WO2004009083A1 - Antitumor compound and therapeutic uses thereof - Google Patents

Antitumor compound and therapeutic uses thereof Download PDF

Info

Publication number
WO2004009083A1
WO2004009083A1 PCT/EP2003/007963 EP0307963W WO2004009083A1 WO 2004009083 A1 WO2004009083 A1 WO 2004009083A1 EP 0307963 W EP0307963 W EP 0307963W WO 2004009083 A1 WO2004009083 A1 WO 2004009083A1
Authority
WO
WIPO (PCT)
Prior art keywords
use according
tumors
ret
treatment
kit
Prior art date
Application number
PCT/EP2003/007963
Other languages
French (fr)
Other versions
WO2004009083A8 (en
Inventor
Cinzia Lanzi
Giuliana Cassinelli
Giuditta Cuccuru
Marco Alessandro Pierotti
Franco Zunino
Ernesto Menta
Original Assignee
Cell Therapeutics Europe S.R.L.
Istituto Nazionale Per Lo Studio E La Cura Dei Tumori
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 Cell Therapeutics Europe S.R.L., Istituto Nazionale Per Lo Studio E La Cura Dei Tumori filed Critical Cell Therapeutics Europe S.R.L.
Priority to EP03765081A priority Critical patent/EP1534271B1/en
Priority to CA002493202A priority patent/CA2493202A1/en
Priority to MXPA05000781A priority patent/MXPA05000781A/en
Priority to US10/522,081 priority patent/US20060258731A1/en
Priority to JP2004522542A priority patent/JP2005537277A/en
Priority to DE60307856T priority patent/DE60307856T2/en
Priority to AU2003251437A priority patent/AU2003251437A1/en
Publication of WO2004009083A1 publication Critical patent/WO2004009083A1/en
Publication of WO2004009083A8 publication Critical patent/WO2004009083A8/en
Priority to US12/313,033 priority patent/US20090130229A1/en

Links

Classifications

    • 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
    • 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
    • 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

