WO2010129509A1 - Vinyl indazolyl compounds - Google Patents

Vinyl indazolyl compounds Download PDF

Info

Publication number
WO2010129509A1
WO2010129509A1 PCT/US2010/033487 US2010033487W WO2010129509A1 WO 2010129509 A1 WO2010129509 A1 WO 2010129509A1 US 2010033487 W US2010033487 W US 2010033487W WO 2010129509 A1 WO2010129509 A1 WO 2010129509A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
inhibition
cells
vinyl
Prior art date
Application number
PCT/US2010/033487
Other languages
French (fr)
Inventor
Daohong Chen
Hong-Yu Li
Genshi Zhao
Original Assignee
Eli Lilly And Company
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to SG2011081627A priority Critical patent/SG175909A1/en
Priority to KR1020117026320A priority patent/KR101373910B1/en
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Priority to SI201030191T priority patent/SI2427449T1/en
Priority to JP2012509889A priority patent/JP5555314B2/en
Priority to NZ595553A priority patent/NZ595553A/en
Priority to ES10717402T priority patent/ES2408117T3/en
Priority to EP10717402A priority patent/EP2427449B1/en
Priority to UAA201112804A priority patent/UA104756C2/en
Priority to DK10717402.1T priority patent/DK2427449T3/en
Priority to CN201080020264.3A priority patent/CN102421769B/en
Priority to AU2010246114A priority patent/AU2010246114B2/en
Priority to EA201171367A priority patent/EA018149B1/en
Priority to MX2011011342A priority patent/MX2011011342A/en
Priority to RS20130207A priority patent/RS52795B/en
Priority to BRPI1013748A priority patent/BRPI1013748A2/en
Priority to MA34312A priority patent/MA33244B1/en
Priority to MEP-2013-32A priority patent/ME01462B/en
Priority to PL10717402T priority patent/PL2427449T3/en
Priority to CA2760535A priority patent/CA2760535C/en
Publication of WO2010129509A1 publication Critical patent/WO2010129509A1/en
Priority to TNP2011000545A priority patent/TN2011000545A1/en
Priority to IL216004A priority patent/IL216004A/en
Priority to HK12103968.8A priority patent/HK1163665A1/en
Priority to HRP20130323AT priority patent/HRP20130323T1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Fibroblast growth factor has been recognized as an important mediator of many physiological processes, such as morphogenesis during development and angiogenesis.
  • the fibroblast growth factor receptor (FGFR) family consists of four members (FGFR 1-FGFR4), which are glycoproteins composed of extracellular immunoglobulin (Ig)-like domains, a hydrophobic transmembrane region and a cytoplasmic part containing a tyrosine kinase domain. FGF binding leads to FGFR dimerization, followed by receptor autophosphorylation and activation of downstream signaling pathways. Receptor activation is sufficient for the recruitment and activation of specific downstream signaling partners that participate in the regulation of diverse processes such as cell growth, cell metabolism and cell survival.
  • Ig immunoglobulin
  • the FGF/FGFR signaling pathway has pleiotropic effects on many biological processes critical to tumor cell proliferation, migration, invasion, and angiogenesis.
  • Vinyl indazoles are known in the art for the treatment of cancer. See for example, WO0210137 and WO2003101968.
  • FGFR inhibitors are also known in the art. See for example, WO2002022598.
  • the present invention provides novel vinyl indazolyl compounds believed to have clinical use for treatment of proliferative disorders such as cancer and particularly in disorders mediated by FGF and/or FGFR dysregulation.
  • certain compounds of the present invention have superior FGFRl and FGFR3 potency compared to certain previously known FGFR inhibitors.
  • the present invention provides a compound which is (E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)- lH-indazol-3-yl)vinyl)- lH-pyrazol- 1 -yl)ethanol or a pharmaceutically acceptable salt thereof.
  • the present invention also provides the compound which is (R)-(E)-2-(4-(2- (5 -( 1 -(3 ,5 -dichloropyridin-4-yl)ethoxy)- 1 H-indazol-3 -yl)vinyl)- 1 H-pyrazol- 1 - yl)ethanol or a pharmaceutically acceptable salt thereof.
  • the present invention provides a method of treating cancer wherein the cancer is selected from the group consisting of breast cancer, non-small cell lung (NSCL) cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer in a mammal comprising administering to a mammal in need of such treatment an effective amount of a compound or salt of the present invention.
  • This invention also provides pharmaceutical compositions comprising a compound or salt of the present invention in combination with one or more pharmaceutically acceptable carriers, diluents, or excipients. In a particular embodiment the composition further comprises one or more other therapeutic agents.
  • This invention also provides a compound or salt of the present invention for use in therapy. Additionally, this invention provides use of a compound or salt of the present invention in the manufacture of a medicament for treating cancer.
  • these cancers are selected from the group consisting of breast cancer, lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma AML, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer.
  • the cancers are selected from the group consisting of breast cancer, non- small cell lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer.
  • the cancer is non-small cell lung cancer.
  • the cancer is gastric cancer.
  • the cancer is multiple myeloma.
  • this invention provides a pharmaceutical composition for treating cancer selected from the group consisting of breast cancer, non-small cell lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer comprising a compound or salt of the present invention as an active ingredient.
  • cancer selected from the group consisting of breast cancer, non-small cell lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer comprising a compound or salt of the present invention as an active ingredient.
  • racemic (E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol can be made essentially as described for (R)-(E)-2-(4-(2-(5-(l-(3,5-dichloropyridin-4- yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol starting from racemic 1- (3,5-dichloropyridin-4-yl)ethanol in place of (S)-I -(3,5-dichloropyridin-4-yl)ethanol.
  • (E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)- 1 H-indazol-3 -yl)vinyl)- 1 H-pyrazol- 1 -yl)ethanol contains one chiral center. It is preferred that (E)-2-(4-(2-(5-(l-(3,5-dichloropyridin- 4-yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol exists as a single enantiomer.
  • the single enantiomer may be prepared beginning with chiral reagents or by stereoselective or stereospecific synthetic techniques. Alternatively, the single enantiomer may be isolated from mixtures by standard chiral chromatographic or crystallization techniques. It will be understood by the skilled reader that all of the compounds of the present invention are capable of forming salts.
  • the compounds of the present invention are amines, and accordingly react with any of a number of inorganic and organic acids to form pharmaceutically acceptable acid addition salts.
  • Such pharmaceutically acceptable acid addition salts and common methodology for preparing them are well known in the art. See, e.g., P.
  • the compounds of the present invention can be prepared as illustrated in the preparations and examples below.
  • the activating mutations in the FGFR kinase domain have been found in several types of tumors, including breast, NSCLC, bladder, gastric, prostate, colon, and multiple myeloma. Genomic amplification of FGFR locus was also detected in many breast, gastric, and multiple myeloma cancer patients. Over-expression of FGFRs and FGFs has also been found in many different types of tumors such as bladder, multiple myeloma, prostate, and lung cancers. Other cancers that might benefit from FGFR family pathway inhibitor therapy include AML, liver cancer, melanoma, head and neck cancer, thyroid cancer, pancreatic cancer, renal cell cancer, glioblastoma, and testicular cancer.
  • FGFs and FGFRs are also key regulators of angiogenesis, especially during tumor growth.
  • the FGF/FGFR axis also plays an important role in augmenting other tumor stromal cells such as cancer associated fibroblasts. Up-regulation of FGFs also leads to resistance to anti- angiogenic and other chemo-therapies.
  • small molecule inhibitors of FGFRs have demonstrated anti-tumor activities in several preclinical tumor models and are being explored in the clinic. Taken together, the FGF/FGFR pathway is essential to several important cellular processes in cancer cells. For these reasons, therapies targeting FGFRs and/or FGF signaling may affect both the tumor cells directly and tumor angiogenesis.
  • FGFRl Enzyme Assay (Filter Binding)
  • the FGFR3 Enzyme Assay (Filter Binding)
  • FGF9 induced p-ERK in RT- 112 cell based assay (in the presence of BSA)
  • the AlphaScreen SureFire Detection of ERK phosphorylation (Thr202/Tyr204) in Human Umbilical Vein Endothelial Cells (HUVEC) cell based assays
  • FGFR in vivo target inhibition assay and RTl 12 human bladder and other cancer xenograft Models.
  • FGFRl and FGFR3 Enzyme Assay (Filter Binding)
  • FGFRl or FGFR3 kinase (0.15 ng/ ⁇ L human FGFRl or 0.32 ng/ ⁇ L human FGFR3) is incubated in 50 ⁇ L of a buffer containing 10 mM 4-(2-hydroxyethyl)- 1 - piperazineethane-sulfonic acid (HEPES ) pH 7.5, 8 mM tris(hydroxymethyl)aminomethane (Tris-HCl), pH 7.5, 5.0 mM dithiothreitol (DTT), 10.0 ⁇ M adenosine triphosphate (ATP), 10 mM MnCl 2 , 150 mM NaCl, 0.01% TRITON® X-100, 0.5 ⁇ Ci 33 P-ATP, and 0.05 ⁇ g/ ⁇ L Poly(Glu-Tyr).
  • HEPES 4-(2-hydroxyethyl)- 1 - piperazineethane-sulfonic acid
  • Tris-HCl tris(hydroxy
  • the reaction is carried out in a volume of 50 ⁇ L at RT for 30 minutes and then quenched by adding 130 ⁇ L of 10% H 3 PO 4 .
  • the reaction 120 ⁇ L is transferred to a 96 well 1.0 ⁇ m glass fiber filter plate, incubated at RT for 20-30 minutes and then washed 3x on a
  • TM of MicroScint 20 (Packard) and then counted on a Wallac Micobeta counter.
  • compounds are provided as 10 mM stocks in dimethyl sulfoxide (DMSO).
  • DMSO dimethyl sulfoxide
  • Compounds are serially diluted 1 :3 in 20% DMSO to create a 10 point concentration-response curve and diluted 1 :5 (20 ⁇ M to 0.001 ⁇ M final in 4% final DMSO concentration) into the reaction plate prior to addition of the reaction mixture in the filter plate to determine compound activity.
  • Control wells contain 4% DMSO only while the baseline is established by control wells containing 0.1 M ethylenediaminetetraacetic acid (EDTA).
