US20080275101A1 - Solid Salt Forms Of A Pyrrole Substituted 2-Indolinone - Google Patents

Solid Salt Forms Of A Pyrrole Substituted 2-Indolinone Download PDF

Info

Publication number
US20080275101A1
US20080275101A1 US12/067,242 US6724206A US2008275101A1 US 20080275101 A1 US20080275101 A1 US 20080275101A1 US 6724206 A US6724206 A US 6724206A US 2008275101 A1 US2008275101 A1 US 2008275101A1
Authority
US
United States
Prior art keywords
salt
compound
phosphate
solvent
kit
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US12/067,242
Other languages
English (en)
Inventor
Michael Hawley
Changquan C. Sun
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Products Inc
Pfizer Inc
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 Pfizer Products Inc, Pfizer Inc filed Critical Pfizer Products Inc
Priority to US12/067,242 priority Critical patent/US20080275101A1/en
Publication of US20080275101A1 publication Critical patent/US20080275101A1/en
Assigned to PFIZER INC. reassignment PFIZER INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PFIZER PRODUCTS INC.
Assigned to PFIZER INC. reassignment PFIZER INC. CORRECTION BY DECLARATION OF INCORRECT NUMBERS RECORDED AT REEL 029041 FRAME 0099. Assignors: PFIZER INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Definitions