Definitions

  • the present invention regards the use of the compound (E)-l,3-dihydro- 5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one in the treatment of tumors involving Met, PDGF-R, FGF-RI, FGF-R3 and Kit tyrosine kinases, or Ret oncoproteins.
  • Met Met, PDGF-R, FGF-RI, FGF-R3 and Kit tyrosine kinases, or Ret oncoproteins.
  • RET PTC oncogenes are involved in the transforming processes of human papillary thyroid tumors and originate from the rearrangement of the tyrosine kinase domain of proto-RET with different donor genes.
  • the products of such gene rearrangements show ligand-independent tyrosine-kinase activity and are localized in the cytoplasm.
  • Ret/ptcl is the product of the RET/PTCl oncogene, which originates from the rearrangement of the proto-RET tyrosine kinase domain with the H4/D10S170 gene.
  • Cpd 1 l,3-dihydro-5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one derivative
  • Cpd 1 has now been found to effectively inhibit tyrosine kinases, other than Ret/ptcl, that play a central role in tumor onset, progression and spreading to distant organs. Specifically, Cpd 1 has been shown to completely inhibit the autophosphorylation of Ret/MEN2A (mutations C634R and C634 ), Ret/MEN2B (mutM918T), Met, PDGF-R, FGF-RI, FGF-R3 and Kit (c-Kit and mut ⁇ 559) tyrosine kinases, to reduce their expression (down- regulation) and to revert the phenotype of cells thereby transformed.
  • Object of the invention is therefore the use of the compound (E)-l,3- dihydro-5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one, or of its salts with pharmaceutically acceptable bases, for the treatment of tumors involving at least one of Met, PDGF-R, FGF-RI, FGF-R3 and Kit tyrosine kinases, or involving at least one oncoprotein of the Ret family, including Ret receptors carrying MEN2-associated mutations, in the initial stages of cell transformation or in the following stages of tumor proliferation and dissemination.
  • the invention also concerns the use of any stereoisomer or tautomeric form of Cpd.1.
  • MTC medullary thyroid carcinomas
  • MEN2- associated diseases including MTC, pheochromocytoma, parathyroid hyperplasia, and enteric ganglioneuromas.
  • Met inhibition is useful to antagonize the invasive/metastatic phenotype of tumors of epithelial origin. With respect to Met- activating alterations, Cpd 1 may also have a specific indication in the therapy of kidney tumors. Kit inhibition is useful in the treatment of gastrointestinal stromal tumors, small cell lung carcinomas, seminomas and hematological malignancies such as mastocytosis and acute myelogenous leukemia.
  • Cpd 1 can be used for the treatment of melanomas and gliomas expressing high levels of FGF-RI and of the respective bFGF ligand eventually involved in autocrine loops. Since this receptor has an important role in angiogenic processes, its inhibition by means of Cpd 1 is useful for the control of tumor vascularization.
  • FGF-R3 inhibition is useful in the treatment of multiple myeloma, bladder and cervix carcinomas.
  • RPI-1 and its salts can be formulated with pharmaceutically acceptable vehicles and excipients.
  • the phenol function of Cpd 1 can be salif ⁇ ed by treatment with suitable organic or inorganic bases.
  • the pharmaceutical compositions can be administered by the oral, parenteral, sublingual or transdermic routes, preferably in the form of tablets, capsules, granules, powders, syrups, solutions, suspensions, suppositories, controlled release forms.
  • the compositions can be prepared with conventional techniques, using ingredients known in the art.
  • the quantity of active principle can be varied depending on the severity of the disease, age of the patient, type and route of administration, but in general an amount of 0.1 to 1000 mg/Kg, preferably from 5 to 300 mg/Kg, more preferably from 20 to 200 mg/Kg, in single or multiple doses one or more times a day, is used.
  • Ret oncoproteins carrying aminoacid substitutions which cause constitutive tyrosine kinase activity are involved in sporadic MTC and in the inherited type 2 Multiple Endocrine Neoplasia syndromes (MEN2A, MEN2B and Familial MTC), all characterized by the occurrence of MTC (Jhiang S.M. et al. Oncogene 19, 5590, 2000).
  • MEN2A, MEN2B and Familial MTC all characterized by the occurrence of MTC (Jhiang S.M. et al. Oncogene 19, 5590, 2000).
  • MEN2A sporadic MTC RET mutations are somatic
  • MEN2 patients RET mutations are present at the germline level. These mutations cause constitutive activation of the receptor without modifying its localization at the cell membrane.
  • Ret oncoproteins used in this study involve Cys634 (indicated as Ret/MEN2A C634R and Ret/MEN2A C634W ) or Met918 (indicated as Ret/MEN2B M918T ), and represent the most frequently expressed Ret oncoproteins in MEN2A and MEN2B, respectively.
  • Cpd 1 The inhibitory effect of Cpd 1 has been demonstrated in murine cells transfected with the RET/MEN2A(C634R) gene ( N iH3T3 MEN2A(c634R) cells) and in the human medullary thyroid carcinoma cell lines TT and MZ- CRC-1, respectively characterized by the expression of Ret/MEN2A(C634W) and Ret/MEN2B(M918T) ( Figures 1, 2). A reduction of the oncoprotein tyrosine-phosphorylation and expression is observed in these cell lines ( Figures 1C and 2A). The inhibition of Ret/MEN2A and Ret/MEN2B receptor autophosphorylation by Cpd 1 is associated with an antiproliferative effect ( Figures IB and 2B).
  • N ⁇ H 3T3 MEN2A(C634R) transfectants reverted their transformed phenotype following exposure to Cpdl (Fig. 1A).
  • a significant dose-dependent antitumor activity has been observed in nude mice xenografted with the TT tumor: after oral administration of a daily dose of 50- 100 mg/Kg (twice a day), Cpd 1 treatment reached 80% tumor weight inhibition (TWI) without inducing toxicity (Fig. 3).
  • TWI tumor weight inhibition
  • Fig. 3 The demonstrated pharmacological and biochemical efficacy of Cpd 1 in controlling the proliferation of MTC cells is particularly important considering the aggressiveness of such tumors and the inefficacy of conventional therapies.
  • Met the hepatocyte growth factor receptor
  • PDGF-R receptor tyrosine kinases
  • FGF-R3 receptor tyrosine kinase such as chromosomal traslocations or point mutations produce deregulated, constitutively active, FGF-R3 receptors which have been involved in multiple mieloma and in bladder and cervix carcinomas (Powers CJ. et al. Endocr. Rel. Cancer 7, 165, 2000).
  • Cpd 1 The ability of Cpd 1 to down regulate both tyrosine autophosphorylation and expression of FGF-R3 (mutY373C) exogenously expressed in NIH3T3 transfectants is illustrated in Fig. 6B.
  • Kit tyrosine kinase is constitutively activated as a consequence of mutations or of its involvement in autocrine loops in different tumors such as small cell lung cancers, gastro-intestinal stromal tumors, seminomas and leukemias (Heinrich M.C. et al. J. Clin. Oncol. 20, 1692, 2002).
  • Cpd 1 inhibits the constitutive autophosphorylation and expression of the Kit ( ⁇ 559) mutant exogenously expressed in NIH3T3 cells (Fig. 7B).
  • Such inhibition is associated with reversion of the transformed morphologic phenotype of the transfected cells (Fig. 7A).
  • Fig. 7A Fig.
  • tumor is intended to encompass but is not limited to the abnormal cell proliferation of malignant or non-malignant cells of various tissues and/or organs such as muscle, bone or connective tissue, the skin, brain, lungs, sex organs, the lymphatic or renal systems, mammary or blood cells, liver, the digestive system, pancreas and thyroid or adrenal glands.