  • EDTA ethylenediaminetetraacetic acid
  • the percent inhibition values for each of the 10 concentrations are calculated from control wells on each plate and the 10-point concentration response data are subsequently analyzed using ActivityBase software (IDBS) using a 4-parameter logistic equation and absolute IC 50 values estimated from the resulting curve fit.
  • IDBS ActivityBase software
  • the FGFRl and FGFR3 enzyme assays have Minimum Significant Ratios (MSR) for the estimated IC50 of 1.38 and 1.47, respectively.
  • MSR Minimum Significant Ratios
  • the IC 50 results for Example 1 for FGFRl and FGFR3 in these assays are estimated to be 0.0077 and 0.0064 ⁇ M, respectively. This data demonstrates that certain compounds of the present invention are potent FGFRl and FGFR3 enzyme inhibitors.
  • FGF9 induced p-ERK with BSA Human RTl 12 bladder carcinoma cells are seeded at a density of 5,000 cells per well in 100 ⁇ L RPMI 1640 (Gibco 11875-085) supplemented with 10% fetal bovine serum (FBS, Gibco 10082-147) and 1% of a penicillin/streptomycin solution (Gibco 15140-122) in CELLBIND® 96 well plates (Corning 3340) and incubated overnight at 37 0 C. The next morning the growth medium is removed and replaced with 100 ⁇ L RPMI 1640 supplemented with 20 mg/mL bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • Cells are fixed by the addition of 30 ⁇ L of a 25% formaldehyde solution in phosphate buffered saline (PBS) (3.7% formaldehyde final concentration), and incubated 30 minutes at RT. Cells are washed 3x with PBS, followed by the addition of 100 ⁇ L of cold methanol and incubated for 30 minutes at -20 0 C. The methanol is removed and the cells are treated with PBS containing 0.1% TRITON® X-100 (PBST), washed 3x with PBS, and incubated 15 minutes at RT.
  • PBS phosphate buffered saline
  • Cells are then incubated overnight at 4 0 C with gentle shaking in 50 ⁇ L of a 1 :400 dilution of the p-p44/42 MAPK primary antibody (Cell Signaling 9101S) in PBS supplemented with 2% BSA, 0.01% Phosphatase Inhibitor Cocktail 1 (Sigma P2850), 0.01% Phosphatase Inhibitor Cocktail 2 (Sigma P5726), and 0.01% Protease Inhibitor Cocktail (Sigma P8340).
  • the total mean intensity for pERK/well using the Alexa 488 values is subsequently converted to percent inhibition using values obtained from MIN (10 ⁇ M positive control compound in DMSO) and MAX (DMSO alone) controls run on the same plate.
  • the percentage inhibition values and the 10-point concentration response data are subsequently analyzed using a 4-parameter sigmoidal dose response equation and relative IC50 values are estimated from the resulting curve.
  • the FGF9 induced p-ERK with BSA assay has a Minimum Significant Ratio (MSR) for the estimated IC50 of 2.7.
  • the IC50 for Example 1 in this assay is estimated to be 0.0004 ⁇ M.
  • Umbilical Vein Endothelial Cells The effect of compounds on the inhibition of FGF receptor 1 is measured by monitoring the phosphorylation of ERK (pERK) in response to basic-Fibroblast growth factor (b-FGF) stimulation in Human Umbilical Endothelial cells (HUVEC).
  • the levels of pERK formed are measured using the ALPHASCREEN® SUREFIRE ® system (TGR Biosciences, TGRES50K). This is a homogeneous assay format utilizing the immuno-sandwich capture of the phosphorylated analyte followed by detection using antibody-coated ALPHASCREEN® beads (Perkin Elmer) to generate an amplified signal.
  • HUVEC cells are recovered and maintained in growth medium consisting of endothelial cell basal medium (Clonetics, CC-3132) supplemented with 10% FBS 0.4% bovine brain extract 0.1% hydrocortisone, 0.1% gentamicin sulfate amphotericin-B, and 0.1% epidermal growth facter, human recombination until passage 7.
  • endothelial cell basal medium Clonetics, CC-3132
  • bovine brain extract 0.1% hydrocortisone, 0.1% gentamicin sulfate amphotericin-B, and 0.1% epidermal growth facter, human recombination until passage 7.
  • FBS endothelial cell basal medium
  • bovine brain extract 0.1% hydrocortisone
  • 0.1% gentamicin sulfate amphotericin-B 0.1% epidermal growth facter, human recombination until passage 7.
  • epidermal growth facter human recombination until passage 7.
  • cells are harvested
  • EBM endothelial cell basal
  • BSA endothelial cell basal
  • 20 ⁇ M of 3x serially diluted compounds in starvation medium for 1 hour at 37 0 C.
  • cells are stimulated with 50 ⁇ L b-FGF (Sigma, F0291, final b-FGF concentration 50 ng/mL) at 37 0 C for 15 minutes.
  • the plate is sealed and incubated at RT for 2 hours with gentle shaking and then read on Perkin Elmer EnVision plate reader equipped with a TurboModule using standard ALPHASCREEN® settings (Ex680 nm and Em 52 o-6 2 ⁇ nm )-
  • the emission data is converted to percent inhibition determined from MAX (DMSO alone) and MIN (10 ⁇ M positive control compound in DMSO) controls on each plate and ten-point compound concentration data are then fit to a four-parameter logistic equation using ACTIVITYBASE® 4.0 and the IC 50 is estimated.
  • the ALPHASCREEN® SUREFIRE® Detection of ERK phosphorylation ((Thr202/Tyr204) assay has a Minimum Significant Ratio (MSR) for the IC 50 of 2.1.
  • MSR Minimum Significant Ratio
  • the IC50 of Example 1 in this assay is estimated to be 0.0006 ⁇ M.
  • mice Female nude mice (CDl/nu/nu) are acclimated for 1 week prior to treatment. Animals are grouped into positive control, negative control and compound treatment groups. Compound (formulated in 10% Acacia), positive control (10% Acacia), and negative control (10% Acacia) are administered by oral gavage. Compound doses are in the range of 0.15 to 25 mg/kg. After 2 hours, the compound treatment group and the positive control group are treated with freshly prepared mouse bFGF (6 ⁇ g/animal, Biosource PMG0033) in saline administered intravenously. The negative control group is treated with saline administered intravenously. Mice are sacrificed 10 minutes post intravenous dose.
  • Animal heart is harvested and homogenized for 10 seconds in 300 ⁇ L of ice cold lysis buffer (RIPA; Boston BioProduct BP-115) containing 1 : 100 diluted of inhibitors (phosphotase inhibitor cocktail I Sigma P2850; phosphotase inhibitor cocktail II Sigma P5726 and protease inhibitor cocktail Sigma P8340). Homogenates are centrifuged at 14,000 RPM for 15 minutes and supernatants are transferred to a 96-well plate. Protein level is determined by COOMASSIE PLUSTM protein assay method (Pierce # 1856210). Assay procedures are the same per manufacturer's recommendation (see instruction booklet included in the assay kit).
  • Heart tissue homogenates are analyzed using MSD® phospho-Erk ELISA (Meso Scale Discovery, catalog number N4 ICB-I) to determine tissue phospho-Erk level.
  • MSD® phospho-Erk ELISA Meso Scale Discovery, catalog number N4 ICB-I
  • the ELISA procedures are the same per manufacturer's recommendation (see instruction booklet included in the assay kit; the only modification is that 0.2% sodium dodecyl sulfate is added to the lysis buffer).
  • Positive control is used as minimum phospho-Erk inhibition (0%) and the negative control is used as maximum phospho-Erk inhibition (100%).
  • Percent inhibition of compound treated groups is calculated relative to the maximum and minimum inhibition groups.
  • TEC 9 0 is calculated from a dose response study and is the concentration necessary to achieve 90% inhibition at this time point.
  • the compound of Example 1 has an estimated TEC90 of 28 nM.
  • This data demonstrates that certain compounds of the present invention are potent inhibitors of bFGF induced ERK phosphorylation in vivo.
  • VEGF is used to induce Kdr autophosphorylation ⁇ VEGF (6 ug/animal, R & D Systems 493-MV/CF).
  • Heart tissues are collected and homogenized as described above.
  • the resulting homogenates are analyzed using MSD® phospho-Kdr ELISA (Meso Scale Discovery, catalog number N4 IZA-I) to determine tissue phospho-Kdr level.
  • the ELISA procedures are the same per manufacturer's recommendation (see instruction booklet included in the assay kit; the only modification is that 0.2% sodium dodecyl sulfate is added to the lysis buffer).
  • the positive control group is treated with VEGF 96 ug/animal) in saline administered intravenously (as minimum p-KDR inhibition of 0%).
  • the negative control group is treated with saline administered intravenously (as maximum p-KDR inhibition of 100%). Percent inhibition of compound treated groups is calculated relative to the maximum and minimum inhibition groups.
  • TED50 is calculated from a dose response study and is the dose necessary to achieve 50% inhibition at this time point.
  • the compound of Example 1 has an estimated TED 5 O of 1.34 mg/kg.
  • the TEC 9 0 is calculated from a dose response study and is the concentration necessary to achieve 90% inhibition at this time point.
  • the compound of Example 1 has an estimated TEC 9 0 of 252 nM.
  • Human bladder cancer cells RTl 12 European Collection of Cell Cultures
  • human multiple myeloma cells OPM-2 German Collection of Microorganisms and Cell Cultures
  • human non-small cell lung cancer (NSCL) cells NCI-H460 (American Type Culture Collection)
  • human pancreatic cancer cells BxPC- 3 American Type Culture Collection
  • Example 1 demonstrated dose dependent anti-tumor activity in several xenograft tumor models.
  • NSCLC tumor model NSCLC tumor model (NCI-H460), when dosed at 3 mg/kg (QD for 17 days), 46% inhibition was achieved; when dosed at 3 mg/kg (BID for 17 days), 69% inhibition was achieved.
  • pancreatic tumor model BxPC-3
  • BxPC-3 pancreatic tumor model
  • the compounds of the present invention are preferably formulated as pharmaceutical compositions administered by a variety of routes. Most preferably, such compositions are for oral or intravenous administration. Such pharmaceutical compositions and processes for preparing same are well known in the art. See, e.g., REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY (D. Troy, et al, eds., 21 st ed., Lippincott Williams & Wilkins, 2005).
  • the compounds of the present invention are generally effective over a wide dosage range. For example, dosages per day normally fall within the range of about 0.5 to about 100 mg/kg of body weight.
  • dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, and therefore the above dosage range is not intended to limit the scope of the invention in any way.
  • the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound or compounds administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms.