  • the present invention relates to solid salt forms of a 3-pyrrole substituted 2-indolinone compound, 5-[5-fluoro-2-oxo-1,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl-1H-pyrrole-3-carboxylic acid (2-pyrrolidin-1-yl-ethyl)-amide.
  • PKs protein kinases
  • the compounds of this invention are therefore useful in treating disorders related to abnormal PK activity.
  • Pharmaceutical compositions comprising salts of this compound and methods of preparing them are disclosed.
  • the present invention is also directed to polymorphs of the phosphate salt form of the amide.
  • Polymorphism occurs when a compound crystallizes in a multiplicity of solid phases that differ in crystal packing. Numerous examples are cited in the standard references of solid state properties of pharmaceuticals, Byrn, S. R., Solid-State Chemistry of Drugs, New Your, Academic Press (1982); Kuhnert-Brandstatter, M., Thermomiscroscopy In The Analysis of Pharmaceuticals, New York, Pergamon Press (1971) and Haleblian, J. K. and McCrone, W. Pharmaceutical applications of polymorphism. J. Pharm. Sci., 58, 911 (1969). Byrn states that, in general, polymorphs exhibit different physical characteristics including solubility and physical and chemical stability.
  • polymorphs may differ in ways that influence drug release, solid-state stability, and pharmaceutical manufacturing.
  • the relative stability and the interconversions of polymorphs are particularly important to the selection of a marketed drug.
  • a suitable polymorph may hinge upon the issue of physical stability.
  • the selection of a marketed drug may depend upon the availability and selection of a suitable polymorph having desirable characteristics, such as excellent physical stability or the ability to be manufactured in large scale.
  • the performance of the solid dosage form should not be limited by polymorphic transformations during the shelf life of the product. It is important to note that there is no reliable method to predict the observable crystal structures of a given drug or to predict the existence of polymorphs with desirable physical properties.
  • PKs are enzymes that catalyze the phosphorylation of hydroxy groups on tyrosine, serine, and threonine residues of proteins.
  • the consequences of this seemingly simple activity are staggering since virtually all aspects of cell life (e.g., cell growth, differentiation, and proliferation) in one way or another depend on PK activity.
  • abnormal PK activity has been related to a host of disorders, ranging from relatively non-life threatening diseases such as psoriasis to extremely virulent diseases such as glioblastoma (brain cancer).
  • Receptor tyrosine kinases are excellent candidates for molecular targeted therapy, because they play key roles in controlling cell proliferation and survival and are frequently dysregulated in a variety of malignancies.
  • the mechanisms of dysregulation include overexpression (Her2/neu in breast cancer, epidermal growth factor receptor in non-small cell lung cancer), activating mutations (KIT in gastrointestinal stromal tumors, fms-related tyrosine kinase 3/Flk2 (FLT3) in acute myelogenous leukemia), and autocrine loops of activation (vascular endothelial growth factor/VEGF receptor (VEGF/VEGFR) in melanoma, platelet-derived growth factor/PDGF receptor (PDGF/PDGFR) in sarcoma).
  • overexpression Her2/neu in breast cancer, epidermal growth factor receptor in non-small cell lung cancer
  • KIT gastrointestinal stromal tumors
  • FLT3 fms-related tyrosine kinase 3/Fl
  • the RTKs and their ligands, VEGF, PDGF, and FGF mediate neo-vascularization, known as angiogenesis, in solid tumors. Consequently, by inhibiting the RTKs, the growth of new blood vessels into tumors may be inhibited.
  • Antiangiogenesis agents a class of molecules that inhibits the growth of blood vessels into tumors, have much less toxicity to the body compared to conventional anti-cancer drugs.
  • U.S. Pat. No. 6,573,293, incorporated herein by reference discloses, among other compounds, 5-[5-fluoro-2-oxo-1,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl-1H-pyrrole-3-carboxylic acid (2-pyrrolidin-1-yl-ethyl)-amide (hereinafter “Compound I”). It has the following structure:
  • Compound I is a small molecule that exhibits PK modulating ability. The compound is therefore useful in treating disorders related to abnormal PK activity. It is an inhibitor of the RTKs, PDGFR, VEGFR, KIT, and FLT3. Compound I has been shown to inhibit KIT phosphorylation, arrest cell proliferation, and induce cell cycle arrest and apoptosis in malignant mast cell lines in vitro expressing various forms of mutant KIT. Compound I and related molecules are effective in preclinical models against tumor xenografts arising from cell lines of diverse human tumor origin.
  • Compound I is useful for treating cancers in companion animals, mainly dogs, and is also useful for the treatment of, inter alia, cancer in humans.
  • cancers include, but are not limited to, leukemia, brain cancer, non-small cell lung cancer, squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, lung cancer, bladder cancer, head and neck cancer, small-cell lung cancer, glioma, colorectal cancer, genitourinary cancer, and gastrointestinal stromal cancer.
  • Compound I is useful for the treatment of diseases related to overexpression of mast cells, including but not limited to, mastocytosis in humans and mast cell tumors in dogs.
  • Compound I readily crystallizes. Its solubility is about 10 ⁇ g/1 mL in pH 6 phosphate buffer at 25° C. When the compound was synthesized, very fine particles precipitated out of solution during the last step of synthesis. Subsequent isolation of these fine particles by filtration was slow, and a hard cake resulted after filtration. There is a need for a salt of Compound I which has physical stability and desirable physical properties.
  • This invention comprises salt forms of Compound I.
  • Five different salt forms of Compound I were synthesized and are described herein. (See Table 1) These include the hydrochloride, fumarate, citrate, phosphate, and ascorbate salts of Compound I. Based on characterization of these salts, the 1:1 phosphate salt, Compound I phosphate, was identified as a salt form with highly desirable characteristics. Polymorph screening revealed the existence of 10 polymorphs of Compound I phosphate, herein named Forms I through X.
  • this invention provides two salt forms of Compound I, wherein the salt form is selected from the citrate and phosphate salts, and solvates and polymorphs thereof.
  • the salt form is selected from the citrate and phosphate salts, and solvates and polymorphs thereof.
  • the phosphate salt form with a molecular formula of C 22 H 25 FN 4 O 2 .H 3 O 4 P is selected.
  • the phosphate salt form with a melting point from about 285 to about 290° C. is selected.
  • Compound I phosphate has a structure of
  • the citrate salt, Compound I citrate which has a molecular formula of C 22 H 25 FN 4 O 2 .C 6 H 8 O 7 is selected.
  • the citrate salt form with a melting point from about 178 to about 183° C. is selected.
  • Compound I citrate has a structure of
  • a second aspect of the invention is a pharmaceutical composition
  • a pharmaceutical composition comprising the phosphate salt or the citrate salt of Compound I, or solvates or polymorphs thereof, and a pharmaceutically acceptable carrier or excipient.
  • a third aspect of the invention is a method for the modulation of the catalytic activity of protein kinases comprising contacting said protein kinase with the phosphate or citrate salts of Compound I, or solvates or polymorphs thereof.
  • the protein kinase may be selected from the group consisting of receptor tyrosine kinases, non-receptor protein tyrosine kinases, and serine/threonine protein kinases.
  • a fourth aspect of the invention is a method of preventing or treating a protein kinase related disorder in an organism comprising administering to said organism a therapeutically effective amount of a pharmaceutical composition comprising the phosphate salt or the citrate salt of Compound I, or solvates or polymorphs thereof, and a pharmaceutically acceptable carrier or excipient.
  • the organism is a human.
  • the organism is a companion animal.
  • the companion animal is a cat or a dog.
  • the protein kinase related disorder may be selected from the group consisting of a receptor tyrosine kinase related disorder, a non-receptor protein tyrosine kinase related disorder, and a serine/threonine protein kinase related disorder.
  • the protein kinase related disorder may be selected from the group consisting of an EGFR related disorder, a PDGFR related disorder, an IGFR related disorder, a c-kit related disorder, and a FLK related disorder.
  • Such disorders include by way of example and not limitation, leukemia, brain cancer, non-small cell lung cancer, squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, lung cancer, bladder cancer, head cancer, neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, small cell lung cancer, glioma, mastocytosis, mast cell tumor, colorectal cancer, genitourinary cancer, gastrointestinal cancer, diabetes, an autoimmune disorder, a hyperproliferation disorder, restenosis, fibrosis, psoriasis, von Heppel-Lindau disease, osteoarthritis, rheumatoid arthritis, angiogenesis, an inflammatory disorder, an immunological disorder, and a cardiovascular disorder.
  • a fifth aspect of the invention is a method of preparing phosphate salt crystals of base 5-[5-fluoro-2-oxo-1,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl-1H-pyrrole-3-carboxylic acid (2-pyrrolidin-1-yl-ethyl)-amide which comprises introducing a stoichiometric amount of phosphoric acid to the base in a solution comprising a solvent or a mixture of solvents, forcing the phosphate salt in solution to crystallize, separating the phosphate salt crystals from the solvent solution, and drying the crystals.