  • the abnormal cell proliferation can include but is not limited to tumors of the ovary, breast, brain, prostate, colon, liver, lung, ovary, uterus, cervix, pancreas, gastrointestinal tract, head, neck, nasopharynx, skin, bladder, stomach, kidney or testicles, Kaposi's sarcoma, cholangiocarcinoma, choriocarcinoma, neuroblastoma, Wilms' tumor, Hodgkin's disease, melanoma, multiple myeloma, chronic lymphocytic leukemia and acute or chronic granulocytic lymphoma.
  • the compounds according to the present invention can be administered alone or in combination with other anti-tumor or anti-cancer agents including but not limited to: adriamycin, daunomycin, methotrexate, vincristin, 6- mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L_PAM, BCNU and VP-16.
  • the compounds according to the present invention can also be included in a kit for the treatment of tumors.
  • the kit can include additional anti-cancer or anti- tumor agents.
  • FIG. 1 Effects of Cpd 1 on NLH3T3 MEN2A(C634R) transfectants expressing exogenous RET/MEN2A(C634R).
  • B Antiproliferative effect. N IH3T3 MEN2A(C634R) cells and the parental NIH3T3 cells were treated with increasing concentrations of the drug for 72h and then counted with a Coulter Counter.
  • FIG. 1 Effects of Cpdl on human MTC cell lines harboring MEN2- associated RET mutants.
  • Whole cell extracts were processed for Western blotting and probed with anti-pTyr and anti-Ret antibodies. As evidenced in anti-pTyr blots, cell treatment with the compound induced a dose-dependent inhibition of tyrosine phosphorylation of Ret receptors in both cell lines.
  • a reduced expression of receptor concentrations was observed in cells treated with the highest concentrations of the drug.
  • FIG. 3 Antitumor activity of Cpd 1 in nude mice harboring TT medullary thyroid carcinoma xenografts.
  • Drug treatment started 25 days after s.c. inoculum of the tumor cells.
  • Cpd 1 was delivered per os at 50 or 100 mg/Kg, twice in a day (2qd), for 10 consecutive days (indicated by arrows).
  • Control mice received the vehicle.
  • the treatment induced a significant dose- dependent inhibition of tumor growth.
  • TWI were 60% (P ⁇ 0.005) and 80% (P ⁇ 0.0005) for the 50 mg/Kg and 100 mg/Kg doses, respectively.
  • FIG. 4 Inhibition of Met autophosphorylation and expression in human papillary thyroid carcinoma cells (TPC-1) treated with Cpdl.
  • TPC- 1 cells were exposed to solvent (-) or the indicated concentrations of Cpd 1 for 72h. Equal amounts of protein were used for immunoprecipitation (IP) with anti-Met antibody or for the preparation of whole cell lysates (WCL). Immunoprecipitated proteins and WCLs were separated by SDS-PAGE, transferred to nitrocellulose membranes, and subjected to Western blotting with anti-pTyr or anti-Met antibodies.
  • FIG. 5 Inhibition of PDGF-R autophosphorylation and expression in whole cells by Cpd 1.
  • Figure 7 Effects of Cpdl on cell lines expressing constitutively activated forms of Kit.
  • A) Reversion of the transformed morphologic phenotype of NIH3T3 transfectants expressing exogenous mutated KIT ( ⁇ 559). Cells were treated with 20 ⁇ M Cpdl and photographed 24h later under a phase-contrast microscope (original magnification X 100).
  • the following human medullary thyroid carcinoma (MTC) cell lines were used in this study.
  • the TT cell line derived from a MEN2A-associated MTC characterized by expression of the RET oncogene carrying the mutation C634W.
  • the MZ-CRC-1 cell line derived from MEN2B-associated MTC characterized by expression of the RET oncogene carrying the mutation M918T.
  • TT cells were cultivated in Ham's F12 medium (BioWhittaker, Verviers, Belgium) supplemented with 15% fetal calf serum (FCS) (Life Technologies, Inc., Gaithersburg, MD) whereas MZ-CRC-1 cells were grown in Dulbecco's modified Eagle's medium (DMEM) (BioWhittaker) supplemented with 10% FCS.
  • FCS fetal calf serum
  • DMEM Dulbecco's modified Eagle's medium
  • the human papillary thyroid carcinoma cell line TPC-1 which was used as a model of Met overexpression, was routinely cultivated in DMEM with 10% FCS.
  • N592 cells derived from a human SCLC, were characterized by Kit activation through autocrine stimulation by the SCF ligand. N592 cells were cultured in RPMI 1640 supplemented with 10% FCS.
  • the mouse SWISS3T3 and NIH3T3 fibroblasts were cultured in DMEM with 10% calf serum (Colorado Serum Company, Denver, CO).
  • NIH3T3 cells transfected with different oncogenes were used.
  • NIH3T3 MEN2A transfectants express the short isoform of the RET-MEN2A (C634R) oncogene;
  • NIH3T3 KITA559 cells express KIT carrying the activating mutation ⁇ 559 found in GISTs;
  • NIH 3T3 FGFR3(Y373C) express the FGF-R3 gene carrying the activating mutation Y373C found in a human multiple myeloma cell line.
  • 2N5A cells are NIH3T3 cells transformed by the COL1A1/PDGFB rearrangement generating autocrine stimulation of PDGF-R. All NIH3T3 transfectants were maintained in DMEM plus 5% calf serum in a 10% C0 2 atmosphere. Antiproliferative assays
  • NIH3T3 and NIH3T3 MEN2A cells were trypsinized after 3 days (NIH3T3 and NIH3T3 MEN2A cells) or 7 days (MTC cells) of treatment with Cpdl and counted by a Coulter Counter (Coulter Electronics, Luton U.K.). The concentrations able to inhibit cell proliferation by 50% (IC 50 ) were calculated from the dose-response curves. Each experiment was performed in duplicate Antibodies The following polyclonal antibodies were used: anti-Ret recognizing a
  • COOH-terminal sequence (aa 1000-1014) common to the two Ret isoforms (Borrello M. G., et al., Mol. Cell Biol. 16, 2151, 1996); anti-cKit, anti-Met and anti-FGF-R3 from Santa Cruz Biotechnology (Santa Cruz, CA); anti-actin from Sigma (St. Louis, MO); anti-PDGF-R ⁇ / ⁇ from Upstate Biotechnology (Lake Placid, NY); anti phospho-cKit (Tyr 719) from Cell Signaling Technology (Beverly, MA).
  • the mouse monoclonal anti-phosphotyrosine (anti-pTyr) 4G10 and anti-FGF-Rl antibodies were from Upstate Biotechnology.
  • Protein concentration was determined in appropriately diluted aliquots by the bicinchionic acid (BCA) method (Pierce, Rockford, IL), then samples were adjusted to a final concentration of 10%» glycerol, 5% ⁇ -mercaptoethanol, 0.001% bromophenol blue.
  • BCA bicinchionic acid
  • cells were treated with Cpdl or solvent for the indicated times.
  • Cell monolayers were rinsed twice with cold phosphate-buffered saline plus 0.1 mM sodium orthovanadate and then left for 20 min on ice in lysis buffer (50mM HEPES pH7.6, 150mM NaCl, 10 % glycerol, 1% Triton X-100, 1.5 mM MgCl 2 , 1 mM EGTA, 100 mM NaF, 10 mM sodium pyrophosphate, 10 ⁇ g/ml antipain, 20 ⁇ g/ml chymostatin, 10 ⁇ g/ml E64, 1 mg/ml pefabloc SC).
  • lysis buffer 50mM HEPES pH7.6, 150mM NaCl, 10 % glycerol, 1% Triton X-100, 1.5 mM MgCl 2 , 1 mM EGTA, 100 mM NaF, 10 mM sodium pyr
  • Cells were then collected, aspirated through a 22-gauge needle and centrifuged at lOOOOg, for 20 min, at 4 °C. Protein concentration was determined by the BCA reagent (Pierce). Cell extracts were incubated with protein A-agarose and the indicated antibody for 2 hr at 4°C under rotation. After washing 3 times with 20mM HEPES pH 7.6, 150 mM NaCl, 10% glycerol, 0.1% Triton X-100, the immunoprecipitates were eluted with complete sample buffer.
  • the E configuration of the esocyclic double bond in position 2 was determined by means of ID NOE NMR experiments.

