respectively. DSC thermogram exhibits two endothermic peaks with onset temperature of about 139.5 °C and 275.3 °C
Table 33
Figure imgf000042_0001
14.12 6.27 10.52
14.60 6.07 22.03
15.78 5.62 100.00
17.29 5.13 5.12
17.96 4.94 5.54
18.96 4.68 10.77
20.80 4.27 33.13
21.72 4.09 12.23
22.01 4.04 26.68
22.63 3.93 33.67
23.83 3.73 57.02
25.22 3.53 20.37
26.85 3.32 20.21
27.75 3.21 6.21
Example 34. Preparation of palbociclib isethionate Form δ
To 10 mL of ethanol/water (19: 1, v/v) was added 30.9 mg of palbociclib isethionate. The mixture was placed at a 80°C hot-stage plate for 3 hours and filtered by 0.45 μιη filter at 80 °C. the filtrate was crash cooled to -20 °C and equilibrated at -20 °C until precipitation. The precipitate was isolated by centrifugation and palbociclib isethionate Form δ was obtained, which was analyzed by XRPD and DSC. The XRPD data of the palbociclib isethionate Form δ obtained in this example are listed in Table 34.
The XRPD pattern and DSC thermogram of palbociclib isethionate Form δ obtained from this example are displayed in FIGs. 63 and 64. DSC thermogram exhibits two endothermic peaks with onset temperature of about 47.2 °C and 273.9 °C.
Table 34
Figure imgf000043_0001
10.84 8.16 100.00
11.83 7.48 35.24
14.31 6.19 25.06
15.81 5.60 38.54
17.50 5.07 10.32
18.00 4.93 35.36
18.59 4.77 55.88
25.70 3.47 5.83
29.02 3.08 6.72
30.63 2.92 3.86
Example 35. Preparation of palbociclib isethionate Form ε
To 4.0 mL of 1 ,4-dioxane/water (133:21, v/v) was added 40.2 mg of palbociclib isethionate. The mixture was placed at a 80°C hot-stage plate for 3 hours and filtered by 0.45 μιη filter at 80 °C. the filtrate was crash cooled to -20 °C and equilibrated at -20 °C until precipitation. The precipitate was isolated by centrifugation and palbociclib isethionate Form ε was obtained, which was analyzed by XRPD and DSC. The XRPD data of the palbociclib isethionate Form ε obtained in this example are listed in Table 35.
The XRPD pattern and DSC thermogram of palbociclib isethionate Form ε obtained from this example is displayed in FIGs. 65 and 66. DSC thermogram exhibits two endothermic peaks with onset temperature of about 186.0 °C and 276.7 °C.
Table 35
Figure imgf000044_0001
13.38 6.62 18.43
15.95 5.56 13.87
16.76 5.29 13.19
17.64 5.03 11.43
18.26 4.86 83.13
18.58 4.78 69.81
18.81 4.72 40.65
20.14 4.41 21.75
20.64 4.30 6.99
21.44 4.14 8.53
22.14 4.02 38.18
23.15 3.84 17.15
24.55 3.63 8.38
25.50 3.49 35.52
25.90 3.44 9.65
26.81 3.33 5.66
27.52 3.24 9.95
31.26 2.86 7.52
32.35 2.77 3.82
33.92 2.64 2.68
37.37 2.41 5.82
Example 36. Stability of Palbociclib Form I
To obtain the stability data of Form I, a sample of the solid form was placed in a glass vial sealed with a pin-holed parafilm and subsequently stored in a 25°C/60%RH or 40°C/75%RH chamber, respectively. After storage for a period of time, the solid was tested by XRPD.
The XRPD patterns of Form I remain unchanged after storage at 25°C/60%RH or 40°C/75%RH for 225 days, as shown in FIG. 67 and FIG. 68, respectively. These data illustrate the long-term stability of Crystalline Form I of palbociclib under the storage conditions. See Table 36.
Table 36
Figure imgf000046_0001
Example 37. Purity improvement
Using Patent Form A as starting material to generate Form I, the purity was improved as shown in Table 37. Form A (409.9 mg) was added into 20 mL of dichloromethane to suspend in a glass vial, then placed in an oven at 50°C for 7 days stirring (500 r/min), and the solids were isolated (Form I) and analyzed by HPLC. See Table 37.
Table 37
Figure imgf000046_0002
Example 38. Kinetic solubility
To obtain the kinetic solubility data of crystalline Form I, about 10 mg of Form I was added into 2.0 mL of relevant media in a glass vial, and the suspension was subsequently rolling (25 r/min) in an incubator at RT. Then, the suspension was sampled at lh, 4h and 24h, respectively, by filtering each sample with a 0.45 μιη filter, and the supernatants were analyzed by HPLC. The results are shown in Table 38 below:
Table 38
Figure imgf000046_0003
Example 39. Conversion of Form A into Form I at 50°C
Form A (409.9 mg) (see WO2014128588) was added and suspended in 20 mL of dichloromethane in a glass vial, and the suspension then placed in an oven at 50°C for 7 days while stirring (500 r/min). The solids were isolated and analyzed by XRPD, which shows the formation of crystalline Form I. See FIG. 69.
Example 40. Conversion of Form A into Form I at RT
Form A (335.4 mg) was added and suspended in 2.5 mL of dichloromethane/methanol/H20 (V:V:V=18:6: 1) in a glass vial, and the suspension then placed at RT and stirred (500 r/min) for 30 days. The solids were isolated and analyzed by XRPD, which shows the conversion of Form A into Form I. See FIG. 70.
The foregoing examples and description of the preferred embodiments should be taken as illustrating, rather than as limiting, the present invention as defined by the claims. As will be readily appreciated, numerous variations and combinations of the features set forth above can be utilized without departing from the present invention as set forth in the claims.