  • the phosphoric acid may be introduced in an amount which is 40% molar excess to the base.
  • the solvent may comprise isopropanol.
  • the step of separating the crystals from the solvent solution may comprise adding acetonitrile to the solution and rotovapping the solution.
  • the step of separating the crystals from the solvent solution may also
  • a sixth aspect of the invention is a method of preparing citrate salt crystals of base 5-[5-fluoro-2-oxo-1,2-dihydro-indol-(3Z)-ylidenemethyl]-2,4-dimethyl-1H-pyrrole-3-carboxylic acid (2-pyrrolidin-1-yl-ethyl)-amide which comprises introducing a stoichiometric amount of citric acid to the base in a solution comprising a solvent or a mixture of solvents, forcing the citrate salt in solution to crystallize, separating the citrate salt crystals from the solvent solution, and drying the crystals.
  • the citric acid may also be introduced in an amount of about 40% molar excess to the base.
  • the solvent may comprise methanol.
  • the step of separating the crystals from the solvent solution may comprise adding acetonitrile to the solution and rotovapping the solution.
  • the step of separating the crystals from the solvent solution may comprise filtration.
  • the invention provides the polymorphs Forms I-X (as described herein) of the phosphate salt of Compound I.
  • Form I is provided.
  • An eighth aspect of the invention is a pharmaceutical composition comprising the Form I polymorph of Compound I phosphate and a pharmaceutically acceptable carrier or excipient.
  • a ninth aspect of the invention is a method for the modulation of the catalytic activity of protein kinases comprising contacting said protein kinase with the Form I polymorph of Compound I phosphate.
  • a tenth aspect of the invention is a method of preventing or treating a protein kinase related disorder in an organism comprising administering to said organism a therapeutically effective amount of the Form I polymorph of Compound I phosphate.
  • the organism is a human or companion animal.
  • the companion animal is a cat or a dog.
  • disorders include by way of example and not limitation, mast cell tumor and mastocytosis.
  • An eleventh aspect of the invention is a method of preparing polymorphs of Compound I phosphate, which comprises introducing the phosphate salt to a solution comprising a solvent or a mixture of solvents, optionally, adding a bridging solvent to the solution, and separating the polymorph crystals from the solvent solution.
  • the solution may comprise water plus acetonitrile.
  • the solution may comprise methanol.
  • the bridging solvent may be methanol.
  • a twelfth aspect of the invention is the use of the phosphate or citrate salts of Compound I or the Form I polymorph of the phosphate salt in the preparation of a medicament which is useful in the treatment of a disease mediated by abnormal PK activity.
  • FIG. 1 Moisture sorption data for salts of Compound I.
  • FIG. 2 Powder X-ray Diffraction patterns for Compound I citrate and Compound I phosphate.
  • FIG. 3 Powder X-ray Diffraction patterns of the ten unique solids obtained from the polymorph screening study (See Example 5). Form I through Form X as designated in Tables 5 and 6 are presented.
  • FIG. 4 TGA curves of solids from CH 2 Cl 2 (Form VI, immediately after precipitation), Hexane (Form VII, after standing overnight), and acetonitrile (Form VIII, after standing 3 days).
  • FIG. 5 Results of agarose gel electrophoresis of PCR products from MCTs evaluated in Example 7. Lanes 1-5 correspond to patients 1-5 in Table 8; Lanes 6-14 correspond to patients 6-14 in Table 8. Controls consisted of PCR products generated from the C2 canine mast cell line containing a 48-bp ITD (Lane 15) and from normal canine cerebellum (wild type; Lane 16).
  • FIG. 6 Reductions in MCT phosphorylated KIT and phosphorylated extracellular signal-regulated kinase (ERK)1/2 after a single dose of Compound I phosphate
  • C when used in reference to temperature means centigrade or Celsius.
  • catalytic activity refers to the rate of phosphorylation of tyrosine under the influence, direct or indirect, of RTKs and/or CTKs or the phosphorylation of serine and threonine under the influence, direct or indirect, of STKs.
  • Companion animal refers to domesticated animals offering companionship to humans, and includes, but is not limited to, cats and dogs.
  • contacting refers to bringing a compound of the present invention and a target PK together in such a manner that the compound can affect the catalytic activity of the PK, either directly, i.e., by interacting with the kinase itself, or indirectly, i.e., by interacting with another molecule on which the catalytic activity of the kinase is dependent.
  • IC 50 means the concentration of a test compound which achieves a half-maximal inhibition of the PK activity.
  • modulation refers to the alteration of the catalytic activity of RTKs, CTKs, and STKs.
  • modulating refers to the activation or inhibition of the catalytic activity of RTKs, CTKs, and STKs, preferably the activation of the catalytic activity of RTKs, CTKs, and STKs, depending on the concentration of the compound or salt to which the RTK, CTK, or STK is exposed or, more preferably, the inhibition of the catalytic activity of RTKs, CTKs, and STKs.
  • PK refers to receptor protein tyrosine kinase (RTKs), non-receptor or “cellular” tyrosine kinase (CTKs) and serine-threonine kinases (STKs).
  • RTKs receptor protein tyrosine kinase
  • CTKs non-receptor or “cellular” tyrosine kinase
  • STKs serine-threonine kinases
  • polymorph refers to a solid phase of a substance, which occurs in several distinct forms due to different arrangements and/or confirmations of the molecules in crystal lattice. Polymorphs typically have different chemical and physical properties.
  • pharmaceutically acceptable excipient refers to any substance other than a compound of the invention, added to a pharmaceutical composition.
  • composition refers to a mixture of one or more of the salts of the present invention or the polymorphs of such salts, as described herein, with other chemical components, such as physiologically/pharmaceutically acceptable carriers and excipients.
  • the purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.
  • physiologically/pharmaceutically acceptable carrier refers to a carrier or diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • polymorph may also be defined as different unsolvated crystal forms of a compound.
  • the term also includes solvates (i.e., forms containing solvent, or water), amorphous forms (i.e., noncrystalline forms), and desolvated solvates (i.e., forms which can only be made by removing the solvent from a solvate).
  • solvate is used to describe a molecular complex comprising a compound of the invention and one or more pharmaceutically acceptable solvent molecules, for example, ethanol.
  • solvent molecules for example, ethanol.
  • hydrate is employed when said solvent is water.
  • substantially free in relation to the amount of a certain polymorph in a sample means that other polymorphs are present in an amount less than about 15 weight percent. In another embodiment, “substantially free” means less than about 10 weight percent. In another embodiment, “substantially free” means less than about 5 weight percent. In still another embodiment, “substantially free” means less than about 1 weight percent.
  • the phrase “in an amount less than about 15 weight percent” means that the polymorph of interest is present in an amount of more than about 85 weight percent.
  • the phrase “less than about 10 weight percent” means that the polymorph of interest is present in an amount of more than about 90 weight percent, and so on.
  • terapéuticaally effective amount refers to that amount of the compound being administered which will prevent, alleviate, or ameliorate one or more of the symptoms of the disorder being treated, or prolong the survival of the subject being treated.
  • a therapeutically effective amount refers to that amount which has the effect of:
  • the base compound may be in solution.
  • the solution is generally a solvent.
  • the solution is an alcohol.
  • the solvent may be isopropanol, methanol, acetonitrile, or water plus acetonitrile.
  • the solution may also comprise a mixture of solvents.
  • the salts may be crystallized using a stoichiometric addition/crystallization technique.
  • a stoichiometric amount of the counterion is introduced to the base in solution.
  • the amount of counterion is in a 1:1 ratio to the base.
  • the amount of counterion is from 0% to about 60% molar excess to the base.
  • the amount of counterion is from about 10% to about 50% molar excess to the base.
  • the amount of counterion is about 40% molar excess to the base.
  • the counterions may include hydrochloride, fumarate, citrate, phosphate, and ascorbate ions.
  • the counterion is the phosphate ion.
  • the counterion is the citrate ion.
  • the salt in solution is then forced to crystallize by a variety of common techniques including cooling, evaporation, drowning, etc., known to one skilled in the art.
  • Excess solvents may be removed from the samples by methods known to one skilled in the art.
  • the solvents are removed from the solution by adding acetonitrile (ACN) and rotovapping the solution.
  • ACN acetonitrile
  • the solution may be rotovapped from about 40° C. to about 60° C.
  • additional solvents may be added to the solution (eg, isopropanol and methyl ethyl ketone) prior to rotovapping.
  • the crystallizations may be conducted in the dark to prevent light-induced isomerization.
  • the crystals are removed by filtration.
  • filtration may be performed at ambient laboratory atmosphere.
  • the ascorbate, citrate, fumarate, hydrochloride, and phosphate salts of Compound I were crystallized. Specific examples of crystallization methods are provided below. HPLC analysis may be used to determine purity of the resultant sample. The physical properties of the compounds may be determined by tests known to one skilled in the art, including melting point determination, powder X-ray diffraction, and dynamic moisture sorption gravimetry. Parameters for these tests are described below.