Landscapes

  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Cardiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Indole Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)

Abstract

The use of an arylidene 2-indolinone derivative for treating tumors involving Met, PDGF-R, FGF-RI, FGF-R3 and Kit tyrosine kinases, or the oncoproteins of Ret family is disclosed. The invention also provides pharmaceutical compositions comprising laid arylidene 2-indolinone derivative as well as a kit comprising, in separate containers, said derivative and an anti-cancer or anti-tumor agent.

Description

ANTITUMOR COMPOUND AND THERAPEUTIC USES THEREOF
The present invention regards the use of the compound (E)-l,3-dihydro- 5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one in the treatment of tumors involving Met, PDGF-R, FGF-RI, FGF-R3 and Kit tyrosine kinases, or Ret oncoproteins. BACKGROUND ART
RET PTC oncogenes are involved in the transforming processes of human papillary thyroid tumors and originate from the rearrangement of the tyrosine kinase domain of proto-RET with different donor genes. The products of such gene rearrangements show ligand-independent tyrosine-kinase activity and are localized in the cytoplasm. Ret/ptcl is the product of the RET/PTCl oncogene, which originates from the rearrangement of the proto-RET tyrosine kinase domain with the H4/D10S170 gene.
Int. J. Cancer 85, 384-390 (2000) reports the tyrosine kinase activity inhibition of Ret/ptcl oncoprotein by arylidene 2-indolinone compounds. The l,3-dihydro-5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one derivative (hereafter "Cpd 1") is indicated as particularly effective in reverting the morphologic phenotype of NIH3T3 cells transformed with the RET/PTCl oncogene. In the same paper, tyrosine kinase inhibition by arylidene 2- indolinorie compounds is proposed for the study of Ret signaling and for controlling cell proliferation in Ret- and Ret/ptcs-associated diseases. DESCRIPTION OF THE INVENTION
Cpd 1 has now been found to effectively inhibit tyrosine kinases, other than Ret/ptcl, that play a central role in tumor onset, progression and spreading to distant organs. Specifically, Cpd 1 has been shown to completely inhibit the autophosphorylation of Ret/MEN2A (mutations C634R and C634 ), Ret/MEN2B (mutM918T), Met, PDGF-R, FGF-RI, FGF-R3 and Kit (c-Kit and mutΔ559) tyrosine kinases, to reduce their expression (down- regulation) and to revert the phenotype of cells thereby transformed.
Object of the invention is therefore the use of the compound (E)-l,3- dihydro-5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one, or of its salts with pharmaceutically acceptable bases, for the treatment of tumors involving at least one of Met, PDGF-R, FGF-RI, FGF-R3 and Kit tyrosine kinases, or involving at least one oncoprotein of the Ret family, including Ret receptors carrying MEN2-associated mutations, in the initial stages of cell transformation or in the following stages of tumor proliferation and dissemination.
The invention also concerns the use of any stereoisomer or tautomeric form of Cpd.1.
Inhibition of deregulated, constitutively active, Ret receptors is useful in the treatment of sporadic medullary thyroid carcinomas (MTC) and MEN2- associated diseases including MTC, pheochromocytoma, parathyroid hyperplasia, and enteric ganglioneuromas.
Met inhibition is useful to antagonize the invasive/metastatic phenotype of tumors of epithelial origin. With respect to Met- activating alterations, Cpd 1 may also have a specific indication in the therapy of kidney tumors. Kit inhibition is useful in the treatment of gastrointestinal stromal tumors, small cell lung carcinomas, seminomas and hematological malignancies such as mastocytosis and acute myelogenous leukemia.
The uncontrolled activation of PDGF-R and its involvement in autocrine loops support the therapeutic use of Cpd 1 in tumors unresponsive to conventional therapies, such as glioma and dermatofibrosarcoma protuberans. In addition, PDGF-R is involved in tumor angiogenesis and vascular development thus supporting the use of Cpdl for the control of neoangiogenesis in solid tumors. Cpd 1 can be used for the treatment of melanomas and gliomas expressing high levels of FGF-RI and of the respective bFGF ligand eventually involved in autocrine loops. Since this receptor has an important role in angiogenic processes, its inhibition by means of Cpd 1 is useful for the control of tumor vascularization.
FGF-R3 inhibition is useful in the treatment of multiple myeloma, bladder and cervix carcinomas.
For use in therapy, RPI-1 and its salts can be formulated with pharmaceutically acceptable vehicles and excipients. The phenol function of Cpd 1 can be salifϊed by treatment with suitable organic or inorganic bases. The pharmaceutical compositions can be administered by the oral, parenteral, sublingual or transdermic routes, preferably in the form of tablets, capsules, granules, powders, syrups, solutions, suspensions, suppositories, controlled release forms. The compositions can be prepared with conventional techniques, using ingredients known in the art. The quantity of active principle can be varied depending on the severity of the disease, age of the patient, type and route of administration, but in general an amount of 0.1 to 1000 mg/Kg, preferably from 5 to 300 mg/Kg, more preferably from 20 to 200 mg/Kg, in single or multiple doses one or more times a day, is used.
DETAILED DESCRIPTION OF THE INVENTION
Ret oncoproteins carrying aminoacid substitutions which cause constitutive tyrosine kinase activity are involved in sporadic MTC and in the inherited type 2 Multiple Endocrine Neoplasia syndromes (MEN2A, MEN2B and Familial MTC), all characterized by the occurrence of MTC (Jhiang S.M. et al. Oncogene 19, 5590, 2000). Whereas in sporadic MTC RET mutations are somatic, in MEN2 patients RET mutations are present at the germline level. These mutations cause constitutive activation of the receptor without modifying its localization at the cell membrane. The Ret oncoproteins used in this study involve Cys634 (indicated as Ret/MEN2AC634R and Ret/MEN2AC634W) or Met918 (indicated as Ret/MEN2BM918T), and represent the most frequently expressed Ret oncoproteins in MEN2A and MEN2B, respectively. The inhibitory effect of Cpd 1 has been demonstrated in murine cells transfected with the RET/MEN2A(C634R) gene (NiH3T3MEN2A(c634R) cells) and in the human medullary thyroid carcinoma cell lines TT and MZ- CRC-1, respectively characterized by the expression of Ret/MEN2A(C634W) and Ret/MEN2B(M918T) (Figures 1, 2). A reduction of the oncoprotein tyrosine-phosphorylation and expression is observed in these cell lines (Figures 1C and 2A). The inhibition of Ret/MEN2A and Ret/MEN2B receptor autophosphorylation by Cpd 1 is associated with an antiproliferative effect (Figures IB and 2B). NτH3T3MEN2A(C634R) transfectants reverted their transformed phenotype following exposure to Cpdl (Fig. 1A). A significant dose-dependent antitumor activity has been observed in nude mice xenografted with the TT tumor: after oral administration of a daily dose of 50- 100 mg/Kg (twice a day), Cpd 1 treatment reached 80% tumor weight inhibition (TWI) without inducing toxicity (Fig. 3). The demonstrated pharmacological and biochemical efficacy of Cpd 1 in controlling the proliferation of MTC cells is particularly important considering the aggressiveness of such tumors and the inefficacy of conventional therapies.
Met, the hepatocyte growth factor receptor, is a protein tyrosine kinase involved in the invasive process characteristic of tumor progression and metastatic growth (Maulik G. et al. Cytok. Growth Factor Rev. 13, 41, 2000). Alterations such as mutations, overexpression or the involvement in autocrine loops are the cause of uncontrolled constitutive activation of the kinase. The uncontrolled kinase activity of Met is involved in the invasiveness of many tumors of epithelial origin. In papillary thyroid carcinomas Met is frequently overexpressed. The results illustrated in Fig. 4 show a dose-dependent inhibition of Met autophosphorylation in the papillary thyroid carcinoma cell line TPC- 1 treated with Cpd 1. Met protein levels are also reduced in treated cells. Other receptor tyrosine kinases, such as PDGF-R (Rosenkranz S. and
Kazlauskas A. Growth Factors 16, 201, 1999) and FGF-RI (Powers CJ. et al. Endocr. Rel. Cancer 7, 165, 2000), which are involved either in autocrine loops or in neoangiogenic processes, have an important role in cancer growth. A deregulated activation of these receptors is observed in tumors unresponsive to conventional therapies, such as gliomas and melanomas. The results illustrated in Figures. 5 and 6 show a dose-dependent inhibition by Cpd 1 of receptor autophosphorylation induced by autocrine stimulation (Fig. 5A) or by exogenous ligand (Figures 5B and 6A). These effects are associated with a reduced receptor expression. Concentrations of Cpd 1 higher than 15 μM cause full inhibition of PDGF-R phosphorylation.
Activating mutations of the FGF-R3 receptor tyrosine kinase such as chromosomal traslocations or point mutations produce deregulated, constitutively active, FGF-R3 receptors which have been involved in multiple mieloma and in bladder and cervix carcinomas (Powers CJ. et al. Endocr. Rel. Cancer 7, 165, 2000). The ability of Cpd 1 to down regulate both tyrosine autophosphorylation and expression of FGF-R3 (mutY373C) exogenously expressed in NIH3T3 transfectants is illustrated in Fig. 6B.
Kit tyrosine kinase is constitutively activated as a consequence of mutations or of its involvement in autocrine loops in different tumors such as small cell lung cancers, gastro-intestinal stromal tumors, seminomas and leukemias (Heinrich M.C. et al. J. Clin. Oncol. 20, 1692, 2002). As shown in Fig. 7, Cpd 1 inhibits the constitutive autophosphorylation and expression of the Kit (Δ559) mutant exogenously expressed in NIH3T3 cells (Fig. 7B). Such inhibition is associated with reversion of the transformed morphologic phenotype of the transfected cells (Fig. 7A). In addition, Fig. 7C shows the dose-dependent inhibition by Cpd 1 of c-Kit activated through autocrine loop in the small cell lung carcinoma cell line N592. As used herein, the term "tumor" is intended to encompass but is not limited to the abnormal cell proliferation of malignant or non-malignant cells of various tissues and/or organs such as muscle, bone or connective tissue, the skin, brain, lungs, sex organs, the lymphatic or renal systems, mammary or blood cells, liver, the digestive system, pancreas and thyroid or adrenal glands. The abnormal cell proliferation can include but is not limited to tumors of the ovary, breast, brain, prostate, colon, liver, lung, ovary, uterus, cervix, pancreas, gastrointestinal tract, head, neck, nasopharynx, skin, bladder, stomach, kidney or testicles, Kaposi's sarcoma, cholangiocarcinoma, choriocarcinoma, neuroblastoma, Wilms' tumor, Hodgkin's disease, melanoma, multiple myeloma, chronic lymphocytic leukemia and acute or chronic granulocytic lymphoma.
The compounds according to the present invention can be administered alone or in combination with other anti-tumor or anti-cancer agents including but not limited to: adriamycin, daunomycin, methotrexate, vincristin, 6- mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L_PAM, BCNU and VP-16. The compounds according to the present invention can also be included in a kit for the treatment of tumors. The kit can include additional anti-cancer or anti- tumor agents.
The following Figures illustrate the invention in greater detail. Description of the Figures
Figure 1. Effects of Cpd 1 on NLH3T3MEN2A(C634R) transfectants expressing exogenous RET/MEN2A(C634R). A) Reversion of the transformed morphologic phenotype of NIH3T3MEN2A(C634R) cells. Cells were exposed to 6 μM Cpdl for 24h and then photographed under a phase-contrast microscope (original magnification X 100). B) Antiproliferative effect. NIH3T3MEN2A(C634R) cells and the parental NIH3T3 cells were treated with increasing concentrations of the drug for 72h and then counted with a Coulter Counter. Dose-response curves, from which the reported IC50 values were calculated, showed the higher sensitivity to the drug of the Ret oncoprotein- positive cell line. C) Inhibition of Ret/MEN2A(C634R) autophosphorylation and expression. Cells were treated with solvent (-) or 10 μM Cpdl (+), for the indicated times. Whole cell lysates were prepared and subjected to SDS-PAGE and Western blotting with anti-pTyr antibody. After stripping, the filter was reblotted with anti-Ret antibody. Arrows indicate the partial and fully glycosilated forms of the Ret/MEN2A receptor. Following 2h of exposure to the drug, the receptor appeared partially dephosphorylated. At longer times, a complete tyrosine dephosphorylation was associated with reduced expression of the receptor
Figure 2. Effects of Cpdl on human MTC cell lines harboring MEN2- associated RET mutants. A) Inhibition of Ret autophosphorylation. TT and MZ-CRC-1 cells, expressing respectively RET/MEN2A(C634W) and RET/MEN2B(M918T), were treated with the indicated concentrations of Cpdl, for 24 h. Control cells (C) received the solvent. Whole cell extracts were processed for Western blotting and probed with anti-pTyr and anti-Ret antibodies. As evidenced in anti-pTyr blots, cell treatment with the compound induced a dose-dependent inhibition of tyrosine phosphorylation of Ret receptors in both cell lines. A reduced expression of receptor concentrations was observed in cells treated with the highest concentrations of the drug. B) Antiproliferative effect. TT and MZ-CRC-1 cells were treated with increasing concentrations of Cpd 1 for 7 days and then counted with a Coulter Counter. Dose-response curves documented the ability of the drug to interfere with the proliferation potential of the two MTC cell lines.
Figure 3. Antitumor activity of Cpd 1 in nude mice harboring TT medullary thyroid carcinoma xenografts. Drug treatment started 25 days after s.c. inoculum of the tumor cells. Cpd 1 was delivered per os at 50 or 100 mg/Kg, twice in a day (2qd), for 10 consecutive days (indicated by arrows). Control mice received the vehicle. The treatment induced a significant dose- dependent inhibition of tumor growth. TWI were 60% (P < 0.005) and 80% (P< 0.0005) for the 50 mg/Kg and 100 mg/Kg doses, respectively.
Figure 4. Inhibition of Met autophosphorylation and expression in human papillary thyroid carcinoma cells (TPC-1) treated with Cpdl. A): TPC- 1 cells were exposed to solvent (-) or the indicated concentrations of Cpd 1 for 72h. Equal amounts of protein were used for immunoprecipitation (IP) with anti-Met antibody or for the preparation of whole cell lysates (WCL). Immunoprecipitated proteins and WCLs were separated by SDS-PAGE, transferred to nitrocellulose membranes, and subjected to Western blotting with anti-pTyr or anti-Met antibodies. B): Cells were treated as in A. Cell extracts were immunoprecipitated with anti-pTyr antibody and probed with anti-Met antibody. C): Cells were serum-starved for 24h and exposed to solvent or Cpdl (60 μM) during the last 18h. Then they were left untreated (-) or stimulated with 20ng/ml HGF (+), for 10 min. Cell lysates were immunoprecipitated with anti-Met and probed with anti-pTyr or anti-Met antibody. Drug treatment abolished the constitutive or HGF-induced Met tyrosine phosphorylation and induced a reduction of Met expression.
Figure 5. Inhibition of PDGF-R autophosphorylation and expression in whole cells by Cpd 1. A): 2N5A cells (NIH3T3 transformed by the COL1A1/PDGFB rearrangement generating autocrine stimulation of PDGF-R) were treated with Cpd 1 at the indicated concentrations, for 72 h. Cell extracts were immunoprecipitated with anti-PDGF-R and subjected to Western blotting with anti-pTyr or anti-PDGF-R antibodies. B): Swiss 3T3 cells were serum starved for 24 h and then treated with Cpdl at the indicated concentrations, for 18 h. After stimulation with 1 nM PDGF for 5 min, whole cell lysates were prepared and subjected to Western blotting with anti-pTyr or anti-PDGF-R. Protein loading by anti-actin blot is shown. In both cell systems, a dose- dependent drug-induced inhibition of receptor tyrosine phosphorylation and expression was documented. Figure 6. Inhibition of autophosphorylation and expression of FGF-RI and FGF-R3 receptors in whole cells by Cpdl. A): Swiss3T3 cells were serum starved for 24 h and then treated with Cpdl at the indicated concentrations, for 18 h. After stimulation with 100 ng/ml FGF for 5 min, whole cell lysates were prepared and subjected to Western blotting with anti-pTyr or anti-FGF-Rl. Protein loading by anti-actin blot is shown. B): NIH3T3 transformed by the FGF-R3 mutant Y373C were treated with the indicated concentrations, for 72 h. Cell extracts were immunoprecipitated with anti-FGF-R3 and then subjected to Western blotting with anti p-Tyr or anti-FGF-R3. In both cell systems, a dose-dependent inhibition by Cpd 1 of the receptor tyrosine phosphorylation and expression was documented.
Figure 7. Effects of Cpdl on cell lines expressing constitutively activated forms of Kit. A): Reversion of the transformed morphologic phenotype of NIH3T3 transfectants expressing exogenous mutated KIT (Δ559). Cells were treated with 20 μM Cpdl and photographed 24h later under a phase-contrast microscope (original magnification X 100). B): Inhibition of Kit (Δ559) autophosphorylation and expression in NIH3T3 transfectants. Cells were treated with Cpd 1 at the indicated concentrations, for 72h. Cell lysates were immunoprecipitated with anti-Kit antibody and subjected to Western blotting with anti-p-Tyr or anti-Kit antibodies. C): Inhibition of expression and autocrine loop-activated autophosphorylation of c-Kit in the SCLC cell line N592. Cells were treated with Cpd 1 at the indicated concentrations, for 24h. Whole cell lysates were subjected to Western blotting with anti-Kit antibody or with an antibody specifically recognizing the tyrosine phosphorylated/activated Kit (p-Kit). In both cell systems, a dose-dependent drug-induced inhibition of receptor tyrosine phosphorylation and expression was documented. MATERIALS AND METHODS Cell lines and culture conditions
The following human medullary thyroid carcinoma (MTC) cell lines were used in this study. The TT cell line derived from a MEN2A-associated MTC characterized by expression of the RET oncogene carrying the mutation C634W. The MZ-CRC-1 cell line derived from MEN2B-associated MTC characterized by expression of the RET oncogene carrying the mutation M918T. TT cells were cultivated in Ham's F12 medium (BioWhittaker, Verviers, Belgium) supplemented with 15% fetal calf serum (FCS) (Life Technologies, Inc., Gaithersburg, MD) whereas MZ-CRC-1 cells were grown in Dulbecco's modified Eagle's medium (DMEM) (BioWhittaker) supplemented with 10% FCS. The human papillary thyroid carcinoma cell line TPC-1 which was used as a model of Met overexpression, was routinely cultivated in DMEM with 10% FCS. N592 cells, derived from a human SCLC, were characterized by Kit activation through autocrine stimulation by the SCF ligand. N592 cells were cultured in RPMI 1640 supplemented with 10% FCS. The mouse SWISS3T3 and NIH3T3 fibroblasts were cultured in DMEM with 10% calf serum (Colorado Serum Company, Denver, CO). In addition, NIH3T3 cells transfected with different oncogenes were used. NIH3T3MEN2A transfectants express the short isoform of the RET-MEN2A (C634R) oncogene; NIH3T3KITA559 cells express KIT carrying the activating mutation Δ559 found in GISTs; NIH3T3FGFR3(Y373C) express the FGF-R3 gene carrying the activating mutation Y373C found in a human multiple myeloma cell line. 2N5A cells are NIH3T3 cells transformed by the COL1A1/PDGFB rearrangement generating autocrine stimulation of PDGF-R. All NIH3T3 transfectants were maintained in DMEM plus 5% calf serum in a 10% C02 atmosphere. Antiproliferative assays
Cells were trypsinized after 3 days (NIH3T3 and NIH3T3MEN2A cells) or 7 days (MTC cells) of treatment with Cpdl and counted by a Coulter Counter (Coulter Electronics, Luton U.K.). The concentrations able to inhibit cell proliferation by 50% (IC50) were calculated from the dose-response curves. Each experiment was performed in duplicate Antibodies The following polyclonal antibodies were used: anti-Ret recognizing a
COOH-terminal sequence (aa 1000-1014) common to the two Ret isoforms (Borrello M. G., et al., Mol. Cell Biol. 16, 2151, 1996); anti-cKit, anti-Met and anti-FGF-R3 from Santa Cruz Biotechnology (Santa Cruz, CA); anti-actin from Sigma (St. Louis, MO); anti-PDGF-R α/β from Upstate Biotechnology (Lake Placid, NY); anti phospho-cKit (Tyr 719) from Cell Signaling Technology (Beverly, MA). The mouse monoclonal anti-phosphotyrosine (anti-pTyr) 4G10 and anti-FGF-Rl antibodies were from Upstate Biotechnology. Immunoprecipitation and Western blotting For whole-cell extract preparation, cells were lysed in sodium dodecyl sulfate (SDS) sample buffer (62.5 mM Tris-HCl (pH 6.8), 2% SDS) with 1 mM PMSF, 10 μg/ml pepstatin, 12.5 μg/ml leupeptin, 100 KIU aprotinin, 1 mM sodium orthovanadate, linM sodium molybdate. Protein concentration was determined in appropriately diluted aliquots by the bicinchionic acid (BCA) method (Pierce, Rockford, IL), then samples were adjusted to a final concentration of 10%» glycerol, 5% β-mercaptoethanol, 0.001% bromophenol blue.
For immunoprecipitation experiments, cells were treated with Cpdl or solvent for the indicated times. Cell monolayers were rinsed twice with cold phosphate-buffered saline plus 0.1 mM sodium orthovanadate and then left for 20 min on ice in lysis buffer (50mM HEPES pH7.6, 150mM NaCl, 10 % glycerol, 1% Triton X-100, 1.5 mM MgCl2, 1 mM EGTA, 100 mM NaF, 10 mM sodium pyrophosphate, 10 μg/ml antipain, 20 μg/ml chymostatin, 10 μg/ml E64, 1 mg/ml pefabloc SC). Cells were then collected, aspirated through a 22-gauge needle and centrifuged at lOOOOg, for 20 min, at 4 °C. Protein concentration was determined by the BCA reagent (Pierce). Cell extracts were incubated with protein A-agarose and the indicated antibody for 2 hr at 4°C under rotation. After washing 3 times with 20mM HEPES pH 7.6, 150 mM NaCl, 10% glycerol, 0.1% Triton X-100, the immunoprecipitates were eluted with complete sample buffer.
Normalized immunoprecipitates (0.5 - 4 mg of cell extracts) or whole- cell lysates (30 - 60 μg) were resolved on SDS-PAGE and transferred to nitrocellulose filters. Membranes were incubated with primary antibodies at 4°C, overnight. Immunoreactive bands were revealed by . horseradish peroxidase-coniugated anti-mouse or anti-rabbit antibodies using enhanced chemiluminescence detection systems from Amersham Biosciences (Little Chalfont, United Kingdom) or Pierce. In vivo studies All experiments were carried out using female athymic nude CD-I mice,
8-11 -weeks old (Charles River, Calco, Italy). Mice were maintained in laminar flow rooms with constant temperature and humidity. Experimental protocols were approved by the Ethic Committee for Animal Experimentation of the Istituto Nazionale per lo Studio e la Cura dei Tumori (Milan, Italy), according to the United Kingdom Coordinating Committee on Cancer Research Guidelines (Workman P. et al., British Journal of Cancer, 77, 1, 1998).
The MTC cells TT (1.6xl07 cells) were s.c. inoculated in mice as a cell suspension from in vitro cell culture. Each control or drug-treated group included 8-10 tumors. Tumor cells were injected on day 0, and tumor growth was followed by biweekly measurements of tumor diameters with a Vernier caliper. Tumor weight (TW) was calculated according to the formula: TW (mg) = tumor volume (mm ) = d xD/2, where d and D are the shortest and the longest diameter, respectively. Drug treatment started when tumors were just measurable (mean TW about 50 mg), 25 days after the inoculum of tumor cells. Cpdl was dissolved in 5% ethanol, 5% Cremophor EL, 90% saline (0.9% NaCl) and delivered per os twice in a day (2qd) by a daily schedule for 10 days. Control mice received the solvent solution. Drug efficacy was assessed as percentage TW inhibition (TWP/o in drug- treated versus control mice expressed as: TWI%=100 - (mean TW treated/mean TW control x 100). For statistical analysis, TW in control and treated mice were compared on the day of TWI% evaluation, by Student's t test (two-tailed). P values less than 0.05 were considered statistically significant. Preparation of (E) l,3-dihydro-4-hydroxybenzyliden-5,6-dimethoxy-
(lH)-indol -2-one (Cpd 1). 1) Synthesis of 2-nitro-4,5-dimethoxyphenylacetic acid
Figure imgf000014_0001
C10H„NO6, M.W. 241,20 45 g (0.23 moles, 1 eq.) of 3,4-dimethoxyphenylacetic acid were dissolved in lOOmL (2.2 volumes) glacial acetic acid at 28°C-35°C, under N2 atmosphere and mechanic stirring. The solution was cooled to 15°-20°C and added with a mixture of fuming nitric acid (98%, 33 mL) in glacial acetic acid (25 mL) over a period of 45' . Once the addition was completed, precipitation of a red solid was observed. The suspension was poured in ice water (600 mL) and kept under stirring for 2h. The solid was filtrated, washed with water and dried at 60°C for 8h. 44g of the end product were obtained.
Yield 79.3% (mmol/mmol) TLC (Si02; Ethyl acetate 10/AcOH 0.5) Rfacid = 0.6; Rfproduct = 0.5 m.p. 199°-202°C
1H-NMR, (DMSO): 3.9 ppm (s, 6H); 4.0 ppm (s., 2H); 7.12 ppm (s.,
1H); 7.7 ppm (s., 1H) 2) Synthesis of l,3-dihydro-5,6-dimethoxy-(lH)-indol-2-one
Figure imgf000015_0001
9.2 g (38.14 mmoles, 1 eq.) of 3,4-dimethoxy-2-nitro-phenylacetic acid were suspended in glacial acetic acid (92 mL, 10 volumes) at 25°C under N2 atmosphere and mechanic stirring. The suspension was added with Fe° powder, 325 mesh, 97% (12.0g, 214.86 mmoles, 5.6 eq.) in two equal portions. The first portion was added at room temperature; then the mixture was refluxed and 30 min later the second Fe° portion was added. 30' later the reaction was complete, TLC (Si02; CHC13 9/MeOH 1), Rfnitro = 0.65,
• tindolinone ~ U. / 1. The grey suspension was cooled to room temperature, the acetic acid was evaporated under low pressure to a crude solid, which was suspended in chloroform (200 mL). The salts were filtrated off and the organic phase was washed with a NaCl saturated solution (100 mL), dried over Na2S04 and evaporated to dryness. The solid was suspended in ethyl ether (35 mL) for 30', filtrated and dried at 50°C for 2h. 6.7 g of a beige solid were obtained.
Yield 90.9% (mmol/mmol) m.p. 199°-201°C
TLC (Si02; Ethyl acetate 10/AcOH 0.5) Rfacid = 0.6; Rfproduct = 0.5 1H-NMR, (DMSO): 3.4 ppm (s, 2H); 3.69 ppm (s., 3H); 3.72 ppm (s.,
3H); 6.49 ppm (s., 1H); 6.92 ppm (s., 1H); 10.15 (S., 1H). 3) Synthesis of (E)-l,3-dihydro-4-hydroxybenzyliden-5,6-dimethoxy-lH- indol-2-one
Figure imgf000016_0001
C17H15N045 M.W. 297,31
6.7 g (36.9 mmoles, 1 eq.) of l,3-dihydro-5,6-dimethoxy-(lH)-indol-2- one were dissolved in anhydrous DMSO (50 mL) at room temperature. The solution was added with 4-hydroxybenzaldehyde (5.41 g, 44.3 mmoles, 1.2 eq.) and piperidine (4.38 g, 44.3 mmoles, 1.2 eq.), then stirred for 16 hours. The mixture was poured in H20 (250 mL) and HCl 0.5N (150mL), and precipitation of a solid was observed. The solution was cooled to 5°-10°C for 1 h, filtrated and dried under vacuum at 80°C for 2h. 13g of wet solid were obtained and then crystallized from absolute ethanol, yielding 6.77g of end product.
Yield 61.6% (mmol/mmol) m.p. 238°-240°C
Rf (Silica; Ethyl acetate 100%) = 0.68
Η-NMR, (DMSO): 3.6 ppm (s, 3H); 3.8 ppm (s., 3H); 6.5 ppm (s., IH);
6.9 ppm (d., 2H, J=8.6 Hz); 10 ppm (broad s.); 12.8 ppm (s.).
The E configuration of the esocyclic double bond in position 2 was determined by means of ID NOE NMR experiments.

