Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Bioorg Med Chem ; 23(10): 2387-407, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25882525

ABSTRACT

Compound 1, a hit from the screening of our chemical collection displaying activity against JAK2, was deconstructed for SAR analysis into three regions, which were explored. A series of compounds was synthesized leading to the identification of the potent and orally bioavailable JAK2 inhibitor 16 (NMS-P830), which showed an encouraging tumour growth inhibition in SET-2 xenograft tumour model, with evidence for JAK2 pathway suppression demonstrated by in vivo pharmacodynamic effects.


Subject(s)
Amides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Janus Kinase 2/antagonists & inhibitors , Leukemia, Megakaryoblastic, Acute/drug therapy , Protein Kinase Inhibitors/chemical synthesis , Pyrroles/chemical synthesis , Amides/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression , High-Throughput Screening Assays , Humans , Janus Kinase 2/chemistry , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Leukemia, Megakaryoblastic, Acute/enzymology , Leukemia, Megakaryoblastic, Acute/genetics , Leukemia, Megakaryoblastic, Acute/pathology , Megakaryocyte Progenitor Cells/drug effects , Megakaryocyte Progenitor Cells/enzymology , Megakaryocyte Progenitor Cells/pathology , Mice , Mice, Nude , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Kinase Inhibitors/pharmacology , Pyrroles/pharmacology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
2.
Bioorg Med Chem ; 22(15): 4135-50, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24980703

ABSTRACT

In the last decade the heat shock protein 90 (Hsp90) has emerged as a major therapeutic target and many efforts have been dedicated to the discovery of Hsp90 inhibitors as new potent anticancer agents. Here we report the identification of a novel class of Hsp90 inhibitors by means of a biophysical FAXS-NMR based screening of a library of fragments. The use of X-ray structure information combined with modeling studies enabled the fragment evolution of the initial triazoloquinazoline hit to a class of compounds with nanomolar potency and drug-like properties suited for further lead optimization.


Subject(s)
HSP90 Heat-Shock Proteins/antagonists & inhibitors , Quinazolines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Design , Drug Evaluation, Preclinical , HSP90 Heat-Shock Proteins/metabolism , Humans , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Molecular Dynamics Simulation , Protein Structure, Tertiary , Quinazolines/chemical synthesis , Quinazolines/pharmacology , Structure-Activity Relationship
3.
Bioorg Med Chem ; 21(22): 7047-63, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24100158

ABSTRACT

Novel small molecule inhibitors of heat shock protein 90 (Hsp90) were discovered with the help of a fragment based drug discovery approach (FBDD) and subsequent optimization with a combination of structure guided design, parallel synthesis and application of medicinal chemistry principles. These efforts led to the identification of compound 18 (NMS-E973), which displayed significant efficacy in a human ovarian A2780 xenograft tumor model, with a mechanism of action confirmed in vivo by typical modulation of known Hsp90 client proteins, and with a favorable pharmacokinetic and safety profile.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Isoxazoles/chemistry , Isoxazoles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Binding Sites , Biomarkers, Tumor/metabolism , Catalytic Domain , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Drug Design , Drug Evaluation, Preclinical , Female , HSP90 Heat-Shock Proteins/metabolism , Humans , Isoxazoles/therapeutic use , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Binding/drug effects , Structure-Activity Relationship , Transplantation, Heterologous
4.
Mol Cancer Ther ; 17(3): 603-613, 2018 03.
Article in English | MEDLINE | ID: mdl-29237806

ABSTRACT

Chordomas are rare bone tumors with no approved therapy. These tumors express several activated tyrosine kinase receptors, which prompted attempts to treat patients with tyrosine kinase inhibitors. Although clinical benefit was observed in phase II clinical trials with imatinib and sorafenib, and sporadically also with EGFR inhibitors, therapies evaluated to date have shown modest activity. With the goal of identifying new drugs with immediate therapeutic potential for chordoma patients, we collected clinically approved drugs and other advanced inhibitors of MET, PDGFRß, and EGFR tyrosine kinases, and assessed their antiproliferative activity against a panel of chordoma cell lines. Chordoma cell lines were not responsive to MET and PDGFRß inhibitors. U-CH1 and UM-Chor1 were sensitive to all EGFR inhibitors, whereas the remaining cell lines were generally insensitive to these drugs. Afatinib was the only EGFR inhibitor with activity across the chordoma panel. We then investigated the molecular mechanisms behind the responses observed and found that the antiproliferative IC50s correlate with the unique ability of afatinib to promote degradation of EGFR and brachyury, an embryonic transcription factor considered a key driver of chordoma. Afatinib displayed potent antitumor efficacy in U-CH1, SF8894, CF322, and CF365 chordoma tumor models in vivo In the panel analyzed, high EGFR phosphorylation and low AXL and STK33 expression correlated with higher sensitivity to afatinib and deserve further investigation as potential biomarkers of response. These data support the use of afatinib in clinical trials and provide the rationale for the upcoming European phase II study on afatinib in advanced chordoma. Mol Cancer Ther; 17(3); 603-13. ©2017 AACR.