  • the 1:1 phosphate salt, Compound I phosphate was identified as a salt form with highly desirable characteristics, including good crystallinity, low moisture uptake, ease of crystallization, good purity, and lack of hydrate.
  • the citrate salt also demonstrated desirable characteristics, such as low moisture uptake and good crystallinity.
  • Polymorphs of the compounds of the present invention are desirable because a particular polymorph of a compound may have better physical and chemical properties than other polymorphic forms of the same compound.
  • one polymorph may have increased solubility in certain solvents. Such added solubility may facilitate formulation or administration of the compounds of the present invention.
  • Different polymorphs may also have different mechanical properties (e.g., different compressibility, compactibility, tabletability), which may influence tableting performance of the drug, and thus influence formulation of the drug.
  • a particular polymorph may also exhibit different dissolution rate in the same solvent, relative to another polymorph.
  • Different polymorphs may also have different physical (solid-state conversion from metastable polymorph to a more stable polymorph) and chemical (reactivity) stability.
  • An embodiment of the present invention contemplates the Form I polymorph of Compound I phosphate, as described herein.
  • pure, single polymorphs as well as mixtures comprising two or more different polymorphs are contemplated.
  • a pure, single polymorph may be substantially free from other polymorphs.
  • compositions comprising one or more of the salts of Compound I or the polymorphs of such salts, as described herein, and a pharmaceutically acceptable carrier or excipient.
  • Polymorphs were generated from concentrated solutions of Compound I phosphate.
  • the concentrated solutions may be in a range of 60 to 100 mg of Compound I phosphate per mL of solution. In one embodiment, about 70 mg of Compound I may be dissolved in 1 mL of phosphoric acid.
  • the polymorph crystals may be precipitated from a solvent by various methods including, for example, slow evaporation, cooling a supersaturated solution, precipitation from anti-solvents, etc., which are known to one skilled in the art.
  • the polymorph crystals are generated by adding the solution to an anti-solvent.
  • the anti-solvent may be water plus acetonitrile (ANC), ethanol, methanol, acetone, acetonitrile, THF, ethyl acetate, hexane, methylene chloride (CH 2 Cl 2 ), isopropyl alcohol (IPA), methyl ethyl ketone (MEK), and dioxane.
  • an additional solvent eg, methanol
  • the samples are allowed to stand overnight prior to removing the crystals.
  • the samples are allowed to stand for three days prior to removing the crystals.
  • the crystals may be characterized using standard methods known to one skilled in the art, including PXRD dynamic moisture sorption gravimetry, differential scanning calorimetry, thermal gravimetric analysis, and optical microscopy. These techniques are described below.
  • compositions suitable for the delivery of compounds of the present invention and methods for their preparation will be readily apparent to those skilled in the art. Such compositions and methods for their preparation may be found, for example, in Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company, 1995).
  • excipient The choice of a pharmaceutically acceptable excipient will to a large extent depend on factors such as the particular mode of administration, the effect of the excipient on solubility and stability, and the nature of the dosage form.
  • excipients include, without limitation, calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.
  • Carriers and excipients for formulation of pharmaceutically acceptable compositions comprising Compound I are well known in the art and are disclosed, for example, in U.S. Pat. No. 6,573,293, which is incorporated herein in its entirety. Methods of administration for such are also known in the art and also described, for example, in U.S. Pat. No. 6,573,293. Similar methods could also be used to formulate and administer pharmaceutically acceptable compositions of the salts of Compound I, or the polymorphs of such salts, of this invention.
  • the compounds of the present invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks' solution, Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • parenteral administration e.g., by bolus injection or continuous infusion
  • formulations may be presented in unit dosage forms, such as in ampoules or in multi-dose containers.
  • the compositions may take such forms as suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulating materials such as suspending, stabilizing, or dispersing agents.
  • the compounds of the invention may be administered directly into the blood stream, into muscle, or into an internal organ.
  • Suitable means for parenteral administration include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial and subcutaneous.
  • Suitable devices for parenteral administration include needle (including microneedle) injectors, needle-free injectors and infusion techniques.
  • Parenteral formulations are typically aqueous solutions which may contain excipients such as salts, carbohydrates and buffering agents (preferably adjusted to a pH of from 3 to 9), but, for some applications, they may be more suitably formulated as a sterile non-aqueous solution or as a dried form to be used in conjunction with a suitable vehicle such as sterile, pyrogen-free water. Additionally, suspensions of the compounds of the present invention may be prepared in a lipophilic vehicle. Suitable lipophilic vehicles include fatty oils such as sesame oil, synthetic fatty acid esters, such as ethyl oleate and triglycerides, or materials such as liposomes.
  • the compounds of the invention may be administered orally.
  • Oral administration may involve swallowing, so that the compound enters the gastrointestinal tract, and/or buccal, lingual, or sublingual administration by which the compound enters the blood stream directly from the mouth.
  • the compounds can be formulated by combining the compounds of the present invention with pharmaceutically acceptable carriers well known in the art.
  • Formulations suitable for oral administration include solid, semi-solid and liquid systems such as tablets; soft or hard capsules containing multi- or nano-particulates, liquids, or powders; lozenges (including liquid-filled); chews; gels; fast dispersing dosage forms; films; ovules; sprays; and buccal/mucoadhesive patches.
  • the compounds of the invention may also be administered topically, (intra)dermally, or transdermally to the skin or mucosa.
  • Typical formulations for this purpose include gels, hydrogels, lotions, solutions, creams, ointments, dusting powders, dressings, foams, films, skin patches, wafers, implants, sponges, fibres, bandages and microemulsions. Liposomes may also be used.
  • Typical carriers include alcohol, water, mineral oil, liquid petrolatum, white petrolatum, glycerin, polyethylene glycol and propylene glycol.
  • Penetration enhancers may be incorporated—see, for example, J Pharm Sci, 88 (10), 955-958, by Finnin and Morgan (October 1999).
  • Other means of topical administration include delivery by electroporation, iontophoresis, phonophoresis, sonophoresis and microneedle or needle-free (e.g. PowderjectTM, BiojectTM, etc.) injection
  • the compounds of the present invention may be formulated for rectal administration, such as suppositories or retention enemas using, for example, conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds of the present invention may also exist in unsolvated and solvated forms.
  • the embodiments of the present invention also contemplate a method for the modulation of the catalytic activity of a PK comprising contacting said PK with a one or more of the salts of Compound I or the polymorphs of such salts of the present invention.
  • contacting can be accomplished “in vitro,” i.e., in a test tube, a petri dish, or the like.
  • contacting may involve only a compound and a PK of interest or it may involve whole cells. Cells may also be maintained or grown in cell culture dishes and contacted with a compound in that environment.
  • the ability of a particular compound to affect a PK-related disorder i.e., the IC 50 of the compound, defined below, can be determined before use of the compounds is attempted in vivo with more complex living organisms.
  • a PK-related disorder i.e., the IC 50 of the compound, defined below.
  • Embodiments of the present invention contemplate a method for treating or preventing a protein kinase related disorder in an organism (e.g., a companion animal or a human) comprising administering a therapeutically effective amount of a pharmaceutical composition comprising one or more of the salts of Compound I or the polymorphs of such salts of the present invention and a pharmaceutically acceptable carrier or excipient to the organism.
  • an organism e.g., a companion animal or a human
  • a pharmaceutical composition comprising one or more of the salts of Compound I or the polymorphs of such salts of the present invention and a pharmaceutically acceptable carrier or excipient to the organism.
  • the protein kinase related disorder is selected from the group consisting of a receptor tyrosine kinase related disorder, a non-receptor tyrosine kinase related disorder, and a serine-threonine kinase related disorder.
  • the protein kinase related disorder is selected from the group consisting of an EGFR related disorder, a PDGFR related disorder, an IGFR related disorder, and a FLK related disorder.