Claims

I . A crystalline form of palbociclib, designated as Form I, having an X-ray powder diffraction pattern comprising the following 2Θ values measured using CuKa radiation: 5.1°±0.2°, 9.8°±0.2°, and 19.0°±0.2°.
2. The crystalline Form I of claim 1, wherein the X-ray powder diffraction pattern further comprises the following 2Θ values measured using CuKa radiation: 11.5o±0.2°, 9.5°±0.2°, and 22.2°±0.2°.
3. The crystalline Form I of claim 1 or 2, wherein the X-ray powder diffraction pattern further comprises the following 2Θ values measured using CuKa radiation: 7.9°±0.2°, 12.2°±0.2°, 14.8°±0.2°, and 16.3°±0.2°.
4. The crystalline Form I of claim 1, having an X-ray powder diffraction pattern substantially as depicted in FIG. 1.
5. The crystalline Form I of claim 1, having a differential scanning calorimetric thermogram substantially as depicted in FIG. 2.
6. The crystalline Form I of claim 1, having a thermal gravimetric analysis thermogram substantially as depicted in FIG. 3.
7. A crystalline form of palbociclib, designated as Form II, having an X-ray powder diffraction pattern comprising the following 2Θ values measured using CuKa radiation: 9.4°±0.2°, 12.6°±0.2°, and 17.5°±0.2°.
8. The crystalline Form II of claim 7, wherein the X-ray powder diffraction pattern further comprises the following 2Θ values measured using CuKa radiation: 7.3°±0.2°, 10.9°±0.2°, and 16.4°±0.2°.
9. The crystalline Form II of claim 7 or 8, wherein the X-ray powder diffraction pattern further comprises the following 2Θ values measured using CuKa radiation: 11.7°±0.2°, 19.3°±0.2°, 22.5°±0.2°, and 23.7°±0.2°.
10. The crystalline Form II of claim 7, having an X-ray powder diffraction pattern substantially as depicted in FIG. 4.
I I . A process for the preparation of palbociclib Form I or Form II, comprising: crystallizing palbociclib in one or two crystallization solvents selected from the group consisting of alcohols, ketones, esters, ethers, aromatic hydrocarbons, nitriles, halogenated hydrocarbons, and water, through a crystallization method selected from slurrying, evaporating solvent, cooling, adding anti-solvent, with or without seeding, and combinations thereof.
12. The process of claim 1 1 , wherein said solvent is dichloromethane, the crystallization method is slurrying, and crystalline form is Form I.
13. The process of claim 1 1 , wherein said solvent is a mixture of ethanol/water, the crystallization method is slowly evaporating solvent, and the crystalline form is Form II.
14. A pharmaceutical composition comprising a crystalline form of palbociclib according to any one of claims 1 to 10, and a pharmaceutically acceptable carrier.
15. The pharmaceutical composition of claim 14, wherein said crystalline form of palbociclib is Form I.
16. The pharmaceutical composition of claim 14, wherein said crystalline form of palbociclib is Form II.
17. A pharmaceutical salt selected from palbociclib isethionate, sulfate, phosphate, acetate, L-lactate, maleate, fumarate, citrate, succinate, L-tartrate, hippurate, glutarate, adipate, glycolate, esylate, and diesylate.