Abstract

The present invention provides vinyl indazoiyl compounds useful in the treatment of cancer.

Description

VINYL INDAZOLYL COMPOUNDS
Fibroblast growth factor (FGF) has been recognized as an important mediator of many physiological processes, such as morphogenesis during development and angiogenesis. The fibroblast growth factor receptor (FGFR) family consists of four members (FGFR 1-FGFR4), which are glycoproteins composed of extracellular immunoglobulin (Ig)-like domains, a hydrophobic transmembrane region and a cytoplasmic part containing a tyrosine kinase domain. FGF binding leads to FGFR dimerization, followed by receptor autophosphorylation and activation of downstream signaling pathways. Receptor activation is sufficient for the recruitment and activation of specific downstream signaling partners that participate in the regulation of diverse processes such as cell growth, cell metabolism and cell survival. Thus, the FGF/FGFR signaling pathway has pleiotropic effects on many biological processes critical to tumor cell proliferation, migration, invasion, and angiogenesis. Vinyl indazoles are known in the art for the treatment of cancer. See for example, WO0210137 and WO2003101968. FGFR inhibitors are also known in the art. See for example, WO2002022598.
The present invention provides novel vinyl indazolyl compounds believed to have clinical use for treatment of proliferative disorders such as cancer and particularly in disorders mediated by FGF and/or FGFR dysregulation.
Additionally, certain compounds of the present invention have superior FGFRl and FGFR3 potency compared to certain previously known FGFR inhibitors.
The present invention provides a compound which is (E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)- lH-indazol-3-yl)vinyl)- lH-pyrazol- 1 -yl)ethanol or a pharmaceutically acceptable salt thereof.
The present invention also provides the compound which is (R)-(E)-2-(4-(2- (5 -( 1 -(3 ,5 -dichloropyridin-4-yl)ethoxy)- 1 H-indazol-3 -yl)vinyl)- 1 H-pyrazol- 1 - yl)ethanol or a pharmaceutically acceptable salt thereof.
The present invention provides a method of treating cancer wherein the cancer is selected from the group consisting of breast cancer, non-small cell lung (NSCL) cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer in a mammal comprising administering to a mammal in need of such treatment an effective amount of a compound or salt of the present invention. This invention also provides pharmaceutical compositions comprising a compound or salt of the present invention in combination with one or more pharmaceutically acceptable carriers, diluents, or excipients. In a particular embodiment the composition further comprises one or more other therapeutic agents. This invention also provides a compound or salt of the present invention for use in therapy. Additionally, this invention provides use of a compound or salt of the present invention in the manufacture of a medicament for treating cancer. In particular these cancers are selected from the group consisting of breast cancer, lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma AML, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer. More particularly, the cancers are selected from the group consisting of breast cancer, non- small cell lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer. Most particularly the cancer is non-small cell lung cancer. Most particularly the cancer is gastric cancer. Most particularly the cancer is multiple myeloma. Furthermore, this invention provides a pharmaceutical composition for treating cancer selected from the group consisting of breast cancer, non-small cell lung cancer, bladder cancer, gastric cancer, pancreatic cancer, prostate cancer, colon cancer, multiple myeloma, liver cancer, melanoma, head and neck cancer, thyroid cancer, renal cell cancer, glioblastoma, and testicular cancer comprising a compound or salt of the present invention as an active ingredient.
The skilled artisan will appreciate that racemic (E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol can be made essentially as described for (R)-(E)-2-(4-(2-(5-(l-(3,5-dichloropyridin-4- yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol starting from racemic 1- (3,5-dichloropyridin-4-yl)ethanol in place of (S)-I -(3,5-dichloropyridin-4-yl)ethanol. Additionally, the skilled artisan will appreciate that (E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)- 1 H-indazol-3 -yl)vinyl)- 1 H-pyrazol- 1 -yl)ethanol contains one chiral center. It is preferred that (E)-2-(4-(2-(5-(l-(3,5-dichloropyridin- 4-yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol exists as a single enantiomer. The single enantiomer may be prepared beginning with chiral reagents or by stereoselective or stereospecific synthetic techniques. Alternatively, the single enantiomer may be isolated from mixtures by standard chiral chromatographic or crystallization techniques. It will be understood by the skilled reader that all of the compounds of the present invention are capable of forming salts. The compounds of the present invention are amines, and accordingly react with any of a number of inorganic and organic acids to form pharmaceutically acceptable acid addition salts. Such pharmaceutically acceptable acid addition salts and common methodology for preparing them are well known in the art. See, e.g., P. Stahl, et ah, HANDBOOK OF PHARMACEUTICAL SALTS: PROPERTIES, SELECTION AND USE, (VCHA/Wiley-VCH, 2008); S.M. Berge, et ah, "Pharmaceutical Salts", Journal of Pharmaceutical Sciences, VoI 66, No. 1, January 1977.
The compounds of the present invention can be prepared as illustrated in the preparations and examples below. The naming of the following preparations and examples is done using the Struct=Name naming feature in ChemDraw® Ultra 10.0.
Preparation 1 l-(3,5-Dichloropyridin-4-yl)ethanol To a 3 -neck 12 L round bottom flask add tetrahydrofuran (THF, 3 L) and diisopropylamine (DIPA, 315 mL, 2.24 mol) and cool to -78 0C. Add slowly n- butyllithium (1.6 M in hexanes, 1400 mL, 2.24 mol). After the addition is complete and the temperature has settled at -78 0C slowly add a solution of 3,5-dichloropyridine (296.7 g, 2.00 mol) which immediately forms a yellow solution that changes to a rust colored suspension. After the addition is complete and the temperature has settled at -78 0C slowly add acetaldehyde (230 mL, 4.05 mol) in THF (600 mL). Continue -A-
stirring at -78 0C. After 3 hours, remove the dry ice bath and begin quenching the reaction by the dropwise addition of saturated aqueous ammonium chloride (1 L). Allow the reaction to warm to room temperature (RT) overnight with stirring. Dilute the mixture with methyl-tert-butylether (MTBE, 2 L), saturated aqueous ammonium chloride (1 L) and water (2 L). Partition and wash organics with saturated aqueous sodium chloride (brine). Extract the aqueous phase with MTBE (1.5 L). Combine the organic layers, dry over sodium sulfate, filter and concentrate in vacuo. Purify the residue by silica gel chromatography [25% ethylacetate (EA) in hexanes] to give the title compound as a red oil. Yield: 352 g (90%). MS (ES) m/z 192 [M+l]+.
Preparation 2
(S)- 1 -(3,5-Dichloropyridin-4-yl)ethanol Separate the mixture of stereoisomers obtained in the above reaction on a CHIRALPAK® AD-H column eluting with 90% heptanes/ 10% ethanol. Peak 2 is the desired enantiomer. To establish the absolute configuration dissolve a sample of the product in CDCI3 (final concentration 100 mg/mL). Obtain the vibrational circular dichroism (VCD) and infra red (IR) spectra with a resolution of 4 cm- 1 using a ChiralIR FT VCD spectrometer (BioTools Inc ®) with an IR cell equipped with BaF2 windows and a path length of 100 mm. Collect the VCD and IR for 6 hours with 150 μL of the sample. Present the data without smoothing or further data processing. Obtain vibrational frequencies and absorption and VCD intensities by optimizing the lowest energy conformer by Gaussian at the B3PW91/6-3 IG** level on a Linux cluster, and simulate the corresponding spectra using a Lorentzian bandwidth of 6 cm- 1 vibrational circular dichroism. The above analysis shows the product to be the S- isomer. Yield: 84.37 g (27%). MS (ES) m/z 192 [MH-I]+.
Preparation 3