Claims

1. The use of the compound l,3-dihydro-5,6-dimethoxy-3-[(4- hydroxyphenyl)methylene]-2H-indol-2-one or of non-toxic salts or isomers thereof for the preparation of a medicament for the treatment of tumors involving a tyrosine kinase selected from Met, PDGF-R, FGF-RI, FGF-R3, Kit, or an oncoprotein of the Ret family.
2. The use according to claim 1, for the treatment of tumors expressing RET oncoproteins carrying activating sequence mutations.
3. The use according to claim 2, wherein the RET oncoproteins include
MEN2 associated mutations.
4. The use according to claim 3, wherein the activated sequence mutations are RET/MEN2A (C634R), RET/MEN2A (C634W) and RET/MEN2B
(M918T).
5. The use according to claim 2-4, for the treatment of medullary thyroid carcinomas, pheochromocytoma, parathyroid hyperplasia, enteric ganglioneuroma.
6. The use according to claim 1, for the treatment of tumors bearing a
Met-activating alteration.
7. The use according to claim 6, wherein said tumors are of epithelial origin.
8. The use according to claim 7, for the treatment of kidney tumor.
9. The use according to claim 1, for the treatment of tumors expressing constitutively-activated Kit.
10. The use according to claim 9, wherein Kit is constitutively activated following to sequence mutations or involvement in autocrine loops.
11. The use according to claim 9, for the treatment of gastrointestinal stromal tumors, small cell lung carcinomas, leukemias or seminomas.
12. The use according to claim 1, for the treatment of tumors involving the uncontrolled activation of PDGF-R.
13. The use according to claim 12, wherein said tumors are gliomas and dermatofibrosarcoma protuberans.
14. The use according to claim 1, for the treatment of tumors highly expressing FGF-RI and/or its ligand bFGF.
15. The use according to claim 14, wherein said tumors are melanomas and gliomas.
16. The use according to claim 1, for the treatment of tumors expressing constitutive activating forms of FGF-R3.
17. The use according to claim 16, wherein said tumors are multiple myeloma, bladder and cervix carcinomas.
18. The use according to claims 12 and 14, for the inhibition of tumor angiogenesis.
19. A pharmaceutical composition containing as active ingredient the comp ound 1,3- dihy dro-5 , 6 -dimethoxy-3 - [(4-hy droxypheny l)methy lene] -2H- indol-2-one or a pharmaceutically acceptable salt thereof in combination with a pharmaceutically acceptable carrier, excipient or diluent.
20. The pharmaceutical composition according to claim 19, wherein said pharmaceutically acceptable carrier or diluent is suitable for oral or parenteral administration.
21. The pharmaceutical composition according to claim 19, further comprising a anti-tumor or anti-cancer agent which is different from 1,3- dihydro-5,6-dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one.
22. The pharmaceutical composition according to claim 19, wherein said anti-tumor or anti-cancer agent is selected from the group consisting of adriamycin, daunomycin, methotrexate, vincristin, 6-mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docitaxel, topotecan, irinotecam, gencitabine, Lpam, BCNU and VP-16.
23. A kit comprising, in separate containers, a compound l,3-dihydro-5,6- dimethoxy-3-[(4-hydroxyphenyl)methylene]-2H-indol-2-one or a pharmaceutically acceptable salt thereof and an anti-cancer or anti-tumor agent.
24. The kit according to claim 23, wherein said anti-tumor or anti-cancer agent is selected from the group consisting of adriamycin, daunomycin, methotrexate, vincristin, 6-mercaptopurine, cytosine arabinoside, cyclophosphamide, 5-FU, hexamethylmelamine, carboplatin, cisplatin, idarubycin, paclitaxel, docetaxel, topotecan, irinotecam, gemcitabine, L-PAM, BCNU and VP-16.
PCT/EP2003/007963 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof WO2004009083A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
EP03765081A EP1534271B1 (en) 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof
CA002493202A CA2493202A1 (en) 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof
MXPA05000781A MXPA05000781A (en) 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof.
US10/522,081 US20060258731A1 (en) 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof
JP2004522542A JP2005537277A (en) 2002-07-23 2003-07-22 Antitumor compounds and their therapeutic use
DE60307856T DE60307856T2 (en) 2002-07-23 2003-07-22 ANTITUMORIC CONNECTION AND ITS THERAPEUTIC USES
AU2003251437A AU2003251437A1 (en) 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof
US12/313,033 US20090130229A1 (en) 2002-07-23 2008-11-13 Antitumor uses of compound

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IT2002MI001620A ITMI20021620A1 (en) 2002-07-23 2002-07-23 ANTI-TUMORAL ACTIVITY COMPOUND
ITMI02A001620 2002-07-23

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/313,033 Continuation US20090130229A1 (en) 2002-07-23 2008-11-13 Antitumor uses of compound

Publications (2)

Publication Number Publication Date
WO2004009083A1 true WO2004009083A1 (en) 2004-01-29
WO2004009083A8 WO2004009083A8 (en) 2005-02-03

Family

ID=30130912

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/007963 WO2004009083A1 (en) 2002-07-23 2003-07-22 Antitumor compound and therapeutic uses thereof

Country Status (12)

Country Link
US (2) US20060258731A1 (en)
EP (1) EP1534271B1 (en)
JP (1) JP2005537277A (en)
CN (1) CN1668298A (en)
AT (1) ATE337001T1 (en)
AU (1) AU2003251437A1 (en)
CA (1) CA2493202A1 (en)
DE (1) DE60307856T2 (en)
ES (1) ES2271649T3 (en)
IT (1) ITMI20021620A1 (en)
MX (1) MXPA05000781A (en)
WO (1) WO2004009083A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005068424A1 (en) * 2004-01-20 2005-07-28 Cell Therapeutics Europe S.R.L. Indolinone derivatives as receptor tyrosine kinase ihibitors
EP1883403A2 (en) * 2005-04-29 2008-02-06 The Ohio State University Research Foundation Keratinocyte growth factor receptor - tyrosine specific inhibitors for the prevention of cancer metastatis
WO2008029123A1 (en) * 2006-09-07 2008-03-13 Astrazeneca Ab Method for evaluating patients for treatment with drugs targeting ret receptor tyrosine kinase

Families Citing this family (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103965200B (en) 2008-09-22 2016-06-08 阵列生物制药公司 As imidazo [1,2-B] pyridazine compound of the replacement of TRK kinase inhibitor
AR077468A1 (en) 2009-07-09 2011-08-31 Array Biopharma Inc PIRAZOLO COMPOUNDS (1,5-A) PYRIMIDINE SUBSTITUTED AS TRK-QUINASA INHIBITORS
SG11201703962XA (en) 2014-11-16 2017-06-29 Array Biopharma Inc Crystalline form of (s)-n-(5-((r)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide hydrogen sulfate
AU2016291676B2 (en) 2015-07-16 2020-04-30 Array Biopharma, Inc. Substituted pyrazolo[1,5-a]pyridine compounds as RET kinase inhibitors
RU2744852C2 (en) 2015-10-26 2021-03-16 Локсо Онколоджи, Инк. Point mutations in trk inhibitor-resistant malignant tumors and related methods
US10045991B2 (en) 2016-04-04 2018-08-14 Loxo Oncology, Inc. Methods of treating pediatric cancers
KR102400423B1 (en) 2016-04-04 2022-05-19 록쏘 온콜로지, 인코포레이티드 (S)-N-(5-((R)-2-(2,5-difluorophenyl)-pyrrolidin-1-yl)-pyrazolo[1,5-A]pyrimidine-3- Liquid formulation of yl)-3-hydroxypyrrolidine-1-carboxamide
CN109310694A (en) 2016-04-04 2019-02-05 洛克索肿瘤学股份有限公司 The method for treating Paediatric cancer
LT3800189T (en) 2016-05-18 2023-10-10 Loxo Oncology, Inc. Preparation of (s)-n-(5-((r)-2-(2,5-difluorophenyl)pyrrolidin-1-yl)pyrazolo[1,5-a]pyrimidin-3-yl)-3-hydroxypyrrolidine-1-carboxamide
TWI704148B (en) 2016-10-10 2020-09-11 美商亞雷生物製藥股份有限公司 Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190077A1 (en) 2016-10-10 2019-04-09 Array Biopharma Inc Substituted pyrazolo[1,5-a]pyridine compounds as ret kinase inhibitors
JOP20190092A1 (en) 2016-10-26 2019-04-25 Array Biopharma Inc PROCESS FOR THE PREPARATION OF PYRAZOLO[1,5-a]PYRIMIDINES AND SALTS THEREOF
WO2018136663A1 (en) 2017-01-18 2018-07-26 Array Biopharma, Inc. Ret inhibitors
JP6888101B2 (en) 2017-01-18 2021-06-16 アレイ バイオファーマ インコーポレイテッド Substituted pyrazolo [1,5-a] pyrazine compounds as RET kinase inhibitors
JOP20190213A1 (en) 2017-03-16 2019-09-16 Array Biopharma Inc Macrocyclic compounds as ros1 kinase inhibitors
TWI791053B (en) 2017-10-10 2023-02-01 美商亞雷生物製藥股份有限公司 Crystalline forms of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile and pharmaceutical composition thereof
TWI812649B (en) 2017-10-10 2023-08-21 美商絡速藥業公司 Formulations of 6-(2-hydroxy-2-methylpropoxy)-4-(6-(6-((6-methoxypyridin-3-yl)methyl)-3,6-diazabicyclo[3.1.1]heptan-3-yl)pyridin-3-yl)pyrazolo[1,5-a]pyridine-3-carbonitrile
TW201938169A (en) 2018-01-18 2019-10-01 美商亞雷生物製藥股份有限公司 Substituted pyrazolo[3,4-d]pyrimidine compounds as RET kinase inhibitors
CN111971286B (en) 2018-01-18 2023-04-14 阿雷生物药品公司 Substituted pyrrolo [2,3-d ] pyrimidine compounds as RET kinase inhibitors
US11472802B2 (en) 2018-01-18 2022-10-18 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridine compounds as RET kinase inhibitors
CN112996794A (en) 2018-09-10 2021-06-18 阿雷生物药品公司 Fused heterocyclic compounds as RET kinase inhibitors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6130238A (en) * 1997-06-20 2000-10-10 Sugen, Inc. 3-(cyclohexanoheteroarylidenyl)-2-indolinone protein tyrosine kinase inhibitors
US6133305A (en) * 1997-09-26 2000-10-17 Sugen, Inc. 3-(substituted)-2-indolinones compounds and use thereof as inhibitors of protein kinase activity
US6147106A (en) * 1997-08-20 2000-11-14 Sugen, Inc. Indolinone combinatorial libraries and related products and methods for the treatment of disease
US6268391B1 (en) * 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5502072A (en) * 1993-11-26 1996-03-26 Pfizer Inc. Substituted oxindoles
ES2290117T3 (en) * 2000-02-15 2008-02-16 Sugen, Inc. PROTEIN QUINASE 2-INDOLIN INHIBITORS REPLACED WITH PIRROL.

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6130238A (en) * 1997-06-20 2000-10-10 Sugen, Inc. 3-(cyclohexanoheteroarylidenyl)-2-indolinone protein tyrosine kinase inhibitors
US6268391B1 (en) * 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
US6147106A (en) * 1997-08-20 2000-11-14 Sugen, Inc. Indolinone combinatorial libraries and related products and methods for the treatment of disease
US6133305A (en) * 1997-09-26 2000-10-17 Sugen, Inc. 3-(substituted)-2-indolinones compounds and use thereof as inhibitors of protein kinase activity

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CINZIA LANZI AND ALL.: "Inhibition of transforming activity of the ret/ptc1 oncoprotein by a 2-indolinone derivative", INT.J.CANCER, vol. 85, 2000, pages 384 - 390, XP002257784 *
SARASIN A ET AL: "Mechanisms of mutagenesis in mammalian cells. Application to human thyroid tumours", COMPTES RENDUS DES SEANCES DE L'ACADEMIE DES SCIENCES. SERIE III: SCIENCES DE LA VIE, ELSEVIER, AMSTERDAM, NL, vol. 322, no. 2-3, February 1999 (1999-02-01), pages 143 - 149, XP004270260, ISSN: 0764-4469 *
SUAREZ H G: "Molecular basis of epithelial thyroid tumorigenesis", COMPTES RENDUS DES SEANCES DE L'ACADEMIE DES SCIENCES. SERIE III: SCIENCES DE LA VIE, ELSEVIER, AMSTERDAM, NL, vol. 323, no. 6, June 2000 (2000-06-01), pages 519 - 528, XP004330637, ISSN: 0764-4469 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005068424A1 (en) * 2004-01-20 2005-07-28 Cell Therapeutics Europe S.R.L. Indolinone derivatives as receptor tyrosine kinase ihibitors
EP1883403A2 (en) * 2005-04-29 2008-02-06 The Ohio State University Research Foundation Keratinocyte growth factor receptor - tyrosine specific inhibitors for the prevention of cancer metastatis
EP1883403A4 (en) * 2005-04-29 2011-02-16 Univ Ohio State Res Found Keratinocyte growth factor receptor - tyrosine specific inhibitors for the prevention of cancer metastatis
WO2008029123A1 (en) * 2006-09-07 2008-03-13 Astrazeneca Ab Method for evaluating patients for treatment with drugs targeting ret receptor tyrosine kinase

Also Published As

Publication number Publication date
CA2493202A1 (en) 2004-01-29
ES2271649T3 (en) 2007-04-16
CN1668298A (en) 2005-09-14
DE60307856T2 (en) 2007-04-12
US20060258731A1 (en) 2006-11-16
JP2005537277A (en) 2005-12-08
EP1534271A1 (en) 2005-06-01
WO2004009083A8 (en) 2005-02-03
AU2003251437A8 (en) 2004-02-09
ATE337001T1 (en) 2006-09-15
ITMI20021620A1 (en) 2004-01-23
MXPA05000781A (en) 2005-09-21
EP1534271B1 (en) 2006-08-23
DE60307856D1 (en) 2006-10-05
AU2003251437A1 (en) 2004-02-09
US20090130229A1 (en) 2009-05-21

Similar Documents

Publication Publication Date Title
US20090130229A1 (en) Antitumor uses of compound
EP3009428B1 (en) Diamino heterocyclic carboxamide compound
JP2011518174A (en) Compounds and methods for treating diseases associated with estrogen receptors
US20210154198A1 (en) Combination therapy involving diaryl macrocyclic compounds
EA028452B1 (en) Treatment of breast cancer
CA3074418C (en) Exon 18 and/or exon 21 mutant egfr selective inhibitor
KR20160013164A (en) Pharmaceutical combination comprising metformin and dihydroquercetin and its use for the treatment of cancer
US20180250261A1 (en) Method for treating cancer with a stat3 pathway inhibitor and kinase inhibitor
KR20160110952A (en) Novel methods for treating cancer
KR20100051837A (en) Treatment of pediatric tumors
CN104271129A (en) Methods of treating cancer using aurora kinase inhibitors
KR20170040338A (en) Treatment of androgen deprivation therapy associated symptoms
KR100847413B1 (en) Pharmaceutical Combinations Based on Pyridoindolone Derivatives and Anticancer Agents
JP2021527119A (en) Salt form
TW202227450A (en) Brain-migrating tumor therapeutic agent containing fused pyrimidine compound as active ingredient
WO2023198078A1 (en) Polycyclic compounds as kras g12d inhibitors
KR20230110296A (en) Kinase Inhibitor Combinations for Cancer Treatment
CN113164438A (en) Novel combination regimen for treating chemotherapy-refractory cancer
EA046190B1 (en) NEW COMBINED SOLUTION FOR THE TREATMENT OF CANCER RESISTANT TO CHEMOTHERAPY
KR20200061823A (en) Use of pyrimidinone derivatives having apoptosis of brain cancer stem cells
KR20200061822A (en) Use of pyrimidinedione derivatives having apoptosis of brain cancer stem cells
TW201143766A (en) Antitumor agent containing indole compound

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref document number: PA/a/2005/000781

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2493202

Country of ref document: CA

Ref document number: 2004522542

Country of ref document: JP

Ref document number: 20038173352

Country of ref document: CN

CFP Corrected version of a pamphlet front page
CR1 Correction of entry in section i

Free format text: IN PCT GAZETTE 05/2004 UNDER (71) THE NAME SHOULD READ "ISTITUTO NAZIONALE PER LO STUDIO E LA CURA DEI TUMORI"

WWE Wipo information: entry into national phase

Ref document number: 2003765081

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 2003765081

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006258731

Country of ref document: US

Ref document number: 10522081

Country of ref document: US

WWG Wipo information: grant in national office

Ref document number: 2003765081

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 10522081

Country of ref document: US