18. The pharmaceutical salt of claim 17, which is a crystalline form selected from palbociclib isethionate Forms α, β, γ, δ, and ε, sulfate Form A, phosphate Form A, acetate Forms A and B, L-lactate Form A, maleate Forms A and B, fumarate Forms A and B, citrate Forms A and B, succinate Forms A, B and C, L-tartrate Forms A and B, glutarate Forms A and B, adipate Forms A and B, glycolate Form A, diesylate Forms A, B and C, hippurate Forms A and B, and esylate Forms A and B.
19. The pharmaceutical salt of claim 18, wherein each said palbociclib crystalline form is characterized by an X-ray powder diffraction (XRPD) pattern comprising the peaks as shown in the following table:
Figure imgf000049_0001
fumarate Form B 15.9, 11.6, 4.1, 19.6, 7.7, 11.2, 12.5, 8.6, 25.1 citrate Form A 6.7, 4.2, 9.0, 11.0, 10.8, 8.4, 7.9, 5.2, 20.8 citrate Form B 6.0, 8.2, 9.3, 8.7, 4.1, 11.6, 18.2, 14.6, 22.8 succinate Form A 9.2, 18.7, 4.2, 24.6, 19.3, 21.3, 13.9, 10.6, 8.3 succinate Form B 7.0, 17.2, 4.8, 4.2, 8.0, 21.1, 10.5, 13.9, 18.5 succinate Form C 4.2, 12.8, 18.9, 10.8, 16.8, 8.5, 12.3, 9.4, 21.4
L-tartrate Form A 4.2, 10.5, 6.8, 4.8, 15.5, 12.6, 17.8, 13.9, 21.2
L-tartrate Form B 10.9, 3.1, 12.0, 19.3, 17.2, 4.0, 9.0, 11.5, 21.9 glutarate Form A 17.3, 13.0, 9.5, 19.8, 9.8, 7.7, 15.2, 26.3, 14.8 glutarate Form B 6.9, 16.9, 10.0, 10.1, 13.9, 20.4, 11.7, 4.9, 7.7 adipate Form A 4.3, 9.2, 13.0, 19.5, 14.9, 19.4, 17.6, 18.4, 11.4 adipate Form B 11.7, 3.9, 10.9, 7.8, 19.5, 21.8, 16.3, 13.9, 8.5 glycolate Form A 10.8, 9.2, 13.8, 4.6, 19.5, 18.4, 22.9, 21.7, 14.4 diesylate Form A 6.0, 9.0, 15.0, 18.2, 21.1, 23.8
diesylate Form B 6.8, 3.4, 17.1, 16.8, 13.6, 20.5, 24.1, 11.9, 10.1 diesylate Form C 5.8, 6.8, 8.8, 22.2, 7.3, 11.7, 13.6, 16.5, 11.0 hippurate Form A 5.2, 5.4, 10.8, 10.1, 8.4, 20.9, 15.2, 15.8, 12.9 hippurate Form B 6.0, 4.2, 4.8, 7.5, 18.3, 10.7, 12.4, 9.7, 15.8 esylate Form A 6.4, 4.3, 9.8, 15.0, 12.9, 16.8, 17.2, 25.9, 8.6 esylate Form B 9.0, 8.5, 19.3, 5.0, 10.1, 11.0, 4.2, 22.1, 12.6 isethionate Form a 17.9, 18.2, 22.9,9.6, 14.9, 26.8,10.0,12.5, 19.1 isethionate Form β 8.0 , 10.3, 18.6,10.1, 15.9, 18.3,9.2, 11.9,18.0 isethionate Form γ 15.8, 22.6, 23.8,10.7, 20.8, 22.0,14.6, 25.2,26.9 isethionate Form δ 7.9, 10.8, 18.6,11.8, 15.8, 18.0,14.3, 17.5, 29.0 isethionate Form ε 3.4, 10.0,18.3,9.1, 18.8, 22.1,4.0, 6.7, 25.5
wherein the 2-theta values can each vary within ±0.2°.
20. A process for preparing a palbociclib salt, comprising a step of reacting palbociclib freebase with a corresponding acid in the presence of a suitable solvent or mixture of solvents selected from the group consisting of alcohols, ketones, ethers, esters, and water.
21. A process for preparing palbociclib isethionate Form α, β, γ, δ or ε, comprising: reacting palbociclib freebase with 2-hydroethanolsulfonic acid, or crystallizing palbociclib isethionate in one or two crystallization solvents selected from the group consisting of alcohols, ketones, esters, ethers, aromatic hydrocarbons, nitriles, halogenated hydrocarbons, and water, through a crystallization method selected from slurrying, evaporating solvent(s), cooling, adding anti-solvent, with or without seeding, and combinations thereof.
22. The process of claim 21, wherein said solvent is acetonitrile, the crystallization method is reacting palbociclib freebase with 2-hydroethanolsulfonic acid , and the crystalline form is palbociclib isethionate Form a.
23. The process of claim 21, wherein said solvent is ethanol, the crystallization method is slowly evaporating solvent(s), and the crystalline form is palbociclib isethionate Form β.
24. The process of claim 21, wherein said solvent is Ν,Ν-Dimethylformamide, the crystallization method is slurrying, and the crystalline form is palbociclib isethionate Form
Y-
25. The process of claim 21, wherein said solvent is a mixture of ethanol/water, the crystallization method is cooling, and the crystalline form is palbociclib isethionate Form δ.
26. The process of claim 21, wherein said solvent is a mixture of 1 ,4-dioxane/water, the crystallization method is cooling, and the crystalline form is palbociclib isethionate Form ε.
27. A pharmaceutical composition comprising a crystalline form of palbociclib isethionate, and a pharmaceutically acceptable carrier.
28. The pharmaceutical composition of claim 27, wherein said crystalline form of palbociclib isethionate is Form a.
29. The pharmaceutical composition of claim 27, wherein said crystalline form of palbociclib isethionate is Form β.
30. The pharmaceutical composition of claim 27, wherein said crystalline form of palbociclib isethionate is Form γ.
31. The pharmaceutical composition of claim 27, wherein said crystalline form of palbociclib isethionate is Form δ.
32. The pharmaceutical composition of claim 27, wherein said crystalline form of palbociclib isethionate is Form ε.
33. A method of treating or delaying the progression or onset of a disease or disorder in connection with activity of cyclin-dependent kinase (CDK) 4/6 inhibitor, comprising administering to a subject in need thereof a therapeutically effective amount of palbociclib selected from the group consisting of a crystalline Form I according to any one of claims 1 to 6, a crystalline Form II according to any one of claims 7 to 10, a pharmaceutical salt according to any one of claims 17 to 19, a pharmaceutical composition according to any one of claims 14 to 16 and claims 27 to 32.
34. The method of claim 33, wherein said disease or disorder is selected from the group consisting of breast cancer, ovary cancer, cervix cancer, prostate cancer, testis cancer, esophagus cancer, stomach cancer, skin cancer, and lung cancer.
35. Use of a crystalline Form of palbociclib according to any one of claims 1 to 17, and/or a crystalline Form of palbociclib salts according to any one of claims 18 to 32 in the manufacture of a medicament for treating or delaying the progression or onset of a disease or disorder in connection with activity of cyclin-dependent kinase (CDK) 4/6 inhibitor.
36. The use of claim 35, wherein said disease or disorder is selected from the group consisting of breast cancer, ovary cancer, cervix cancer, prostate cancer, testis cancer, esophagus cancer, stomach cancer, skin cancer, and lung cancer.
PCT/US2015/064010 2014-12-05 2015-12-04 Salts and crystalline forms of 6-acetyl-8-cyclopentyl-5-methyl-2((5-(piperazin-1-yl)pyridin-2-yl)amino)pyrido[2,3-d] pyrimidin-7(8h)-one (palbociclib) WO2016090257A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462088438P 2014-12-05 2014-12-05
US62/088,438 2014-12-05