  • the receptor protein kinase whose catalytic activity is modulated by a compound of this invention is selected from the group consisting of EGF, HER2, HER3, HER4, IR, IGF-1R, IRR, PDGFR ⁇ , PDGFR ⁇ , CSFIR, C-Kit, C-fms, Flk-1R, Flk4, KDR/Flk-1, Flt-1, FGFR-1R, FGFR-2R, FGFR-3R and FGFR-4R.
  • the cellular tyrosine kinase whose catalytic activity is modulated by a compound of this invention is selected from the group consisting of Src, Frk, Btk, Csk, Abl, ZAP70, Fes/Fps, Fak, Jak, Ack, Yes, Fyn, Lyn, Lck, Blk, Hck, Fgr and Yrk.
  • the serine-threonine protein kinase whose catalytic activity is modulated by a compound of this invention is selected from the group consisting of CDK2 and Raf.
  • the protein kinase related disorder is selected from the group consisting of squamous cell carcinoma, astrocytoma, Kaposi's sarcoma, glioblastoma, lung cancer, bladder cancer, head and neck cancer, melanoma, ovarian cancer, prostate cancer, breast cancer, small cell lung cancer, glioma, colorectal cancer, genitourinary cancer, gastrointestinal cancer, mastocytosis, and mast cell tumors.
  • the protein kinase related disorder is selected from the group consisting of diabetes, an autoimmune disorder, a hyperproliferation disorder, restenosis, fibrosis, psoriasis, von Heppel-Lindau disease, osteoarthritis, rheumatoid arthritis, angiogenesis, an inflammatory disorder, an immunological disorder, and a cardiovascular disorder.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an amount sufficient to achieve the intended purpose, e.g., the modulation of PK activity or the treatment or prevention of a PK-related disorder.
  • the therapeutically effective amount or dose can be estimated initially from cell culture assays. Then, the dosage can be formulated for use in animal models so as to achieve a circulating concentration range that includes the IC 50 as determined in cell culture. Such information can then be used to more accurately determine useful doses in humans or companion animals.
  • the amount of the compound to be administered ranges from about 0.001 to about 100 mg per kg of body weight, such total dose being given at one time or in divided doses.
  • the amount of a composition administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician or veterinarian, etc.
  • the effective local concentration of the drug may not be related to plasma concentration and other procedures known in the art may be employed to determine the correct dosage amount and interval.
  • Embodiments of the present invention also contemplate a method of treating cancer in companion animals comprising administering a pharmaceutical composition comprising one or more of the salts of Compound I or the polymorphs of such salts of the present invention and a pharmaceutically acceptable carrier or excipient.
  • the salts of Compound I or the polymorphs of such salts, as described herein would be metabolized by enzymes in the body of an organism such as a companion animal or a human being to generate a metabolite that can modulate the activity of the protein kinases. Such metabolites are within the scope of the present invention.
  • Compounds of the invention may be administered alone or in combination with one or more other compounds of the invention or in combination with one or more other drugs (or as any combination thereof). It is also contemplated that the salts of Compound I or the polymorphs of such salts, as described herein, might be combined with other chemotherapeutic agents for treatment of the diseases and disorders discussed above. For example, a compound of the present invention may be combined with fluorouracil alone or in further combination with leukovorin or other alkylating agents. A compound of the present invention may be used in combination with other antimetabolite chemotherapeutic agents such as, without limitation, folic acid analogs or the purine analogs.
  • a compound may also be used in combination with natural product based chemotherapeutic agents, antibiotic chemotherapeutic agents, enzymatic chemotherapeutic agents, platinum coordination complexes, and hormone and hormone antagonist. It is also contemplated that a compound of the present invention could be used in combination with mitoxantrone or paclitaxel for the treatment of solid tumor cancers or leukemias.
  • Tests to determine the physical properties of the salts of Compound I included melting point determination, HPLC purity, powder X-ray diffraction, and dynamic moisture sorption gravimetry.
  • Powder X-ray Diffraction was performed using a Scintag X2 Advanced Diffraction System (lab 259-1088, controlled by Scintag DMS/NT 1.30a and Microsoft Windows NT 4.0 software.
  • the system uses a Copper X-ray source (45 kV and 40 mA) to provide CuK ⁇ 1 emission of 1.5406 ⁇ and a solid-state Peltier cooled detector.
  • the beam aperture was controlled using tube divergence and anti-scatter slits of 2 and 4 mm and detector anti-scatter and receiving slits of 0.5 and 0.2 mm width. Data were collected from 2 to 35° two-theta using a step scan of 0.03°/step with a counting time of one second per step. Scintag round, top loading stainless steel sample holders with 9 mm diameter inserts were utilized for the experiments. Powders were packed into the holder and were gently pressed by a glass slide to ensure coplanarity between the sample surface and the surface of holder.
  • DMSG Dynamic Moisture Sorption Gravimetry
  • Table 1 shows a summary of data for the ascorbate, citrate, fumarate, hydrochloride, and phosphate salts of Compound I. HPLC analysis suggested that the salts were of relatively high purity, and no significant change in the purity was induced through the salt formation process.
  • hydrochloride, fumarate, and ascorbate salts were very hygroscopic (see FIG. 1 ).
  • the other two salts (citrate and phosphate) had lower moisture sorption profiles, absorbing less than 3% water at 70% relative humidity.
  • Compound I free base was used to prepare the phosphate salt.
  • a sample (lot number 35282-CS-51) of Compound I phosphate was prepared as described above. 4 mL of 0.977 M phosphoric acid was added to 1.095 g of free base in a flask immediately followed by adding 4 mL of acetonitrile. A suspension was obtained. The suspension was heated slightly on a hot-plate. Adding 40 mL of water and heating while stirring for about one hour did not completely dissolve the solid. The solid was filtered and washed with 10 mL of acetonitrile. PXRD showed it was the phosphate salt of Compound I.)
  • Lot 35282-CS-51 was named polymorph Form I of Compound I Phosphate. It has high crystallinity, good flowability, and large crystal size. Both the absence of the melting event at the melting temperature of Compound I free base (free base polymorph Form A, 256° C.; free base polymorph Form B, 259° C.) and the presence of high melting points (281-297° C.) of the solids suggested that the crystals of Lot 35282-CS-51 are a different salt form and not Compound I free base. The purity of the lot was 99.6% by HPLC.
  • the vial was left on the bench for at least 30 minutes before the next addition of solvent. This step was repeated until no crystals were visible against a black and a white background.
  • the solubility was then bracketed by dividing the weight of the compound by the final volume and the volume before the last addition. If a solid remained after the addition of 10 mL of solvent, the solubility was expressed as less than the weight divided by the final volume. If the solid was completely dissolved after the first addition of solvent, the solubility was expressed as greater than the weight divided by the solvent volume. All experiments were conducted at room temperature.
  • the estimated solubilities of Compound I phosphate in various solvents are presented in Table 4 along with solubilities of the free base, expressed as mg/mL.
  • the solubility of Compound I phosphate is lower than that of Compound I free base in the same solvent, except in water (at various pH levels).
  • the solubility of Compound I phosphate depends on the pH value of a solution, and becomes considerably higher (>3 mg/mL) at pH 2 or lower.
  • the melting point of Compound I phosphate (lot 35282-CS-51) is about 281-297° C., which is substantially higher than the melting point of Compound I free base (free base polymorph Form A, 256° C.; free base polymorph Form B, 260° C.).
  • One important result is that the wettability of Compound I phosphate with water is much better than that of Compound I free base.
  • Example 4C The low solubilities of Compound I phosphate seen in Example 4C indicated that solutions of highly concentrated (60-100 mg/mL, dark orange-red) Compound I phosphate would be beneficial to precipitate polymorphs of Compound I phosphate from various solvents.
  • concentrated solutions were prepared by dissolving Compound I free base in about 1 M phosphoric acid.
  • Compound I free base could be dissolved in 1 mL of 1M phosphoric acid.
  • the amount of Compound I free base and phosphoric acid used depended on the desired concentration and batch size of the solution.
  • organic solvents e.g., ethyl acetate, hexane, CH 2 Cl 2
  • ethyl acetate, hexane, CH 2 Cl 2 Some organic solvents, e.g., ethyl acetate, hexane, CH 2 Cl 2 , are not miscible with water and two layers of solvents were observed. Only a little precipitation was seen at the interface even minutes after addition. In those cases, about 1 mL of methanol was added as a bridging solvent, to increase the miscibility between the two layers. Methanol appeared to work well to increase the miscibility because colorless organic layer became yellow as soon as methanol was added. The vial was then shaken vigorously by hand for about one minute. The solids precipitated from organic solvents were vacuum filtered both immediately after precipitation (within 20 min) and after standing overnight or for three days in order to isolate both metastable and stable polymorphs. The powder was then analyzed. The different solids were numbered in the order
  • DSC Differential Scanning Calorimetry