Subject(s)
Afatinib/therapeutic use , Bone Neoplasms/drug therapy , Chordoma/drug therapy , ErbB Receptors/antagonists & inhibitors , Fetal Proteins/antagonists & inhibitors , T-Box Domain Proteins/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chordoma/genetics , Chordoma/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice, Nude , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
5.
Cancer Chemother Pharmacol ; 60(2): 229-35, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17089166

ABSTRACT

PURPOSE: Edotecarin (J-107088, formerly ED-749) is a potent indolocarbazole topoisomerase-I inhibitor that has the potential to treat solid tumors. The current studies evaluated the potency and antitumor activity of edotecarin, as a single agent and in combination with capecitabine or docetaxel. METHODS: Antiproliferative activity was tested in vitro in a panel of 13 mammary cell lines and antitumor efficacy was tested in vivo in various breast cancer models. RESULTS: Edotecarin inhibited cellular proliferation in breast carcinoma cell lines: 50% inhibitory concentrations ranged from 8 nmol/L in SKBR-3 cells to approximately 30 micromol/L in BT20 cells. Single dose and weekly intravenous treatments with edotecarin 30 and 150 mg/kg produced significant antitumor activity in the SKBR-3 human breast carcinoma xenograft model, with no major toxicities, compared with vehicle solvent treatment. Daily administration of edotecarin 15 mg/kg for 10 days was not well tolerated, whereas the total dose of 150 mg/kg was safe when administered in a single injection. Edotecarin 3 and 30 mg/kg given after docetaxel in the nude mouse SKBR-3 xenograft model produced tumor growth delays that were greater than those observed with either agent alone and with no toxicity as evaluated on the basis of body weight reduction (<20%). Furthermore, edotecarin 3 mg/kg in combination with capecitabine produced more than additive effects and the combination was well tolerated. However, edotecarin at a dose of 30 mg/kg in combination with capecitabine was lethal. Edotecarin also exhibited potent antitumor activity against xenografted human MX-1 cells, MMTV-v-Ha-ras oncogene-driven mouse breast tumors, and chemically induced rat mammary tumors. CONCLUSIONS: The data suggest that edotecarin may be useful as a single agent or a component of combination chemotherapy regimens for treating human breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Carbazoles/therapeutic use , Indoles/therapeutic use , Mammary Neoplasms, Experimental/drug therapy , Topoisomerase I Inhibitors , Animals , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Inhibitory Concentration 50 , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Rats , Rats, Sprague-Dawley , Taxoids/therapeutic use
6.
Sci Rep ; 7(1): 9226, 2017 08 23.
Article in English | MEDLINE | ID: mdl-28835717

ABSTRACT

Chordomas are rare, slowly growing tumors with high medical need, arising in the axial skeleton from notochord remnants. The transcription factor "brachyury" represents a distinctive molecular marker and a key oncogenic driver of chordomas. Tyrosine kinase receptors are also expressed, but so far kinase inhibitors have not shown clear clinical efficacy in chordoma patients. The need for effective therapies is extremely high, but the paucity of established chordoma cell lines has limited preclinical research. Here we describe the isolation of the new Chor-IN-1 cell line from a recurrent sacral chordoma and its characterization as compared to other chordoma cell lines. Chor-IN-1 displays genomic identity to the tumor of origin and has morphological features, growth characteristics and chromosomal abnormalities typical of chordoma, with expression of brachyury and other relevant biomarkers. Chor-IN-1 gene variants, copy number alterations and kinome gene expression were analyzed in comparison to other four chordoma cell lines, generating large scale DNA and mRNA genomic data that can be exploited for the identification of novel pharmacological targets and candidate predictive biomarkers of drug sensitivity in chordoma. The establishment of this new, well characterized chordoma cell line provides a useful tool for the identification of drugs active in chordoma.