Publications (1)

Publication Number Publication Date
WO2016090257A1 true WO2016090257A1 (en) 2016-06-09

Family

ID=56092531

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/064010 WO2016090257A1 (en) 2014-12-05 2015-12-04 Salts and crystalline forms of 6-acetyl-8-cyclopentyl-5-methyl-2((5-(piperazin-1-yl)pyridin-2-yl)amino)pyrido[2,3-d] pyrimidin-7(8h)-one (palbociclib)

Country Status (1)

Country Link
WO (1) WO2016090257A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106220626A (en) * 2016-07-31 2016-12-14 合肥远志医药科技开发有限公司 A kind of polymorphic of Pabuk former times profit cloth and preparation method thereof
WO2017115315A1 (en) * 2015-12-30 2017-07-06 Dr. Reddy's Laboratories Limited Solid forms of palbociclib
WO2018009735A1 (en) 2016-07-07 2018-01-11 Plantex Ltd. Solid state forms of palbociclib dimesylate
CN108929321A (en) * 2018-07-03 2018-12-04 威海贯标信息科技有限公司 A kind of Pa Boxini novel crystal forms
CN115667260A (en) * 2022-08-26 2023-01-31 成都苑东生物制药股份有限公司 Piperazine Bai Xili saccharine salt crystal form and preparation method thereof
WO2024040668A1 (en) * 2022-08-26 2024-02-29 成都苑东生物制药股份有限公司 Palbociclib saccharinate crystal form and preparation method therefor

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030149001A1 (en) * 2002-01-22 2003-08-07 Mark Barvian 2-(Pyridin-2-ylamino)-pyrido[2,3-d]pyrimidin-7-ones
US20050059670A1 (en) * 2003-07-11 2005-03-17 Pfizer Inc Isethionate salt of a selective CKD4 inhibitor
US20080125588A1 (en) * 2006-09-08 2008-05-29 Pfizer Inc. SYNTHESIS OF 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3-d] PRYIMIDIN-7-ONES
WO2014128588A1 (en) * 2013-02-21 2014-08-28 Pfizer Inc. Solid forms of a selective cdk4/6 inhibitor

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030149001A1 (en) * 2002-01-22 2003-08-07 Mark Barvian 2-(Pyridin-2-ylamino)-pyrido[2,3-d]pyrimidin-7-ones
US20050059670A1 (en) * 2003-07-11 2005-03-17 Pfizer Inc Isethionate salt of a selective CKD4 inhibitor
US20080021037A1 (en) * 2003-07-11 2008-01-24 Pfizer Inc. Isethionate salt of a selective cdk4 inhibitor
US20080125588A1 (en) * 2006-09-08 2008-05-29 Pfizer Inc. SYNTHESIS OF 2-(PYRIDIN-2-YLAMINO)-PYRIDO[2,3-d] PRYIMIDIN-7-ONES
WO2014128588A1 (en) * 2013-02-21 2014-08-28 Pfizer Inc. Solid forms of a selective cdk4/6 inhibitor

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017115315A1 (en) * 2015-12-30 2017-07-06 Dr. Reddy's Laboratories Limited Solid forms of palbociclib
WO2018009735A1 (en) 2016-07-07 2018-01-11 Plantex Ltd. Solid state forms of palbociclib dimesylate
US10597393B2 (en) 2016-07-07 2020-03-24 Plantex Ltd. Solid state forms of Palbociclib dimesylate
US11332467B2 (en) 2016-07-07 2022-05-17 Plantex Ltd. Solid state forms of palbociclib dimesylate
CN106220626A (en) * 2016-07-31 2016-12-14 合肥远志医药科技开发有限公司 A kind of polymorphic of Pabuk former times profit cloth and preparation method thereof
CN108929321A (en) * 2018-07-03 2018-12-04 威海贯标信息科技有限公司 A kind of Pa Boxini novel crystal forms
CN115667260A (en) * 2022-08-26 2023-01-31 成都苑东生物制药股份有限公司 Piperazine Bai Xili saccharine salt crystal form and preparation method thereof
WO2024040668A1 (en) * 2022-08-26 2024-02-29 成都苑东生物制药股份有限公司 Palbociclib saccharinate crystal form and preparation method therefor
CN115667260B (en) * 2022-08-26 2024-04-23 成都硕德药业有限公司 Saccharin salt crystal form of piperaquine Bai Xili and preparation method thereof