(S)- 1 -(3,5-Dichloropyridin-4-yl)ethyl methanesulfonate Dissolve (S)- l-(3,5-dichloropyridin-4-yl)ethanol (5.02 g, 26.14 mmol) in dichloromethane (DCM, 100 mL) and cool the flask in an ice bath. Add triethylamine (TEA, 3.5 mL, 25.11 mmol) followed by the dropwise addition of methanesulfonyl chloride (2.2 mL, 28.42 mmol). Remove the ice bath and allow the reaction to warm to RT. After 4 hours, quench the reaction with water (100 mL) and separate layers. Extract the aqueous layer with DCM (50 mL) followed by 20% isopropyl alcohol (IPA)/chloroform (50 mL). Combine the organic extracts, dry over anhydrous sodium sulfate, filter and concentrate in vacuo. Yield: 7.15 g, (100%). MS (ES) m/z 270 [M+ 1]+.
Preparation 4 4-Iodo- 1 -(2-(tetrahydro-2H-pyran-2-yloxy)ethyl)- 1 H-pyrazole
In a 1 L 3 -neck flask equipped with magnetic stir bar, nitrogen blanket and internal temperature probe dissolve 2-(2-bromoethoxy)tetrahydro-2H-pyran (34 g, 156 mmol) in acetonitrile (ACN, 400 mL). Add 4-iodopyrazole (29.34 g, 149.74 mmol) followed by cesium carbonate (73.4 g, 223.02 mmol). Stir the mixture at RT for 18 hours. Filter the reaction mixture through CELITE®, wash the filter cake with ACN and concentrate the filtrate to a golden oil. Use without further purification. Yield: 47.819 g (99%). MS (ES) m/z 323 [M+ 1]+.
Preparation 5 5-(tert-Butyldimethylsilyloxy)- lH-indazole
Charge a 10 L reaction vessel with N,N-dimethylformamide (DMF, 2.50 L), 5 -hydroxy indazole (150.20 g, 1.12 mol) and lH-imidazole (114.35 g, 1.68 mol). Cool the mixture to 0 0C and add tert-butyldimethylchlorosilane (253.16 g, 1.68 mol) over 0.5 hours. Stir the mixture at 18 0C for 3 hours. Add water (2.5 L) to the reaction slowly with an ice bath at 5 0C to maintain an internal temperature at around 20 0C. Transfer the mixture to a separating funnel and extract with EA (2 x 2.5 L). Combine the extracts and wash with water (3 x 2.5 L) and brine. Dry the organic solutions over anhydrous sodium sulfate, filter, and evaporate to a red oil. Pass the oil through a silica gel pad and elute with eluent (0% to 30% EA in hexane) to afford the title compound as an orange oil which crystallizes. Yield: 300 g (100%). MS (ES) m/z 249 [M+l]+. Preparation 6
5-(tert-Butyldimethylsilyloxy)-3-iodo-lH-indazole
Cool a solution of 5-(tert-butyldimethylsilyloxy)-lH-indazole (300.00 g, 1.21 mol) in DCM (4.00 L) to 10 0C in a 10 L jacketed reactor vessel. To the resulting solution add N-iodosuccinimide (298.89 g, 1.33 mol) in portions over 0.5 hours. Stir the mixture at RT for 3 hours to give complete conversion as indicated by liquid chromatography mass spectrometry (LC-MS) and thin layer chromatography (TLC). Cool the mixture to 10 0C and quench with water (2.5 L). Transfer the mixture to a separatory funnel and extract the aqueous layer into DCM (2.5 L). Wash the combined organic extracts with a 10% aqueous sodium thiosulfate solution (5 L) and brine. Dry the organic solution over magnesium sulfate, filter and concentrate in vacuo to afford the title compound as an orange solid. Yield: 388 g (90%). MS (ES)
Figure imgf000007_0001
Preparation 7
5-(tert-Butyldimethylsilyloxy)-3-iodo-l-(tetrahydro-2H-pyran-2-yl)-lH-indazole Cool a solution of 5-(tert-butyldimethylsilyloxy)-3-iodo-lH-indazole (387.00 g, 1.08 mol) in DCM (2.50 L) and THF (1.00 L) to 10 0C in a 10 L jacketed reactor vessel. To the resulting mixture add methanesulfonic acid (14.0 mL, 216.02 mmol), followed by 3,4-dihydro-2H-pyran (296 mL, 3.24 mol) over 0.5 hours, observing a slight exotherm. Stir the mixture at RT for 3 hours. Cool the reaction to 10 0C and quench with saturated aqueous sodium bicarbonate (2 L). Dilute the mixture with water (2 L) and extract the aqueous layer with DCM (2 L). Wash the combined organic extracts with water (2 L) and brine. Dry the organic mixture over anhydrous sodium sulfate, filter and concentrate in vacuo. Elute the residue through a silica gel pad with eluent (0 to 10% EA/hexanes) to give the title compound. Yield: 150 g (31%). MS (ES) m/z 459 [M+l]+.
Preparation 8 (E)- 1 -(Tetrahydro-2H-pyran-2-yl)-3-(2-(l -(2-(tetrahydro-2H-pyran-2-yloxy)ethyl)-
1 H-pyrazol-4-yl)vinyl)- 1 H-indazol-5 -ol
Sparge with nitrogen a mixture of 5-(tert-butyldimethylsilyloxy)-3-iodo-l- (tetrahydro-2H-pyran-2-yl)-lH-indazole (14 g, 30.54 mmol) in DMF (150 mL) in a 500 mL 3 -neck round bottom flask equipped with magnetic stirring, temperature probe, and condenser with septa for 10 minutes. To the resulting solution add tributylamine (TBA, 6.7 g, 36.1 mmol) and 4,4,5, 5-tetramethyl-2-vinyl- 1,3,2- dioxaborolane (7.0 g, 43.18 mmol) and continue sparging for 10 minutes. To the resulting mixture add bis(triphenylphosphine) palladium (II) chloride (0.45 g, 0.63 mmol) and continue to sparge for an additional 0.5 hours. Heat the mixture at 95-100 0C for 18 hours. Cool the reaction mixture to below 40 0C and charge with 4-iodo-l- (2-(tetrahydro-2H-pyran-2-yloxy)ethyl)-lH-pyrazole (9.8 g, 30.42 mmol). To the resulting mixture add barium hydroxide octahydrate (19.3 g, 60.3 mmol) and water (13 mL) and continue sparging for 10 minutes. Add 1,1'- bis(diphenylphosphino)ferrocene palladium (II) chloride DCM complex (1.3 g, 1.56 mmol) to the reaction and continue sparging 0.5 hours. Heat the mixture at 95 0C under nitrogen for 3 hours. Dilute the mixture with EA and filter through a Celite® pad. Wash the pad with brine (400 mL) and separate the filtrate layers. Wash the organic layer with brine and extract the combined aqueous layers with EA. Combine the organic solutions and concentrate to a brown oil. Dissolve the oil in DCM (100 mL) and add to a silica gel pad. Elute the pad with eluent (50% EA in hexanes followed by 70% EA in hexanes) to afford a light brown oil. Triturate with MTBE (10O mL) to afford the title compound as a solid. Yield: 5 g (37%). MS (ES) m/z 439 [M+l]+.
Preparation 9
5-((R)- 1 -(3,5-Dichloropyridin-4-yl)ethoxy)- 1 -(tetrahydro-2H-pyran-2-yl)-3-((E)-2-(l - (2-(tetrahydro-2H-pyran-2-yloxy)ethyl)- 1 H-pyrazol-4-yl)vinyl)- 1 H-indazole
In a 3 -neck 250 mL round bottom flask equipped with an internal temperature probe, reflux condenser, nitrogen blanket and magnetic stir bar, slurry (E)-I-
(tetrahydro-2H-pyran-2-yl)-3-(2-(l-(2-(tetrahydro-2H-pyran-2-yloxy)ethyl)-lH- pyrazol-4-yl)vinyl)-lH-indazol-5-ol (10.0 g, 22.83 mmol) and cesium carbonate (7.8£ g, 23.94 mmol) in ACN (92 mL) and warm to 60 0C. To the suspension, add (S)-I- (3,5-dichloropyridin-4-yl)ethyl methanesulfonate (7.03 g, 26.02 mmol) and stir overnight. Cool the reaction mixture to RT, filter and wash solids with ACN. Concentrate the filtrate and purify the residue by silica gel chromatography (2-4% (2 M ammonia in methanol) /DCM). Combine product fractions and concentrate in vacuo to a white foam. Yield: 12.5 g (86%). MS (ES) m/z 612 [M+l]+.
Example 1
(R)-(E)-2-(4-(2-(5-(l-(3,5-Dichloropyridin-4-yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH- pyrazol- 1 -yl)ethanol
Figure imgf000009_0001
Charge a 3 -neck, 250 mL round bottom flask equipped with an addition funnel, nitrogen inlet, internal temperature probe and magnetic stirrer with methanol (57 mL) and cool in an ice bath. To the resulting solution, add acetyl chloride (20 mL, 281.03 mmol) slowly through an addition funnel. To the solution, add 5-((R)-I- (3,5-dichloropyridin-4-yl)ethoxy)- 1 -(tetrahydro-2H-pyran-2-yl)-3-((E)-2-(l -(2- (tetrahydro-2H-pyran-2-yloxy)ethyl)-lH-pyrazol-4-yl)vinyl)-lH-indazole (7.1 g, 11.59 mmol) dissolved in methanol (40 mL) via addition funnel. After addition is complete, remove the ice bath, warm to RT and stir the mixture for 4 hours. Concentrate the reaction mixture in vacuo to a yellow foam. Dissolve the yellow foam in methanol (10 mL) and add slowly to a saturated aqueous sodium bicarbonate solution (120 mL). Stir the mixture at RT for 30 minutes. Filter the mixture, wash the solid with water (100 mL), and dry under vacuum. Recrystallize the solid from hot EA/methanol/hexanes to give the title compound as a white solid. Yield: 2.1 g (41%). MS (ES) m/z AAA [M+ 1]+. Aberrant regulation of the FGF/FGFR pathway has been implicated in many forms of human malignancies. FGFRs and FGFs are often over-expressed in numerous cancers, and their expression often correlates with poor prognosis. The activating mutations in the FGFR kinase domain have been found in several types of tumors, including breast, NSCLC, bladder, gastric, prostate, colon, and multiple myeloma. Genomic amplification of FGFR locus was also detected in many breast, gastric, and multiple myeloma cancer patients. Over-expression of FGFRs and FGFs has also been found in many different types of tumors such as bladder, multiple myeloma, prostate, and lung cancers. Other cancers that might benefit from FGFR family pathway inhibitor therapy include AML, liver cancer, melanoma, head and neck cancer, thyroid cancer, pancreatic cancer, renal cell cancer, glioblastoma, and testicular cancer. In addition to their roles in tumor formation and progression, FGFs and FGFRs are also key regulators of angiogenesis, especially during tumor growth. The FGF/FGFR axis also plays an important role in augmenting other tumor stromal cells such as cancer associated fibroblasts. Up-regulation of FGFs also leads to resistance to anti- angiogenic and other chemo-therapies. Finally, small molecule inhibitors of FGFRs have demonstrated anti-tumor activities in several preclinical tumor models and are being explored in the clinic. Taken together, the FGF/FGFR pathway is essential to several important cellular processes in cancer cells. For these reasons, therapies targeting FGFRs and/or FGF signaling may affect both the tumor cells directly and tumor angiogenesis.
All exemplified compounds are tested essentially as described below in at least one of the following assays: FGFRl Enzyme Assay (Filter Binding), the FGFR3 Enzyme Assay (Filter Binding), FGF9 induced p-ERK in RT- 112 cell based assay (in the presence of BSA), the AlphaScreen SureFire Detection of ERK phosphorylation (Thr202/Tyr204) in Human Umbilical Vein Endothelial Cells (HUVEC) cell based assays, FGFR in vivo target inhibition assay, and RTl 12 human bladder and other cancer xenograft Models. These assays demonstrate that the tested compounds are FGFR family pathway inhibitors and have anti-cancer activity. FGFRl and FGFR3 Enzyme Assay (Filter Binding)
FGFRl or FGFR3 kinase (0.15 ng/μL human FGFRl or 0.32 ng/μL human FGFR3) is incubated in 50 μL of a buffer containing 10 mM 4-(2-hydroxyethyl)- 1 - piperazineethane-sulfonic acid (HEPES ) pH 7.5, 8 mM tris(hydroxymethyl)aminomethane (Tris-HCl), pH 7.5, 5.0 mM dithiothreitol (DTT), 10.0 μM adenosine triphosphate (ATP), 10 mM MnCl2, 150 mM NaCl, 0.01% TRITON® X-100, 0.5 μCi 33P-ATP, and 0.05 μg/μL Poly(Glu-Tyr). The reaction is carried out in a volume of 50 μL at RT for 30 minutes and then quenched by adding 130 μL of 10% H3PO4. The reaction (120 μL) is transferred to a 96 well 1.0 μm glass fiber filter plate, incubated at RT for 20-30 minutes and then washed 3x on a
TITERTEK® Zoom with 0.5% H3PO4. Wells are air dried before addition of 40 μL
TM of MicroScint 20 (Packard) and then counted on a Wallac Micobeta counter. For compound inhibition, compounds are provided as 10 mM stocks in dimethyl sulfoxide (DMSO). Compounds are serially diluted 1 :3 in 20% DMSO to create a 10 point concentration-response curve and diluted 1 :5 (20 μM to 0.001 μM final in 4% final DMSO concentration) into the reaction plate prior to addition of the reaction mixture in the filter plate to determine compound activity. Control wells contain 4% DMSO only while the baseline is established by control wells containing 0.1 M ethylenediaminetetraacetic acid (EDTA). The percent inhibition values for each of the 10 concentrations are calculated from control wells on each plate and the 10-point concentration response data are subsequently analyzed using ActivityBase software (IDBS) using a 4-parameter logistic equation and absolute IC50 values estimated from the resulting curve fit. The FGFRl and FGFR3 enzyme assays have Minimum Significant Ratios (MSR) for the estimated IC50 of 1.38 and 1.47, respectively. The IC50 results for Example 1 for FGFRl and FGFR3 in these assays are estimated to be 0.0077 and 0.0064 μM, respectively. This data demonstrates that certain compounds of the present invention are potent FGFRl and FGFR3 enzyme inhibitors.
FGF9 induced p-ERK with BSA Human RTl 12 bladder carcinoma cells are seeded at a density of 5,000 cells per well in 100 μL RPMI 1640 (Gibco 11875-085) supplemented with 10% fetal bovine serum (FBS, Gibco 10082-147) and 1% of a penicillin/streptomycin solution (Gibco 15140-122) in CELLBIND® 96 well plates (Corning 3340) and incubated overnight at 37 0C. The next morning the growth medium is removed and replaced with 100 μL RPMI 1640 supplemented with 20 mg/mL bovine serum albumin (BSA). After 3 hours of incubation at 37 0C, 20 μL of 3x serially diluted compounds in RPMI 1640 with 20 mg/mL BSA in 6% DMSO are added to each well. This yielded a 10 point dose-response curve ranging from 10 - 0.005 μM in 1% DMSO. The incubation is continued for 1 hour at 37 0C. The cells are stimulated with 50 μL of a 50 μg/mL FGF9 (R&D Systems 273 -F9) solution in serum free RPMI to give a final concentration of 500 ng/mL FGF9. Cells are fixed by the addition of 30 μL of a 25% formaldehyde solution in phosphate buffered saline (PBS) (3.7% formaldehyde final concentration), and incubated 30 minutes at RT. Cells are washed 3x with PBS, followed by the addition of 100 μL of cold methanol and incubated for 30 minutes at -20 0C. The methanol is removed and the cells are treated with PBS containing 0.1% TRITON® X-100 (PBST), washed 3x with PBS, and incubated 15 minutes at RT. Cells are then incubated overnight at 4 0C with gentle shaking in 50 μL of a 1 :400 dilution of the p-p44/42 MAPK primary antibody (Cell Signaling 9101S) in PBS supplemented with 2% BSA, 0.01% Phosphatase Inhibitor Cocktail 1 (Sigma P2850), 0.01% Phosphatase Inhibitor Cocktail 2 (Sigma P5726), and 0.01% Protease Inhibitor Cocktail (Sigma P8340). The next morning, plates are washed 2x with PBST and 2x with PBS, followed by a 1 hour RT incubation in the dark in 80 uL of a 1: 1000 dilution of the Alexa Fluor 488 goat anti-rabbit IgG H+L secondary antibody (Invitrogen Al 1034) in PBS with 1% BSA and 0.1% of Phosphatase Inhibitor
Cocktail 1, 0.01% Phosphatase Inhibitor Cocktail 2, and 0.01% Protease Inhibitor Cocktail. Cells are washed 3x with PBS, followed by the addition of 100 μL of a 1 :200 dilution of propidium iodide (PI) (Molecular Probe P-3566) in PBS and then incubated in the dark for 1 hour. The p-ERK positive cells and total cells per well are identified with the ACUMEN EXPLORER™ (TTP LabTech Ltd) using optical filter 500-530 nM and 575-640 nM for Alexa 488 and PI, respectively. The total mean intensity for pERK/well using the Alexa 488 values is subsequently converted to percent inhibition using values obtained from MIN (10 μM positive control compound in DMSO) and MAX (DMSO alone) controls run on the same plate. The percentage inhibition values and the 10-point concentration response data are subsequently analyzed using a 4-parameter sigmoidal dose response equation and relative IC50 values are estimated from the resulting curve. The FGF9 induced p-ERK with BSA assay has a Minimum Significant Ratio (MSR) for the estimated IC50 of 2.7. The IC50 for Example 1 in this assay is estimated to be 0.0004 μM. This data demonstrates that certain compounds of the present invention are potent inhibitors of FGF9 induced ERK phosphorylation in human cancer cells.
AlphaScreen SureFire Detection of ERK phosphorylation (Thr202/Tyr204) in Human
Umbilical Vein Endothelial Cells (HUVEC) The effect of compounds on the inhibition of FGF receptor 1 is measured by monitoring the phosphorylation of ERK (pERK) in response to basic-Fibroblast growth factor (b-FGF) stimulation in Human Umbilical Endothelial cells (HUVEC). The levels of pERK formed are measured using the ALPHASCREEN® SUREFIRE ® system (TGR Biosciences, TGRES50K). This is a homogeneous assay format utilizing the immuno-sandwich capture of the phosphorylated analyte followed by detection using antibody-coated ALPHASCREEN® beads (Perkin Elmer) to generate an amplified signal.