Subject(s)
Chordoma/genetics , Genomics , Biopsy , Cell Line, Tumor , Chordoma/metabolism , Chordoma/pathology , Chromosome Aberrations , DNA Copy Number Variations , Gene Expression Regulation, Neoplastic , Genomics/methods , Humans , Immunohistochemistry , Karyotype , Male , Middle Aged
7.
Mol Cancer Ther ; 15(4): 628-39, 2016 04.
Article in English | MEDLINE | ID: mdl-26939704

ABSTRACT

Activated ALK and ROS1 tyrosine kinases, resulting from chromosomal rearrangements, occur in a subset of non-small cell lung cancers (NSCLC) as well as other tumor types and their oncogenic relevance as actionable targets has been demonstrated by the efficacy of selective kinase inhibitors such as crizotinib, ceritinib, and alectinib. More recently, low-frequency rearrangements of TRK kinases have been described in NSCLC, colorectal carcinoma, glioblastoma, and Spitzoid melanoma. Entrectinib, whose discovery and preclinical characterization are reported herein, is a novel, potent inhibitor of ALK, ROS1, and, importantly, of TRK family kinases, which shows promise for therapy of tumors bearing oncogenic forms of these proteins. Proliferation profiling against over 200 human tumor cell lines revealed that entrectinib is exquisitely potent in vitro against lines that are dependent on the drug's pharmacologic targets. Oral administration of entrectinib to tumor-bearing mice induced regression in relevant human xenograft tumors, including the TRKA-dependent colorectal carcinoma KM12, ROS1-driven tumors, and several ALK-dependent models of different tissue origins, including a model of brain-localized lung cancer metastasis. Entrectinib is currently showing great promise in phase I/II clinical trials, including the first documented objective responses to a TRK inhibitor in colorectal carcinoma and in NSCLC. The drug is, thus, potentially suited to the therapy of several molecularly defined cancer settings, especially that of TRK-dependent tumors, for which no approved drugs are currently available. Mol Cancer Ther; 15(4); 628-39. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Indazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Anaplastic Lymphoma Kinase , Animals , Benzamides/chemistry , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Disease Models, Animal , Enzyme Activation/drug effects , Humans , Indazoles/chemistry , Magnetic Resonance Imaging , Male , Mice , Mice, Transgenic , Mortality , Protein Kinase Inhibitors/chemistry , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Translocation, Genetic , Treatment Outcome , Xenograft Model Antitumor Assays
8.
Eur J Cancer ; 41(10): 1453-9, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15913986

ABSTRACT

In this study, we have shown the intracellular distribution of choline and phosphatidylcholine fluorescent derivatives in human breast carcinoma cells using confocal microscopy. The fluorescent choline derivatives ethanamimium 2-hydroxy-N,N-dimethyl-N-[2-N-(2,1,3-benzoxadiazol-4-amine,-N-methyl,-7- nitro)-ethyl] bromide (NBD-choline) and C(6)-NBD-phosphatidylcholine (C(6)-NBD-PC) were used in this work. NBD-choline was easily internalised into drug sensitive MCF-7 and in multidrug resistant MCF-7/DX cells. The probe was found to localise in the endoplasmic reticulum of sensitive cells and in the Golgi of multidrug resistant cells. In contrast, very low accumulation was found in normal MCF10A cells. For C(6)-NBD-PC, a similar pattern of localisation was found in tumour cells, but a significant uptake was also observed in normal cells. Unlike NBD-choline, C(6)-NBD-PC appears not to discriminate between normal and tumour cells. These results are consistent with previously published results showing higher levels of (11)C-choline uptake in malignant lesions seen with positron emission tomography (PET) in vivo imaging. Our results suggest that using NBD-choline and laser scanning confocal fluorescence microscopy (LSCFM) could be a useful tool to study choline metabolism in cancer cells and to consolidate PET imaging findings.