  • TGA Thermogravimetry
  • TGA experiments were performed using a high resolution analyzer (TA Instruments model 2950).
  • the TA Instruments Thermal SolutionsTM for NT (version 1.3L) was used for data collection, and the Universal AnalysisTM for NT (version 2.4F) was used for data analysis.
  • Samples (5-10 mg) were placed onto a aluminum pan which was further placed on a platinum weighing pan before being heated. The weights of the aluminum and platinum pans were tared prior to loading the samples. The temperature was increased from 30° C. to 300° C. linearly at a rate of 10° C./min. Dry nitrogen purge was used.
  • Microscopy was conducted on an Olympus BHSP polarized light microscope. Powder was suspended in silicone oil and dispersed between a microscopy slide and a cover slip. Prior to observation, the cover slip was gently rubbed against the slide to render good dispersion of the particles.
  • the temperatures listed in the table are peak temperatures only.
  • c Peak temperatures of a broad endothermal event from the starting temperature of a DSC run. These events are also reflected by gradual weight loss on TGA. This type of heat event is typical of loss of surface adsorbed solvent.
  • d Peak temperatures of a relatively sharp endothermal event on DSC. This event is also recorded by TGA as a sudden loss of weight at the corresponding peak temperature. This type of heat event is typical of desolvation of a solvate.
  • the aggregates are constituted of very fine particles.
  • f ND means that no definite form can be assigned because of the low signal of the diffraction peaks of corresponding PXRD patterns obscured a detailed comparison with PXRD patterns of other forms.
  • g Form VI is a solvent containing solid.
  • the temperatures listed in the table are peak temperatures only.
  • c Peak temperatures of a relatively sharp endothermal event on DSC. This event is also recorded by TGA as a sudden loss of weight at the corresponding peak temperature.
  • d Conversion was complete in 5 hrs (from yellow to orange-red).
  • e Conversion was not obvious in 5 hrs but was mostly completed in three days (yellow loose precipitate to orange-red well-formed needle-shaped crystals).
  • f Conversion was complete in 5 hrs. The color change was not obvious (from yellow to light orange).
  • g Form VII and VIII are solvent containing solids.
  • ERK1/2 mitogenactivated protein kinase downstream of KIT signaling
  • MAPK mitogenactivated protein kinase
  • Study Design This study was a proof of target modulation study in dogs with recurrent or metastatic grade II/III MCTs. Patients received a single oral dose of Compound I phosphate at 3.25 mg/kg. Using a 6-mm punch biopsy instrument, samples were obtained from the tumor before Compound I phosphate administration and 8 hours (h) after treatment. When possible, multiple biopsies were taken. Each sample was flash frozen in liquid nitrogen and stored at ⁇ 70° centigrade (C) before analysis. Blood samples for analysis of plasma levels of Compound I phosphate were obtained at the same time as tumor biopsies (see below).
  • Plasma samples were drawn from the jugular vein and placed into a red-top serum collection vacuum glass tube. Specimens were kept at room temperature, allowed to clot, centrifuged at 1500 rpm at 4° C. for 10 minutes, transferred to cryovials, and plasma frozen at ⁇ 70° C. pending analysis. Briefly, plasma samples (20 ⁇ l) or Compound I phosphate standards in canine plasma were mixed with methanol (200 ⁇ l) containing DL-propranolol hydrochloride (internal standard) in a 96-well polypropylene plate (Orochem Technology, Westmont, Ill.).
  • the plate was mixed by vortex for 1 min, and the samples were centrifuged for 10 min at 4000 rpm. Ten microliters of the supernatant were injected onto the LC/MS/MS system, in which separation occurred on a BataBasic C-18 (5 ⁇ m, 100 ⁇ 4.6 mm) reverse-phase high-performance liquid chromatography column (Keystone Scientific, Foster City, Calif.). The amount of Compound I phosphate and the internal standard in each canine plasma sample were quantified based on standard curves generated using known amounts of compound ranging from 0.2 to 500 ng/ml.
  • the PCR products were separated by electrophoresis on a 4% agarose gel; the expected wild-type c-kit PCR product is 196 bp in size for PCR from cDNA and 190 bp in size for genomic DNA PCR.
  • the PCR products were gel purified using the Promega PCR Wizard Clean-Up kit (Promega) and sequenced using both P1 (forward) and P5 or P2 (reverse) primers at the core sequencing facility at the University of California—Davis, to rule out the presence of very small ITDs, deletions, or point mutations. Sequence alignment and comparison were performed using the DNASIS sequence analysis program.
  • Tumor biopsies were frozen in liquid nitrogen and later pulverized using a liquid nitrogen-cooled cryomortar and pestle, then stored at ⁇ 70° C. until used.
  • pulverized tumors were homogenized, lysed, and immunoprecipitated from 1 mg of starting tumor lysate, as described previously (Abrams, T. J., Lee, L. B., Murray, L. J., Pryer, N. K., Cherrington, J. M. SU11248 inhibits KIT and platelet-derived growth factor receptor beta in preclinical models of human small cell lung cancer. Mol. Cancer Ther.
  • the blots were stripped, reblocked, and reprobed with an antibody to KIT (A-4542; DAKO Corp., Carpinteria, Calif.).
  • KIT KIT
  • p42/44 ERK the same tumor lysates used for KIT analysis were probed by Western blot with an antibody to phospho-Thr 202/Tyr 204 ERK1/2 (9101B; Cell Signaling Technology) and then stripped and reprobed with an antibody to total ERK (9102; Cell Signaling Technology).
  • Evaluable tumor biopsy pairs for both KIT and ERK1/2 were considered those for which detectable total protein was present in both biopsies of the pair.
  • Target modulation was scored by eye by three observers blinded to the JM status and plasma concentration. Reduction of ⁇ 50% in phospho-protein signal relative to total protein signal in the biopsy sample taken post-treatment compared with the pretreatment biopsy was scored as positive for target modulation, whereas a reduction of ⁇ 50% was scored negative.
  • the therapeutic range of Compound I for target inhibition was considered to be 50-100 ng/ml for 12 h of a 24-h dosing period.
  • the plasma concentration of Compound I phosphate at 8 h (approximately Cmax) after a single dose at 3.25 mg/kg ranged from 33.2 to 186 ng/ml, with an average of 105 ⁇ 9 ng/mL (Table 8).
  • the plasma concentration of Compound I phosphate was outside the range of the other samples (0.3 ng/ml). Twelve of 14 dogs had plasma levels considered to be in the therapeutic range established in a Phase I clinical study of Compound I. (London, C. A., Hannah, A.
  • Compound I phosphate also affected a signaling pathway downstream of KIT. Mutations in c-kit in GIST and hematopoietic malignancies have been reported to activate different signaling pathways from each other and from wild-type KIT. In canine MCTs, all but one tumor sample had detectable phosphorylated ERK1/2 at baseline. In 7 of 11 evaluable tumor biopsy pairs, ERK1/2 was inhibited, as measured by a reduction in phosphorylated ERK1/2 after treatment. Not all of the tumors scoring positive for ERK1/2 inhibition were also positive for inhibition of KIT phosphorylation. ERK1/2 target modulation did not correlate with tumor grade or the presence or absence of c-kit ITD mutation. As for KIT target modulation, ERK1/2 target modulation was detected more frequently in tumors that expressed high levels of ERK1/2 and phosphorylated ERK1/2 at baseline.
  • KIT inhibition at this plasma concentration may be reasonably extrapolated to successful inhibition of the other closely related receptor tyrosine kinase targets of Compound I expressed by these tumors, based on in vitro and in vivo potency of Compound I, providing a molecular rationale for objective responses in these tumors.
  • canine mammary tumors express VEGFR, which is inhibited by indolinone tyrosine kinase inhibitors at comparable concentrations to KIT in cellular in vitro assays.
  • Efficacy was based on the objective response (complete response or partial response) at the week 6 visit where the mean of the two evaluators sum of the longest diameter of target lesions (Mean Sum LD) was compared to the Baseline Mean Sum LD for calculation of percent reduction or increase.
  • Assessment of non-target lesions was subjective.
  • a complete response (CR) was defined as the disappearance of all target and non-target lesions and the appearance of no new lesions;
  • a partial response (PR) was defined as at least a 30% decrease in the Mean Sum LD of target lesions compared to the Baseline Mean Sum LD and non-progression of non-target lesions and appearance of no new lesions.
  • Tissue samples from the tumor and distant normal skin were collected prior to randomization and submitted for the assessment of c-kit mutation status.
  • T02 and 65 T01 animals were included in the efficacy analysis.
  • the data analysis indicated a statistically significant improvement in the primary endpoint (objective response) for Compound I phosphate (T02) compared to placebo (T01).
  • the T02 animals had a significantly greater objective response rate (38.3%; 33/86) compared to T01 animals (7.9%; 5/63) (p ⁇ 0.001).
  • Nearly twice as many T01 animals (66.7%; 42/63) experienced progressive disease compared to T02 animals (33.7%; 29/86).
  • Dogs in the T02 group that were positive for the c-kit mutation were almost twice as likely to have an objective response compared to those that were negative for the c-kit mutation (60%, 12/20 vs. 32.8%, 21/64, respectively).
US12/067,242 2005-09-19 2006-09-08 Solid Salt Forms Of A Pyrrole Substituted 2-Indolinone Abandoned US20080275101A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/067,242 US20080275101A1 (en) 2005-09-19 2006-09-08 Solid Salt Forms Of A Pyrrole Substituted 2-Indolinone