HUVEC cells are recovered and maintained in growth medium consisting of endothelial cell basal medium (Clonetics, CC-3132) supplemented with 10% FBS 0.4% bovine brain extract 0.1% hydrocortisone, 0.1% gentamicin sulfate amphotericin-B, and 0.1% epidermal growth facter, human recombination until passage 7. For the assay, cells are harvested by standard procedures and then counted. Cells (20,000/well) are plated in 100 μL of growth medium into 96 well Poly-D-Lysine coated plates (BD, 354640). Plates are incubated overnight at 37 0C, 5% CO2. On the day of the assay, cells are serum starved in 100 μL EBM (endothelial cell basal) medium containing 1.5% FBS and 20 mg/mL BSA for 3 hours at 37 0C, 5% Cθ2,then treated with 20 μM of 3x serially diluted compounds in starvation medium for 1 hour at 37 0C. This yielded a 10 point concentration-response curve ranging from 10-0.005 μM in 1% DMSO. After 1 hour compound treatment, cells are stimulated with 50 μL b-FGF (Sigma, F0291, final b-FGF concentration 50 ng/mL) at 37 0C for 15 minutes. In the wells containing cells and 50 μL stimulator b-FGF yields MAX signal, and cells with 10 μM positive control compound and 50 ul stimulator b- FGF as MIN. The medium then is removed and 50 μL of Ix SUREFIRE® Lysis Buffer (TGR Biosciences SUREFIRE ® Kit component) is added/well and incubation continued at RT for 10 minutes with gentle shaking. For pERK detection, 6 μL Iy sate and 10 μL reaction mixture (60 parts reaction buffer/10 parts activation buffer/0.6 part each of donor and acceptor beads, Perkin Elmer, 6760617R) are transferred to a 384 well proxiplate (Perkin Elmer, 6006280). The plate is sealed and incubated at RT for 2 hours with gentle shaking and then read on Perkin Elmer EnVision plate reader equipped with a TurboModule using standard ALPHASCREEN® settings (Ex680nm and Em52o-62θnm)- The emission data is converted to percent inhibition determined from MAX (DMSO alone) and MIN (10 μM positive control compound in DMSO) controls on each plate and ten-point compound concentration data are then fit to a four-parameter logistic equation using ACTIVITYBASE® 4.0 and the IC50 is estimated. The ALPHASCREEN® SUREFIRE® Detection of ERK phosphorylation ((Thr202/Tyr204) assay has a Minimum Significant Ratio (MSR) for the IC50 of 2.1. The IC50 of Example 1 in this assay is estimated to be 0.0006 μM. This data demonstrates that certain compounds of the present invention are potent inhibitors of bFGF induced ERK phosphorylation in Human Umbilical Endothelial cells.
FGFR in vivo target inhibition assay
Female nude mice (CDl/nu/nu) are acclimated for 1 week prior to treatment. Animals are grouped into positive control, negative control and compound treatment groups. Compound (formulated in 10% Acacia), positive control (10% Acacia), and negative control (10% Acacia) are administered by oral gavage. Compound doses are in the range of 0.15 to 25 mg/kg. After 2 hours, the compound treatment group and the positive control group are treated with freshly prepared mouse bFGF (6 μg/animal, Biosource PMG0033) in saline administered intravenously. The negative control group is treated with saline administered intravenously. Mice are sacrificed 10 minutes post intravenous dose. Animal heart is harvested and homogenized for 10 seconds in 300 μL of ice cold lysis buffer (RIPA; Boston BioProduct BP-115) containing 1 : 100 diluted of inhibitors (phosphotase inhibitor cocktail I Sigma P2850; phosphotase inhibitor cocktail II Sigma P5726 and protease inhibitor cocktail Sigma P8340). Homogenates are centrifuged at 14,000 RPM for 15 minutes and supernatants are transferred to a 96-well plate. Protein level is determined by COOMASSIE PLUS™ protein assay method (Pierce # 1856210). Assay procedures are the same per manufacturer's recommendation (see instruction booklet included in the assay kit).
Heart tissue homogenates are analyzed using MSD® phospho-Erk ELISA (Meso Scale Discovery, catalog number N4 ICB-I) to determine tissue phospho-Erk level. The ELISA procedures are the same per manufacturer's recommendation (see instruction booklet included in the assay kit; the only modification is that 0.2% sodium dodecyl sulfate is added to the lysis buffer). Positive control is used as minimum phospho-Erk inhibition (0%) and the negative control is used as maximum phospho-Erk inhibition (100%). Percent inhibition of compound treated groups is calculated relative to the maximum and minimum inhibition groups. TEC90 is calculated from a dose response study and is the concentration necessary to achieve 90% inhibition at this time point. For example, the compound of Example 1 has an estimated TEC90 of 28 nM. This data demonstrates that certain compounds of the present invention are potent inhibitors of bFGF induced ERK phosphorylation in vivo. To assess the activity of this compound against Kdr, female nude mice (CD 1/nu/nu) are acclimated and treated as described above except VEGF is used to induce Kdr autophosphorylation {VEGF (6 ug/animal, R & D Systems 493-MV/CF). Heart tissues are collected and homogenized as described above. The resulting homogenates are analyzed using MSD® phospho-Kdr ELISA (Meso Scale Discovery, catalog number N4 IZA-I) to determine tissue phospho-Kdr level. The ELISA procedures are the same per manufacturer's recommendation (see instruction booklet included in the assay kit; the only modification is that 0.2% sodium dodecyl sulfate is added to the lysis buffer). The positive control group is treated with VEGF 96 ug/animal) in saline administered intravenously (as minimum p-KDR inhibition of 0%). The negative control group is treated with saline administered intravenously (as maximum p-KDR inhibition of 100%). Percent inhibition of compound treated groups is calculated relative to the maximum and minimum inhibition groups. TED50 is calculated from a dose response study and is the dose necessary to achieve 50% inhibition at this time point. For example, the compound of Example 1 has an estimated TED5O of 1.34 mg/kg. The TEC90 is calculated from a dose response study and is the concentration necessary to achieve 90% inhibition at this time point. For example, the compound of Example 1 has an estimated TEC90 of 252 nM. This data demonstrates that certain compounds of the present invention are less potent inhibitors of VEGF induced Kdr phosphorylation in vivo compared to certain previously known FGFR inhibitors.
Xenograft tumor models
One of Human bladder cancer cells RTl 12 (European Collection of Cell Cultures), human multiple myeloma cells OPM-2 (German Collection of Microorganisms and Cell Cultures), human non-small cell lung cancer (NSCL) cells NCI-H460 (American Type Culture Collection), human pancreatic cancer cells BxPC- 3 (American Type Culture Collection) or human gastric cancer cells SNU- 16
(American Type Culture Collection) are expanded in culture, according to KOREAN CELL LINE BANK (KCLB) recommendations, harvested and injected subcutaneously onto the rear flank of nude mice. When tumors are established (7-21 days after implant), animals are randomized and grouped into control and test groups. Test compound is prepared in an appropriate vehicle (i.e. formulated in 10% Acacia), and the test compound and a vehicle control are administered by oral gavage. Tumor response is determined by tumor volume measurement performed twice a week during the course of treatment and reported as percent inhibition of tumor volume versus the vehicle control group. The compound of Example 1 demonstrated dose dependent anti-tumor activity in several xenograft tumor models. For example, in the bladder tumor model (RT-112), when dosed at 3 mg/kg (QD for 21 days), 41.3% inhibition was achieved; when dosed at 3 mg/kg (BID for 21 days), 85.9% inhibition was achieved. In the gastric tumor model (SNU- 16), when dosed at 3 mg/kg (QD for 17 days), 62% inhibition was achieved; when dosed at 3 mg/kg (BID for 17 days), 83% inhibition was achieved. In the multiple myeloma tumor model (OPM-2), when dosed at 3 mg/kg (QD for 21 days), 68% inhibition was achieved; when dosed at 3 mg/kg (BID for 21 days), 84% inhibition was achieved. In the NSCLC tumor model (NCI-H460), when dosed at 3 mg/kg (QD for 17 days), 46% inhibition was achieved; when dosed at 3 mg/kg (BID for 17 days), 69% inhibition was achieved. In the pancreatic tumor model (BxPC-3), when dosed at 3 mg/kg (QD for 21 days), 1% inhibition was achieved; when dosed at 3 mg/kg (BID for 21 days), 55% inhibition was achieved. This data demonstrates that certain compounds of the present invention are inhibitors of xenograft human tumor growth in several animal models.
The compounds of the present invention are preferably formulated as pharmaceutical compositions administered by a variety of routes. Most preferably, such compositions are for oral or intravenous administration. Such pharmaceutical compositions and processes for preparing same are well known in the art. See, e.g., REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY (D. Troy, et al, eds., 21st ed., Lippincott Williams & Wilkins, 2005). The compounds of the present invention are generally effective over a wide dosage range. For example, dosages per day normally fall within the range of about 0.5 to about 100 mg/kg of body weight. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, and therefore the above dosage range is not intended to limit the scope of the invention in any way. It will be understood that the amount of the compound actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the actual compound or compounds administered, the age, weight, and response of the individual patient, and the severity of the patient's symptoms.