Subject(s)
Breast Neoplasms/metabolism , Choline/metabolism , Phosphatidylcholines/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Choline/analogs & derivatives , Female , Fluorescence , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Positron-Emission Tomography , Tumor Cells, Cultured
9.
ChemMedChem ; 10(2): 276-95, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25430902

ABSTRACT

Aberrant activation of the mitogen-activated protein kinase (MAPK)-mediated pathway components, RAF-MEK-ERK, is frequently observed in human cancers and clearly contributes to oncogenesis. As part of a project aimed at finding inhibitors of B-Raf, a key player in the MAPK cascade, we originally identified a thiazole derivative endowed with high potency and selectivity, optimal in vitro ADME properties, and good pharmacokinetic profiles in rodents, but that suffers from elevated hERG inhibitory activity. An optimization program was thus undertaken, focused mainly on the elaboration of the R(1) and R(2) groups of the scaffold. This effort ultimately led to N-(4-{2-(1-cyclopropylpiperidin-4-yl)-4-[3-(2,5-difluorobenzenesulfonylamino)-2-fluorophenyl]thiazol-5-yl}-pyridin-2-yl)acetamide (20), which maintains favorable in vitro and in vivo properties, but lacks hERG liability. Besides exhibiting potent antiproliferative activity against only cell lines bearing B-Raf V600E or V600D mutations, compound 20 also intriguingly shows a weaker "paradoxical" activation of MEK in non-mutant B-Raf cells than other known B-Raf inhibitors. It also demonstrates very good efficacy in vivo against the A375 xenograft melanoma model (tumor volume inhibition >90% at 10 mg kg(-1) ); it is therefore a suitable candidate for preclinical development.


Subject(s)
Antineoplastic Agents/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/chemistry , Thiazoles/chemistry , Administration, Oral , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , ERG1 Potassium Channel , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Ether-A-Go-Go Potassium Channels/metabolism , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/drug effects , Protein Binding , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Protein Structure, Tertiary , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Sulfonamides/therapeutic use , Sulfonamides/toxicity , Thiazoles/pharmacology , Thiazoles/therapeutic use , Thiazoles/toxicity , Transplantation, Heterologous
10.
Mol Oncol ; 8(8): 1495-507, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24962792

ABSTRACT

The NTRK1 gene encodes Tropomyosin-related kinase A (TRKA), the high-affinity Nerve Growth Factor Receptor. NTRK1 was originally isolated from a colorectal carcinoma (CRC) sample as component of a somatic rearrangement (TPM3-NTRK1) resulting in expression of the oncogenic chimeric protein TPM3-TRKA, but there has been no subsequent report regarding the relevance of this oncogene in CRC. The KM12 human CRC cell line expresses the chimeric TPM3-TRKA protein and is hypersensitive to TRKA kinase inhibition. We report the detailed characterization of the TPM3-NTRK1 genomic rearrangement in KM12 cells and through a cellular screening approach, the identification of NMS-P626, a novel highly potent and selective TRKA inhibitor. NMS-P626 suppressed TPM3-TRKA phosphorylation and downstream signaling in KM12 cells and showed remarkable antitumor activity in mice bearing KM12 tumors. Finally, using quantitative reverse transcriptase PCR and immunohistochemistry (IHC) we identified the TPM3-NTRK1 rearrangement in a CRC clinical sample, therefore suggesting that this chromosomal translocation is indeed a low frequency recurring event in CRC and that such patients might benefit from therapy with TRKA kinase inhibitors.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Receptor, trkA/antagonists & inhibitors , Receptor, trkA/metabolism , Tropomyosin/metabolism , Animals , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Immunoprecipitation , In Vitro Techniques , Mice , Protein Binding/drug effects
11.
Genet Test Mol Biomarkers ; 17(3): 254-9, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23356232

ABSTRACT

The generation of biological data on wide panels of tumor cell lines is recognized as a valid contribution to the cancer research community. However, research laboratories can benefit from this knowledge only after the identity of each individual cell line used in the experiments is verified and matched to external sources. Among the methods employed to assess cell line identity, DNA fingerprinting by profiling Short Tandem Repeat (STR) at variable loci has become the method of choice. However, the analysis of cancer cell lines is sometimes complicated by their intrinsic genetic instability, resulting in multiple allele calls per locus. In addition, comparison of data across different sources must deal with the heterogeneity of published profiles both in terms of number and type of loci used. The aim of this work is to provide the scientific community a homogeneous reference dataset for 300 widely used tumor cell lines, profiled in parallel on 16 loci. This large dataset is interfaced with an in-house developed software tool for Cell Line Identity Finding by Fingerprinting (CLIFF), featuring an original identity score calculation, which facilitates the comparison of STR profiles from different sources and enables accurate calls when multiple loci are present. CLIFF additionally allows import and query of proprietary STR profile datasets.