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US71858605P 2005-09-19 2005-09-19
US12/067,242 US20080275101A1 (en) 2005-09-19 2006-09-08 Solid Salt Forms Of A Pyrrole Substituted 2-Indolinone
PCT/IB2006/002506 WO2007034272A1 (en) 2005-09-19 2006-09-08 Solid salt forms of a pyrrole substituted 2-indolinone

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2006/002506 A-371-Of-International WO2007034272A1 (en) 2005-09-19 2006-09-08 Solid salt forms of a pyrrole substituted 2-indolinone

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/367,009 Continuation US8987320B2 (en) 2005-09-19 2012-02-06 Solid salt forms of a pyrrole substituted 2-indolinone

Publications (1)

Publication Number Publication Date
US20080275101A1 true US20080275101A1 (en) 2008-11-06

Family

ID=37626412

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/067,242 Abandoned US20080275101A1 (en) 2005-09-19 2006-09-08 Solid Salt Forms Of A Pyrrole Substituted 2-Indolinone
US13/367,009 Active 2027-07-07 US8987320B2 (en) 2005-09-19 2012-02-06 Solid salt forms of a pyrrole substituted 2-indolinone
US14/620,835 Active US9290479B2 (en) 2005-09-19 2015-02-12 Solid salt forms of a pyrrole substituted 2-indolinone

Family Applications After (2)

Application Number Title Priority Date Filing Date
US13/367,009 Active 2027-07-07 US8987320B2 (en) 2005-09-19 2012-02-06 Solid salt forms of a pyrrole substituted 2-indolinone
US14/620,835 Active US9290479B2 (en) 2005-09-19 2015-02-12 Solid salt forms of a pyrrole substituted 2-indolinone

Country Status (25)

Country Link
US (3) US20080275101A1 (pt)
EP (2) EP1928858B1 (pt)
JP (2) JP4519114B2 (pt)
KR (1) KR101050906B1 (pt)
CN (2) CN101287724B (pt)
AR (1) AR056521A1 (pt)
AU (1) AU2006293644B2 (pt)
BR (1) BRPI0616374B8 (pt)
CA (1) CA2621569C (pt)
CY (1) CY1109326T1 (pt)
DE (1) DE602006008015D1 (pt)
DK (1) DK1928858T3 (pt)
ES (1) ES2328407T3 (pt)
HK (1) HK1125101A1 (pt)
IL (1) IL189590A0 (pt)
MX (1) MX2008002415A (pt)
NO (1) NO20080866L (pt)
NZ (1) NZ566033A (pt)
PL (1) PL1928858T3 (pt)
PT (1) PT1928858E (pt)
RU (1) RU2399619C2 (pt)
SI (1) SI1928858T1 (pt)
TW (1) TWI310766B (pt)
WO (1) WO2007034272A1 (pt)
ZA (1) ZA200801431B (pt)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE202010017129U1 (de) 2009-01-23 2011-09-14 Teva Pharmaceutical Industries Ltd. Rasagilinformulierung mit verzögerter Freisetzung

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SI1928858T1 (sl) 2005-09-19 2009-10-31 Pfizer Prod Inc Trde solne oblike s pirolom substituiranega 2-indolinona
EP2545034A1 (en) 2010-03-10 2013-01-16 Synthon B.V. A process for amidation of pyrrole carboxylate compounds

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6573293B2 (en) * 2000-02-15 2003-06-03 Sugen, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
US20060009510A1 (en) * 2004-07-09 2006-01-12 Pharmacia & Upjohn Company Llc Method of synthesizing indolinone compounds
US7119209B2 (en) * 2002-02-15 2006-10-10 Pharmacia & Upjohn Company Process for preparing indolinone derivatives
US7211600B2 (en) * 1999-12-22 2007-05-01 Sugen Inc. Methods of modulating c-kit tyrosine protein kinase function with indolinone compounds
US7452913B2 (en) * 2003-02-24 2008-11-18 Pharmacia & Upjohn Company Polymorphs of pyrrole substituted 2-indolinone protein kinase inhibitors

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6574293B1 (en) 1998-10-28 2003-06-03 Ericsson Inc. Receivers and methods for reducing interference in radio communications
US6797725B2 (en) * 2001-04-09 2004-09-28 Sugen, Inc. Prodrugs of a 3-(pyrrol-2-ylmethylidene)-2-indolinone derivatives
PT3168218T (pt) 2001-08-15 2019-01-11 Pharmacia & Upjohn Co Llc Um cristal compreendendo um sal de ácido l-málico de n-[2- (dietilamino)etil]-5-[(5-fluoro-1,2-dihidro-2-oxo-3h-indol- 3-ilideno)metil]-2,4-dimetil-1h-pirrol-3-carboxamida para utilização como um medicamento
US20040209937A1 (en) * 2003-02-24 2004-10-21 Sugen, Inc. Treatment of excessive osteolysis with indolinone compounds
SI1928858T1 (sl) 2005-09-19 2009-10-31 Pfizer Prod Inc Trde solne oblike s pirolom substituiranega 2-indolinona

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7211600B2 (en) * 1999-12-22 2007-05-01 Sugen Inc. Methods of modulating c-kit tyrosine protein kinase function with indolinone compounds
US6573293B2 (en) * 2000-02-15 2003-06-03 Sugen, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
US7125905B2 (en) * 2000-02-15 2006-10-24 Agouron Pharmaceuticals, Inc. Pyrrole substituted 2-indolinone protein kinase inhibitors
US20070010569A1 (en) * 2000-02-15 2007-01-11 Sugen, Inc. & Pharmacia & Upjohn Co. Pyrrole substituted 2-indolinone protein kinase inhibitors
US7119209B2 (en) * 2002-02-15 2006-10-10 Pharmacia & Upjohn Company Process for preparing indolinone derivatives
US7452913B2 (en) * 2003-02-24 2008-11-18 Pharmacia & Upjohn Company Polymorphs of pyrrole substituted 2-indolinone protein kinase inhibitors
US20090048327A1 (en) * 2003-02-24 2009-02-19 Pharmacia & Upjohn Company Polymorphs of Pyrrole Substituted 2-Indolinone Protein Kinase Inhibitors
US20060009510A1 (en) * 2004-07-09 2006-01-12 Pharmacia & Upjohn Company Llc Method of synthesizing indolinone compounds

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE202010017129U1 (de) 2009-01-23 2011-09-14 Teva Pharmaceutical Industries Ltd. Rasagilinformulierung mit verzögerter Freisetzung

Also Published As

Publication number Publication date
CN103601720B (zh) 2016-08-10
EP1928858A1 (en) 2008-06-11
CA2621569C (en) 2010-10-19
AU2006293644B2 (en) 2012-05-31
PT1928858E (pt) 2009-09-17
CN103601720A (zh) 2014-02-26
NO20080866L (no) 2008-03-13
WO2007034272A1 (en) 2007-03-29
AR056521A1 (es) 2007-10-10
RU2008109959A (ru) 2009-10-27
AU2006293644A1 (en) 2007-03-29
CN101287724B (zh) 2013-11-06
CA2621569A1 (en) 2007-03-29
NZ566033A (en) 2011-04-29
CY1109326T1 (el) 2014-07-02
SI1928858T1 (sl) 2009-10-31
ZA200801431B (en) 2009-07-29
US20150158849A1 (en) 2015-06-11
BRPI0616374A2 (pt) 2011-06-21
PL1928858T3 (pl) 2009-12-31
US20120142749A1 (en) 2012-06-07
JP2007084540A (ja) 2007-04-05
JP4519114B2 (ja) 2010-08-04
MX2008002415A (es) 2008-03-27
EP1928858B1 (en) 2009-07-22
TWI310766B (en) 2009-06-11
HK1125101A1 (en) 2009-07-31
BRPI0616374B8 (pt) 2021-05-25
RU2399619C2 (ru) 2010-09-20
DE602006008015D1 (de) 2009-09-03
IL189590A0 (en) 2008-08-07
KR101050906B1 (ko) 2011-07-20
KR20080037093A (ko) 2008-04-29
US8987320B2 (en) 2015-03-24
ES2328407T3 (es) 2009-11-12
JP2010090152A (ja) 2010-04-22
TW200745093A (en) 2007-12-16
BRPI0616374B1 (pt) 2020-05-12
CN101287724A (zh) 2008-10-15
EP2112148A1 (en) 2009-10-28
DK1928858T3 (da) 2009-10-26
US9290479B2 (en) 2016-03-22

Similar Documents

Publication Publication Date Title
KR101749192B1 (ko) 트리아진, 피리미딘 및 피리딘 유사체 및 이의 치료제 및 진단 프로브로의 용도
CN109970745B (zh) 取代的吡咯并三嗪类化合物及其药物组合物及其用途
EA035680B1 (ru) Малеатные соли ингибитора braf киназы, кристаллические формы, способы их получения и применения
US9290479B2 (en) Solid salt forms of a pyrrole substituted 2-indolinone
US9884856B2 (en) Crystal form of Dabrafenib mesylate and preparation method thereof
CN108947974B (zh) 一种氨基嘧啶类化合物及包含该化合物的组合物及其用途
WO2020244612A1 (zh) Cdk9抑制剂的多晶型物及其制法和用途
CN110256408B (zh) 一种二芳基吡唑化合物及包含该化合物的组合物及其用途
WO2019178801A1 (zh) 一类新型的苯氧基哌啶衍生物及其用途
CN111138426B (zh) 吲唑类激酶抑制剂及其用途
WO2014177011A1 (zh) 二氢吲哚酮衍生物的二马来酸盐及其多晶型物
WO2020168963A1 (zh) 取代的芳香稠合环衍生物及其组合物及用途
BR112017020945B1 (pt) Sais cristalinos de sesqui-maleato de um inibidor de quinase b-raf, método e processo de preparação e usos dos mesmos

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: PFIZER INC., NEW YORK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PFIZER PRODUCTS INC.;REEL/FRAME:029041/0099

Effective date: 20120928

AS Assignment

Owner name: PFIZER INC., NEW YORK

Free format text: CORRECTION BY DECLARATION OF INCORRECT NUMBERS RECORDED AT REEL 029041 FRAME 0099;ASSIGNOR:PFIZER INC.;REEL/FRAME:029926/0644

Effective date: 20130131