Claims

We Claim:
1. A compound which is (E)-2-(4-(2-(5-(l-(3,5-dichloropyridin-4-yl)ethoxy)- lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol, or a pharmaceutically acceptable salt thereof.
2. A compound of Claim 1 which is (R)-(E)-2-(4-(2-(5-(l-(3,5- dichloropyridin-4-yl)ethoxy)-lH-indazol-3-yl)vinyl)-lH-pyrazol-l-yl)ethanol, or a pharmaceutically acceptable salt thereof.
3. A pharmaceutical composition comprising a compound or salt of Claim 1 or 2 in combination with a pharmaceutically acceptable carrier, diluent or excipient.
4. A compound or salt of Claim 1 or 2 for use in therapy.
5. A compound or salt of Claim 1 or 2 for the treatment of cancer.
6. The compound of Claim 5 wherein the cancer is non-small cell lung cancer.
7. The compound of Claim 5 wherein the cancer is gastric cancer.
8. The compound of Claim 5 wherein the cancer is multiple myeloma.
PCT/US2010/033487 2009-05-07 2010-05-04 Vinyl indazolyl compounds WO2010129509A1 (en)

Priority Applications (23)

Application Number Priority Date Filing Date Title
MX2011011342A MX2011011342A (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds.
EA201171367A EA018149B1 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
SI201030191T SI2427449T1 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
KR1020117026320A KR101373910B1 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
NZ595553A NZ595553A (en) 2009-05-07 2010-05-04 VINYL INDAZOLYL COMPOUND; (E)-2-(4-(2-(5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-1H-indazol-3-yl)vinyl)-1H-pyrazol-1-yl)ethanol
ES10717402T ES2408117T3 (en) 2009-05-07 2010-05-04 Vinyl Indazolyl Compounds
RS20130207A RS52795B (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
UAA201112804A UA104756C2 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
DK10717402.1T DK2427449T3 (en) 2009-05-07 2010-05-04 VINYLINDAZOLYL COMPOUNDS
CN201080020264.3A CN102421769B (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
AU2010246114A AU2010246114B2 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
SG2011081627A SG175909A1 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
JP2012509889A JP5555314B2 (en) 2009-05-07 2010-05-04 Vinylindazolyl compounds
EP10717402A EP2427449B1 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
BRPI1013748A BRPI1013748A2 (en) 2009-05-07 2010-05-04 "vinyl indazolyl compounds"
MA34312A MA33244B1 (en) 2009-05-07 2010-05-04 VINYLINDAZOLYL COMPOUNDS
MEP-2013-32A ME01462B (en) 2009-05-07 2010-05-04 Vinyl indazol yl compounds
PL10717402T PL2427449T3 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
CA2760535A CA2760535C (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds
TNP2011000545A TN2011000545A1 (en) 2010-02-04 2011-10-25 Vinyl indazolyl compounds
IL216004A IL216004A (en) 2009-05-07 2011-10-27 Vinyl indazolyl compounds and pharmaceutical compositions comprising them
HK12103968.8A HK1163665A1 (en) 2009-05-07 2012-04-20 Vinyl indazolyl compounds
HRP20130323AT HRP20130323T1 (en) 2009-05-07 2013-04-10 Vinyl indazolyl compounds

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US17629009P 2009-05-07 2009-05-07
US61/176,290 2009-05-07
US30141610P 2010-02-04 2010-02-04
US61/301,416 2010-02-04

Publications (1)

Publication Number Publication Date
WO2010129509A1 true WO2010129509A1 (en) 2010-11-11

Family

ID=42238704

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/033487 WO2010129509A1 (en) 2009-05-07 2010-05-04 Vinyl indazolyl compounds

Country Status (35)

Country Link
US (1) US8268869B2 (en)
EP (1) EP2427449B1 (en)
JP (1) JP5555314B2 (en)
KR (1) KR101373910B1 (en)
CN (1) CN102421769B (en)
AR (1) AR078411A1 (en)
AU (1) AU2010246114B2 (en)
BR (1) BRPI1013748A2 (en)
CA (1) CA2760535C (en)
CL (1) CL2011002781A1 (en)
CO (1) CO6450624A2 (en)
CR (1) CR20110580A (en)
DK (1) DK2427449T3 (en)
EA (1) EA018149B1 (en)
EC (1) ECSP11011441A (en)
ES (1) ES2408117T3 (en)
HK (1) HK1163665A1 (en)
HN (1) HN2011002809A (en)
HR (1) HRP20130323T1 (en)
IL (1) IL216004A (en)
JO (1) JO2860B1 (en)
MA (1) MA33244B1 (en)
ME (1) ME01462B (en)
MX (1) MX2011011342A (en)
MY (1) MY160390A (en)
NZ (1) NZ595553A (en)
PE (2) PE20120812A1 (en)
PL (1) PL2427449T3 (en)
PT (1) PT2427449E (en)
RS (1) RS52795B (en)
SG (1) SG175909A1 (en)
SI (1) SI2427449T1 (en)
TW (1) TWI382982B (en)
UA (1) UA104756C2 (en)
WO (1) WO2010129509A1 (en)

Cited By (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012047699A1 (en) * 2010-10-05 2012-04-12 Eli Lilly And Company Crystalline (r) - (e) -2- (4- (2- (5- (1- (3, 5 -dichloropyridin-4 -yl) ethoxy) - 1h - indazol - 3 - yl) vinyl) -1 h- pyrazol- 1 -yl) ethanol and its use as fgfr inhibitor
WO2013133351A1 (en) 2012-03-08 2013-09-12 アステラス製薬株式会社 Novel fgfr3 fusion product
CN103819396A (en) * 2014-02-26 2014-05-28 四川大学 Synthesis method of chiral 1-(3,5-dichloropyridine-4-yl)-ethanol
WO2014151734A1 (en) 2013-03-15 2014-09-25 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214515B2 (en) 2015-08-20 2019-02-26 Zhejiang Hisun Pharmaceutical Co., Ltd. Substituted pyrazoles as inhibitors of fibroblast growth factor receptor
US10544144B2 (en) 2016-06-06 2020-01-28 Shanghai Allist Pharmaceutical And Medical Technol Fused pyrimidine piperidine cyclic derivative, preparation process and use thereof
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
EP3778644A2 (en) 2014-12-23 2021-02-17 The Trustees of Columbia University in the City of New York Fgfr-tacc fusion proteins and methods thereof
WO2021138391A1 (en) * 2019-12-30 2021-07-08 Tyra Biosciences, Inc. Indazole compounds
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
EP4056558A4 (en) * 2019-11-06 2023-12-27 Jinan University Indazole compound, pharmaceutical composition of same, and applications thereof
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11920138B2 (en) 2012-07-24 2024-03-05 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN105906621A (en) * 2015-04-06 2016-08-31 四川百利药业有限责任公司 Ethanol compound used as FGFR inhibitor
WO2016187767A1 (en) * 2015-05-25 2016-12-01 Hutchison Medipharma Limited Pharmaceutical compositions and use thereof
EP3333157B1 (en) * 2015-08-07 2020-12-23 Harbin Zhenbao Pharmaceutical Co., Ltd. Vinyl compounds as fgfr and vegfr inhibitors
WO2022033472A1 (en) * 2020-08-11 2022-02-17 河南迈英诺医药科技有限公司 Fgfr inhibitor compound and use thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002010137A2 (en) * 2000-07-31 2002-02-07 Signal Pharmaceuticals, Inc. Indazole derivatives as jnk inhibitors
EP1510516A1 (en) * 2002-05-31 2005-03-02 Eisai Co., Ltd. Pyrazole compound and medicinal composition containing the same

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK1650203T3 (en) 2000-09-11 2008-06-02 Novartis Vaccines & Diagnostic Quinolinone derivatives as tyrosine kinase inhibitors
EP1447405A4 (en) * 2001-10-17 2005-01-12 Kirin Brewery Quinoline or quinazoline derivatives inhibiting auto-phosphorylation of fibroblast growth factor receptors
DE10342503A1 (en) * 2003-09-12 2005-04-14 Merck Patent Gmbh Benzyl-Benzimidazolylderivate
EP1753763B1 (en) 2004-05-26 2008-08-13 Pfizer Limited Indazole and indolone derivatives and their use as pharmaceuticals
WO2007058626A1 (en) 2005-11-16 2007-05-24 S*Bio Pte Ltd Indazole compounds
JP2009531274A (en) * 2005-12-07 2009-09-03 オーエスアイ・ファーマスーティカルズ・インコーポレーテッド Kinase-inhibiting pyrrolopyridine compounds
DK2120932T3 (en) 2006-12-20 2014-10-13 Nerviano Medical Sciences Srl INDAZOL DERIVATIVES AS KINase INHIBITORS FOR TREATMENT OF CANCER
US7737149B2 (en) * 2006-12-21 2010-06-15 Astrazeneca Ab N-[5-[2-(3,5-dimethoxyphenyl)ethyl]-2H-pyrazol-3-yl]-4-(3,5-dimethylpiperazin-1-yl)benzamide and salts thereof
BRPI0812450A2 (en) 2007-06-05 2019-09-24 Schering Corp polycyclic indazole derivatives and their use as erk inhibitors for cancer treatment
DE102007028521A1 (en) 2007-06-21 2008-12-24 Merck Patent Gmbh Indazolamidderivate
EP2265270A1 (en) * 2008-02-04 2010-12-29 OSI Pharmaceuticals, Inc. 2-aminopyridine kinase inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002010137A2 (en) * 2000-07-31 2002-02-07 Signal Pharmaceuticals, Inc. Indazole derivatives as jnk inhibitors
EP1510516A1 (en) * 2002-05-31 2005-03-02 Eisai Co., Ltd. Pyrazole compound and medicinal composition containing the same

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
S.M. BERGE ET AL.: "Pharmaceutical Salts", JOURNAL OF* PHARMACEUTICAL SCIENCES, vol. 66, no. 1, January 1977 (1977-01-01), XP002675560, DOI: doi:10.1002/jps.2600660104

Cited By (57)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012047699A1 (en) * 2010-10-05 2012-04-12 Eli Lilly And Company Crystalline (r) - (e) -2- (4- (2- (5- (1- (3, 5 -dichloropyridin-4 -yl) ethoxy) - 1h - indazol - 3 - yl) vinyl) -1 h- pyrazol- 1 -yl) ethanol and its use as fgfr inhibitor
AU2011312485B2 (en) * 2010-10-05 2014-09-04 Eli Lilly And Company Crystalline (R) - (E) -2- (4- (2- (5- (1- (3, 5 -dichloropyridin-4 -yl) ethoxy) - 1H - indazol - 3 - yl) vinyl) -1 H- pyrazol- 1 -yl) ethanol and its use as FGFR inhibitor
EA021817B1 (en) * 2010-10-05 2015-09-30 Эли Лилли Энд Компани Crystalline (r)-(e)-2-(4-(2-(5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-1h-indazol-3-yl)vinyl)-1h-pyrazol-1-yl)ethanol monohydrate
JP2013538875A (en) * 2010-10-05 2013-10-17 イーライ リリー アンド カンパニー Crystalline ((R)-(E) -2- (4- (2- (5- (1- (3,5-dichloropyridin-4-yl) ethoxy) -1H-indazol-3-yl) vinyl)- 1H-pyrazol-1-yl) ethanol and use as FGFR inhibitor
US8530665B2 (en) 2010-10-05 2013-09-10 Eli Lilly And Company Crystalline (R)-(E)-2-(4-(2-(5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-1H-indazol-3-yl)vinyl)-1H-pyrazol-1-yl)ethanol
US10213427B2 (en) 2010-12-22 2019-02-26 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US9533954B2 (en) 2010-12-22 2017-01-03 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
US10813930B2 (en) 2010-12-22 2020-10-27 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of FGFR3
WO2013133351A1 (en) 2012-03-08 2013-09-12 アステラス製薬株式会社 Novel fgfr3 fusion product
US11840534B2 (en) 2012-06-13 2023-12-12 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US10131667B2 (en) 2012-06-13 2018-11-20 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US9611267B2 (en) 2012-06-13 2017-04-04 Incyte Holdings Corporation Substituted tricyclic compounds as FGFR inhibitors
US11053246B2 (en) 2012-06-13 2021-07-06 Incyte Corporation Substituted tricyclic compounds as FGFR inhibitors
US11920138B2 (en) 2012-07-24 2024-03-05 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US9388185B2 (en) 2012-08-10 2016-07-12 Incyte Holdings Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9745311B2 (en) 2012-08-10 2017-08-29 Incyte Corporation Substituted pyrrolo[2,3-b]pyrazines as FGFR inhibitors
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
WO2014151734A1 (en) 2013-03-15 2014-09-25 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US11505788B2 (en) 2013-03-15 2022-11-22 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US10208296B2 (en) 2013-03-15 2019-02-19 The Trustees Of Columbia University In The City Of New York Fusion proteins and methods thereof
US10040790B2 (en) 2013-04-19 2018-08-07 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US9533984B2 (en) 2013-04-19 2017-01-03 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US11530214B2 (en) 2013-04-19 2022-12-20 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
US10947230B2 (en) 2013-04-19 2021-03-16 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US10450313B2 (en) 2013-04-19 2019-10-22 Incyte Holdings Corporation Bicyclic heterocycles as FGFR inhibitors
CN103819396A (en) * 2014-02-26 2014-05-28 四川大学 Synthesis method of chiral 1-(3,5-dichloropyridine-4-yl)-ethanol
CN103819396B (en) * 2014-02-26 2016-06-15 四川大学 A kind of 1-(3,5-dichloropyridine-4-base of chirality) synthetic method of-ethanol
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
EP3925979A2 (en) 2014-12-23 2021-12-22 The Trustees of Columbia University in the City of New York Fgfr-tacc fusion proteins and methods thereof
EP3778644A2 (en) 2014-12-23 2021-02-17 The Trustees of Columbia University in the City of New York Fgfr-tacc fusion proteins and methods thereof
US10738048B2 (en) 2015-02-20 2020-08-11 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10016438B2 (en) 2015-02-20 2018-07-10 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10632126B2 (en) 2015-02-20 2020-04-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9708318B2 (en) 2015-02-20 2017-07-18 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10251892B2 (en) 2015-02-20 2019-04-09 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11014923B2 (en) 2015-02-20 2021-05-25 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214528B2 (en) 2015-02-20 2019-02-26 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9801889B2 (en) 2015-02-20 2017-10-31 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11173162B2 (en) 2015-02-20 2021-11-16 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US11667635B2 (en) 2015-02-20 2023-06-06 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US9890156B2 (en) 2015-02-20 2018-02-13 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
US10214515B2 (en) 2015-08-20 2019-02-26 Zhejiang Hisun Pharmaceutical Co., Ltd. Substituted pyrazoles as inhibitors of fibroblast growth factor receptor
US10544144B2 (en) 2016-06-06 2020-01-28 Shanghai Allist Pharmaceutical And Medical Technol Fused pyrimidine piperidine cyclic derivative, preparation process and use thereof
US10611762B2 (en) 2017-05-26 2020-04-07 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11472801B2 (en) 2017-05-26 2022-10-18 Incyte Corporation Crystalline forms of a FGFR inhibitor and processes for preparing the same
US11174257B2 (en) 2018-05-04 2021-11-16 Incyte Corporation Salts of an FGFR inhibitor
US11466004B2 (en) 2018-05-04 2022-10-11 Incyte Corporation Solid forms of an FGFR inhibitor and processes for preparing the same
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
US11591329B2 (en) 2019-07-09 2023-02-28 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11607416B2 (en) 2019-10-14 2023-03-21 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
EP4056558A4 (en) * 2019-11-06 2023-12-27 Jinan University Indazole compound, pharmaceutical composition of same, and applications thereof
US11897891B2 (en) 2019-12-04 2024-02-13 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
US11407750B2 (en) 2019-12-04 2022-08-09 Incyte Corporation Derivatives of an FGFR inhibitor
WO2021138391A1 (en) * 2019-12-30 2021-07-08 Tyra Biosciences, Inc. Indazole compounds
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors

Also Published As

Publication number Publication date
HRP20130323T1 (en) 2013-05-31
MX2011011342A (en) 2011-11-18
IL216004A (en) 2014-01-30
ECSP11011441A (en) 2011-12-30
TWI382982B (en) 2013-01-21
MY160390A (en) 2017-03-15
AU2010246114B2 (en) 2012-12-06
EA201171367A1 (en) 2012-04-30
IL216004A0 (en) 2012-01-31
BRPI1013748A2 (en) 2016-04-05
EP2427449B1 (en) 2013-04-03
CA2760535A1 (en) 2010-11-11
AU2010246114A1 (en) 2011-11-03
KR101373910B1 (en) 2014-03-12
TW201105652A (en) 2011-02-16
JP5555314B2 (en) 2014-07-23
CO6450624A2 (en) 2012-05-31
UA104756C2 (en) 2014-03-11
CN102421769B (en) 2014-04-02
HK1163665A1 (en) 2012-09-14
SI2427449T1 (en) 2013-05-31
ME01462B (en) 2014-04-20
PE20120812A1 (en) 2012-07-08
JO2860B1 (en) 2015-03-15
RS52795B (en) 2013-10-31
CL2011002781A1 (en) 2012-05-25
CA2760535C (en) 2014-01-14
US20100286209A1 (en) 2010-11-11
DK2427449T3 (en) 2013-04-22
AR078411A1 (en) 2011-11-09
NZ595553A (en) 2013-05-31
CR20110580A (en) 2012-01-06
PL2427449T3 (en) 2013-08-30
MA33244B1 (en) 2012-05-02
CN102421769A (en) 2012-04-18
PE20160124A1 (en) 2016-02-24
EA018149B1 (en) 2013-05-30
US8268869B2 (en) 2012-09-18
ES2408117T3 (en) 2013-06-18
SG175909A1 (en) 2011-12-29
HN2011002809A (en) 2013-07-22
PT2427449E (en) 2013-05-06
KR20120004511A (en) 2012-01-12
EP2427449A1 (en) 2012-03-14
JP2012526126A (en) 2012-10-25

Similar Documents

Publication Publication Date Title
US8268869B2 (en) Vinyl indazolyl compounds
US8530665B2 (en) Crystalline (R)-(E)-2-(4-(2-(5-(1-(3,5-dichloropyridin-4-yl)ethoxy)-1H-indazol-3-yl)vinyl)-1H-pyrazol-1-yl)ethanol
JP5868992B2 (en) Substituted benzopyrazine derivatives as FGFR kinase inhibitors for the treatment of cancer diseases
JP6363068B2 (en) Pteridine as an FGFR inhibitor
JP6054406B2 (en) Anti-cancer benzopyrazine via FGFR kinase inhibition
CN113307797B (en) Polysubstituted quinazoline compound and application thereof
CN110903283A (en) Substituted quinazoline compound, pharmaceutical composition containing compound and application of compound
CN115315422B (en) Amide compound and application thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080020264.3

Country of ref document: CN

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

Ref document number: 10717402

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2067/MUMNP/2011

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 595553

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2010717402

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: MX/A/2011/011342

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 216004

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 2760535

Country of ref document: CA

Ref document number: 001877-2011

Country of ref document: PE

ENP Entry into the national phase

Ref document number: 2010246114

Country of ref document: AU

Date of ref document: 20100504

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20117026320

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2012509889

Country of ref document: JP

Ref document number: 11150245

Country of ref document: CO

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: CR2011-000580

Country of ref document: CR

WWE Wipo information: entry into national phase

Ref document number: 201171367

Country of ref document: EA

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: PI1013748

Country of ref document: BR

WWE Wipo information: entry into national phase

Ref document number: P-2013/0207

Country of ref document: RS

WWE Wipo information: entry into national phase

Ref document number: 000013-2016

Country of ref document: PE

ENP Entry into the national phase

Ref document number: PI1013748

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20111104