Similar Documents

Publication Publication Date Title
RU2497820C2 (en) Crystalline forms and two solvate forms of lactic acid salts 4-amino-5-fluorine-3-[5-(4-methylpiperazin-1-yl)-1-h-benzimidazol-2-yl]quinoline-2(1h)one
WO2016090257A1 (en) Salts and crystalline forms of 6-acetyl-8-cyclopentyl-5-methyl-2((5-(piperazin-1-yl)pyridin-2-yl)amino)pyrido[2,3-d] pyrimidin-7(8h)-one (palbociclib)
EP3248983B1 (en) Crystal form a of obeticholic acid and preparation method therefor
JP2018024708A (en) Salts of epidermal growth factor receptor kinase inhibitor
AU2018259089B2 (en) Polymorphs and solid forms of (s)-2-((2-((s)-4-(difluoromethyl)-2-oxooxazolidin-3-yl)-5,6-dihydrobenzo(ƒ)imidazo(1,2-d)(1,4)oxazepin-9-yl)amino)propanamide, and methods of production
JP2015017116A (en) Lactate salt of 4-[6-methoxy-7-(3-piperidin-1-yl-propoxy)quinazolin-4-yl]piperazine-1-carboxylic acid(4-isopropoxyphenyl)-amide and pharmaceutical compositions thereof for treatment of cancer and other diseases or disorders
TWI675839B (en) Form crystal of bisulfate of janus kinase (jak) inhibitor and preparation method thereof
JP5899214B2 (en) Solid form of 4-[-2-[[5-Methyl-1- (2-naphthalenyl) -1H-pyrazol-3-yl] oxy] ethyl] morpholine hydrochloride
JP2015180679A (en) Crystalline forms of sodium 4-{[9-chloro-7-(2-fluoro-6-methoxyphenyl)-5h-pyrimido[5,4-d][2]benzazepin-2-yl]amino}-2-methoxybenzoate
JP7235748B2 (en) Salt of 4-amino-N-(1-((3-chloro-2-fluorophenyl)amino)-6-methylisoquinolin-5-yl)thieno[3,2-D]pyrimidine-7-carboxamide and crystals thereof form
WO2013124385A2 (en) Solid forms of dabigatran etexilate mesylate and processes for their preparation
CN111278808B (en) Solid forms of 2- (5- (4- (2-morpholinoethoxy) phenyl) pyridin-2-yl) -N-benzyl acetamide
TW201617347A (en) Form crystal of bisulfate of janus kinase (JAK) inhibitor and preparation method thereof
JP7314319B2 (en) Crystal polymorph of CDK9 inhibitor, method for producing the same, and use thereof
JP2018537486A (en) Crystalline forms of quinolone analogs and their salts
JP4980431B2 (en) Maleic acid monosalt of antiviral agent and pharmaceutical composition containing the same
TW202404604A (en) New solid forms of [(1s)-1-[(2s,4r,5r)-5-(5-amino-2-oxo-thiazolo[4,5-d]pyrimidin-3-yl)-4-hydroxy-tetrahydrofuran-2-yl]propyl] acetate
US20220162185A1 (en) Crystalline and amorphous forms of n-(5-((4-ethylpiperazin-1-yl)methyl)pyridine-2-yl)-5-fluoro-4-(3-isopropyl-2-methyl-2h-indazol-5-yl)pyrimidin-2-amine and its salts, and preparation methods and therapeutic uses thereof
TW201811326A (en) Salt forms of 4-cyano-N-(2-(4,4-dimethylcyclohex-1-en-1-yl)-6-(2,2,6,6-tetramethyltetrahydro-2H-pyran-4-yl)pyridin-3-yl)-1H-imidazole-2-carboxamide
TW202227406A (en) Polymorphs of an ssao inhibitor
WO2016206633A1 (en) Phenyl amino pyrimidine compound or polymorph of salt thereof
US20220098173A1 (en) Crystalline form of tegavivint, method of preparation, and use thereof
WO2016101912A1 (en) Crystal form of salt of epidermal growth factor receptor kinase inhibitor and preparation method thereof
TW202327593A (en) Polymorph of kras inhibitor and preparation method for polymorph and use thereof
WO2016206634A1 (en) Morpholino phenyl amino pyrimidine compound or polymorph of salt thereof

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 15865815

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 15865815

Country of ref document: EP

Kind code of ref document: A1