Subject(s)
DNA Fingerprinting , Microsatellite Repeats , Neoplasms/genetics , Algorithms , Alleles , Animals , Cell Line, Tumor , Electrophoresis, Capillary , Humans , Mice , Multiplex Polymerase Chain Reaction , Neoplasm Transplantation , Neoplasms/pathology
12.
PLoS One ; 8(3): e58424, 2013.
Article in English | MEDLINE | ID: mdl-23520509

ABSTRACT

CD56 is expressed in 15-20% of acute myeloid leukaemias (AML) and is associated with extramedullary diffusion, multidrug resistance and poor prognosis. We describe the establishment and characterisation of a novel disseminated model of AML (AML-NS8), generated by injection into mice of leukaemic blasts freshly isolated from a patient with an aggressive CD56(+) monoblastic AML (M5a). The model reproduced typical manifestations of this leukaemia, including presence of extramedullary masses and central nervous system involvement, and the original phenotype, karyotype and genotype of leukaemic cells were retained in vivo. Recently Polo-Like Kinase 1 (PLK1) has emerged as a new candidate drug target in AML. We therefore tested our PLK1 inhibitor NMS-P937 in this model either in the engraftment or in the established disease settings. Both schedules showed good efficacy compared to standard therapies, with a significant increase in median survival time (MST) expecially in the established disease setting (MST = 28, 36, 62 days for vehicle, cytarabine and NMS-P937, respectively). Importantly, we could also demonstrate that NMS-P937 induced specific biomarker modulation in extramedullary tissues. This new in vivo model of CD56(+) AML that recapitulates the human tumour lends support for the therapeutic use of PLK1 inhibitors in AML.


Subject(s)
CD56 Antigen , Cell Cycle Proteins/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Neoplasms, Experimental/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyrazoles/pharmacology , Quinazolines/pharmacology , Adult , Animals , Cell Cycle Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
13.
Mol Cancer Ther ; 9(8): 2243-54, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20682657

ABSTRACT

Altered expression and activity of cyclin-dependent kinase (CDK) and tropomyosin receptor kinase (TRK) families are observed in a wide variety of tumors. In those malignancies with aberrant CDK activation, the retinoblastoma protein (pRb) pathway is deregulated, leading to uncontrolled cell proliferation. Constitutive activation of TRKs is instead linked to cancer cell survival and dissemination. Here, we show that the novel small-molecule PHA-848125, a potent dual inhibitor of CDKs and TRKs, possesses significant antitumor activity. The compound inhibits cell proliferation of a wide panel of tumoral cell lines with submicromolar IC(50). PHA-848125-treated cells show cell cycle arrest in G(1) and reduced DNA synthesis, accompanied by inhibition of pRb phosphorylation and modulation of other CDK-dependent markers. The compound additionally inhibits phosphorylation of TRKA and its substrates in cells, which functionally express this receptor. Following oral administration, PHA-848125 has significant antitumor activity in various human xenografts and carcinogen-induced tumors as well as in disseminated primary leukemia models, with plasma concentrations in rodents in the same range as those found active in inhibiting cancer cell proliferation. Mechanism of action was also confirmed in vivo as assessed in tumor biopsies from treated mice. These results show that the dual CDK-TRK inhibitor PHA-848125 has the potential for being a novel and efficacious targeted drug for cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Multigene Family , Protein Kinases/metabolism , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Quinazolines/administration & dosage , Quinazolines/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/metabolism , Humans , Mice , Phosphorylation/drug effects , Pyrazoles/chemistry , Pyrazoles/pharmacokinetics , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Rats , Treatment Outcome , Xenograft Model Antitumor Assays
14.
J Med Chem ; 52(16): 5152-63, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19603809

ABSTRACT

The discovery of a novel class of inhibitors of cyclin dependent kinases (CDKs) is described. Starting from compound 1, showing good potency as inhibitor of CDKs but being poorly selective against a panel of serine-threonine and tyrosine kinases, new analogues were synthesized. Enhancement in selectivity, antiproliferative activity against A2780 human ovarian carcinoma cells, and optimization of the physical properties and pharmacokinetic profile led to the identification of highly potent and orally available compounds. Compound 28 (PHA-848125), which in the preclinical xenograft A2780 human ovarian carcinoma model showed good efficacy and was well tolerated upon repeated daily treatments, was identified as a drug candidate for further development. Compound 28 is currently undergoing phase I and phase II clinical trials.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Pyrazoles/chemical synthesis , Quinazolines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Humans , Male , Mice , Mice, Nude , Models, Molecular , Neoplasm Transplantation , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Solubility , Structure-Activity Relationship , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL