Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Pharmacol Res ; 177: 106076, 2022 03.
Article in English | MEDLINE | ID: mdl-35074524

ABSTRACT

Drug discovery from natural sources is going through a renaissance, having spent many decades in the shadow of synthetic molecule drug discovery, despite the fact that natural product-derived compounds occupy a much greater chemical space than those created through synthetic chemistry methods. With this new era comes new possibilities, not least the novel targets that have emerged in recent times and the development of state-of-the-art technologies that can be applied to drug discovery from natural sources. Although progress has been made with some immunomodulating drugs, there remains a pressing need for new agents that can be used to treat the wide variety of conditions that arise from disruption, or over-activation, of the immune system; natural products may therefore be key in filling this gap. Recognising that, at present, there is no authoritative article that details the current state-of-the-art of the immunomodulatory activity of natural products, this in-depth review has arisen from a joint effort between the International Union of Basic and Clinical Pharmacology (IUPHAR) Natural Products and Immunopharmacology Sections, with contributions from a number of world-leading researchers in the field of natural product drug discovery, to provide a "position statement" on what natural products has to offer in the search for new immunomodulatory argents. To this end, we provide a historical look at previous discoveries of naturally occurring immunomodulators, present a picture of the current status of the field and provide insight into the future opportunities and challenges for the discovery of new drugs to treat immune-related diseases.


Subject(s)
Biological Products , Pharmacology, Clinical , Biological Products/chemistry , Biological Products/pharmacology , Biological Products/therapeutic use , Drug Discovery , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Immunomodulating Agents
2.
Int J Mol Sci ; 22(23)2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34884436

ABSTRACT

Cancer therapies targeting the vascular endothelial growth factor (VEGF) signaling pathway can lead to renal damage by disrupting the glomerular ultrafiltration apparatus. The objective of the current study was to identify sensitive biomarkers for VEGF inhibition-induced glomerular changes in rats. Male Sprague-Dawley rats were administered an experimental VEGF receptor (VEGFR) inhibitor, ABT-123, for seven days to investigate the correlation of several biomarkers with microscopic and ultrastructural changes. Glomeruli obtained by laser capture microdissection were also subjected to gene expression analysis to investigate the underlying molecular events of VEGFR inhibition in glomerulus. ABT-123 induced characteristic glomerular ultrastructural changes in rats, including fusion of podocyte foot processes, the presence of subendothelial electron-dense deposits, and swelling and loss of fenestrations in glomerular endothelium. The subtle morphological changes cannot be detected with light microscopy or by changes in standard clinical chemistry and urinalysis. However, urinary albumin increased 44-fold as early as Day three. Urinary ß2-microglobulin levels were also increased. Other urinary biomarkers that are typically associated with tubular injury were not significantly impacted. Such patterns in urinary biomarkers can provide valuable diagnostic insight to VEGF inhibition therapy-induced glomeruli injuries.


Subject(s)
Kidney Diseases/urine , Protein Kinase Inhibitors/adverse effects , Signal Transduction/drug effects , beta 2-Microglobulin/urine , Albumins/metabolism , Animals , Biomarkers/urine , Disease Models, Animal , Kidney Diseases/chemically induced , Kidney Diseases/metabolism , Laser Capture Microdissection , Male , Rats , Rats, Sprague-Dawley , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
3.
Biochem Biophys Res Commun ; 491(3): 681-686, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28756225

ABSTRACT

Cancer cells have an unusually high requirement for the central and intermediary metabolite nicotinamide adenine dinucleotide (NAD+), and NAD+ depletion ultimately results in cell death. The rate limiting step within the NAD+ salvage pathway required for converting nicotinamide to NAD+ is catalyzed by nicotinamide phosphoribosyltransferase (NAMPT). Targeting NAMPT has been investigated as an anti-cancer strategy, and several highly selective small molecule inhibitors have been found to potently inhibit NAMPT in cancer cells, resulting in NAD+ depletion and cytotoxicity. To identify mechanisms that could cause resistance to NAMPT inhibitor treatment, we generated a human fibrosarcoma cell line refractory to the highly potent and selective NAMPT small molecule inhibitor, GMX1778. We uncovered novel and unexpected mechanisms of resistance including significantly increased expression of quinolinate phosphoribosyl transferase (QPRT), a key enzyme in the de novo NAD+ synthesis pathway. Additionally, exome sequencing of the NAMPT gene in the resistant cells identified a single heterozygous point mutation that was not present in the parental cell line. The combination of upregulation of the NAD+ de novo synthesis pathway through QPRT over-expression and NAMPT mutation confers resistance to GMX1778, but the cells are only partially resistant to next-generation NAMPT inhibitors. The resistance mechanisms uncovered herein provide a potential avenue to continue exploration of next generation NAMPT inhibitors to treat neoplasms in the clinic.


Subject(s)
Cyanides/administration & dosage , Cytokines/antagonists & inhibitors , Cytokines/genetics , Drug Resistance, Neoplasm/drug effects , Fibrosarcoma/drug therapy , Fibrosarcoma/metabolism , Guanidines/administration & dosage , NAD/biosynthesis , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/genetics , Anilides , Apoptosis/drug effects , Apoptosis/genetics , Arginine/analogs & derivatives , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Fibrosarcoma/genetics , Humans , Mutation/genetics , NAD/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Treatment Outcome
5.
Proc Natl Acad Sci U S A ; 107(28): 12634-9, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616035

ABSTRACT

Aurora kinase B inhibitors induce apoptosis secondary to polyploidization and have entered clinical trials as an emerging class of neocytotoxic chemotherapeutics. We demonstrate here that polyploidization neutralizes Mcl-1 function, rendering cancer cells exquisitely dependent on Bcl-XL/-2. This "addiction" can be exploited therapeutically by combining aurora kinase inhibitors and the orally bioavailable BH3 mimetic, ABT-263, which inhibits Bcl-XL, Bcl-2, and Bcl-w. The combination of ABT-263 with aurora B inhibitors produces a synergistic loss of viability in a range of cell lines of divergent tumor origin and exhibits more sustained tumor growth inhibition in vivo compared with aurora B inhibitor monotherapy. These data demonstrate that Bcl-XL/-2 is necessary to support viability during polyploidization in a variety of tumor models and represents a druggable molecular vulnerability with potential therapeutic utility.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Aniline Compounds , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Aurora Kinase B , Aurora Kinases , Enzyme Inhibitors/therapeutic use , Male , Mice , Neoplasms/genetics , Protein Serine-Threonine Kinases , Sulfonamides
6.
J Biol Chem ; 286(51): 43951-43958, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22020937

ABSTRACT

Eukaryotic elongation factor-2 kinase (eEF2K) relays growth and stress signals to protein synthesis through phosphorylation and inactivation of eukaryotic elongation factor 2 (eEF2). 1-Benzyl-3-cetyl-2-methylimidazolium iodide (NH125) is a widely accepted inhibitor of mammalian eEF2K and an efficacious anti-proliferation agent against different cancer cells. It implied that eEF2K could be an efficacious anticancer target. However, eEF2K siRNA was ineffective against cancer cells including those sensitive to NH125. To test if pharmacological intervention differs from siRNA interference, we identified a highly selective small molecule eEF2K inhibitor A-484954. Like siRNA, A-484954 had little effect on cancer cell growth. We carefully examined the effect of NH125 and A-484954 on phosphorylation of eEF2, the known cellular substrate of eEF2K. Surprisingly, NH125 increased eEF2 phosphorylation, whereas A-484954 inhibited the phosphorylation as expected for an eEF2K inhibitor. Both A-484954 and eEF2K siRNA inhibited eEF2K and reduced eEF2 phosphorylation with little effect on cancer cell growth. These data demonstrated clearly that the anticancer activity of NH125 was more correlated with induction of eEF2 phosphorylation than inhibition of eEF2K. Actually, induction of eEF2 phosphorylation was reported to correlate with inhibition of cancer cell growth. We compared several known inducers of eEF2 phosphorylation including AMPK activators and an mTOR inhibitor. Interestingly, stronger induction of eEF2 phosphorylation correlated with more effective growth inhibition. We also explored signal transduction pathways leading to NH125-induced eEF2 phosphorylation. Preliminary data suggested that NH125-induced eEF2 phosphorylation was likely mediated through multiple pathways. These observations identified an opportunity for a new multipathway approach to anticancer therapies.


Subject(s)
Gene Expression Regulation, Neoplastic , Imidazoles/pharmacology , Peptide Elongation Factor 2/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Design , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphorylation , RNA, Small Interfering/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , eIF-2 Kinase/metabolism
7.
J Pharmacol Exp Ther ; 343(3): 617-27, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22935731

ABSTRACT

ABT-348 [1-(4-(4-amino-7-(1-(2-hydroxyethyl)-1H-pyrazol-4-yl)thieno[3,2-c]pyridin-3-yl)phenyl)-3-(3-fluorophenyl)urea] is a novel ATP-competitive multitargeted kinase inhibitor with nanomolar potency (IC(50)) for inhibiting binding and cellular autophosphorylation of Aurora B (7 and 13 nM), C (1 and 13 nM), and A (120 and 189 nM). Cellular activity against Aurora B is reflected by inhibition of phosphorylation of histone H3, induction of polyploidy, and inhibition of proliferation of a variety of leukemia, lymphoma, and solid tumor cell lines (IC(50) = 0.3-21 nM). In vivo inhibition of Aurora B was confirmed in an engrafted leukemia model by observing a decrease in phosphorylation of histone H3 that persisted in a dose-dependent manner for 8 h and correlated with plasma concentration of ABT-348. Evaluation of ABT-348 across a panel of 128 kinases revealed additional potent binding activity (K(i) < 30 nM) against vascular endothelial growth factor receptor (VEGFR)/platelet-derived growth factor receptor (PDGFR) families and the Src family of cytoplasmic tyrosine kinases. VEGFR/PDGFR binding activity correlated with inhibition of autophosphorylation in cells and inhibition of vascular endothelial growth factor (VEGF)-stimulated endothelial cell proliferation (IC(50) ≤ 0.3 nM). Evidence of on-target activity in vivo was provided by the potency for blocking VEGF-mediated vascular permeability and inducing plasma placental growth factor. Activity against the Src kinase family was evident in antiproliferative activity against BCR-ABL chronic myeloid leukemia cells and cells expressing the gleevec-resistant BCR-ABL T315I mutation. On the basis of its unique spectrum of activity, ABT-348 was evaluated and found effective in representative solid tumor [HT1080 and pancreatic carcinoma (MiaPaCa), tumor stasis] and hematological malignancy (RS4;11, regression) xenografts. These results provide the rationale for clinical assessment of ABT-348 as a therapeutic agent in the treatment of cancer.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Phenylurea Compounds/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , src-Family Kinases/antagonists & inhibitors , Aminopyridines/chemistry , Aminopyridines/pharmacokinetics , Aminopyridines/therapeutic use , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Histones/antagonists & inhibitors , Human Umbilical Vein Endothelial Cells , Humans , Leukemia, Experimental/drug therapy , Leukemia, Experimental/enzymology , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Molecular Structure , NIH 3T3 Cells , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/therapeutic use , Time Factors , Xenograft Model Antitumor Assays
9.
Bioorg Med Chem Lett ; 22(14): 4750-5, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22695126

ABSTRACT

In an effort to identify kinase inhibitors with dual KDR/Aurora B activity and improved aqueous solubility compared to the Abbott dual inhibitor ABT-348, a series of novel pyrazole pyrimidines structurally related to kinase inhibitor AS703569 were prepared. SAR work provided analogs with significant cellular activity, measureable aqueous solubility and moderate antitumor activity in a mouse tumor model after weekly ip dosing. Unfortunately these compounds were pan-kinase inhibitors that suffered from narrow therapeutic indices which prohibited their use as antitumor agents.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrazoles/chemistry , Pyrimidines/chemistry , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Amination , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aurora Kinase B , Aurora Kinases , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Models, Molecular , Molecular Structure , Pyrimidines/pharmacology , Structure-Activity Relationship , Xenograft Model Antitumor Assays
10.
Bioorg Med Chem Lett ; 22(9): 3208-12, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22465635

ABSTRACT

In an effort to identify multi-targeted kinase inhibitors with a novel spectrum of kinase activity, a screen of Abbott proprietary KDR inhibitors against a broad panel of kinases was conducted and revealed a series of thienopyridine ureas with promising activity against the Aurora kinases. Modification of the diphenyl urea and C7 moiety of these compounds provided potent inhibitors with good pharmacokinetic profiles that were efficacious in mouse tumor models after oral dosing. Compound 2 (ABT-348) of this series is currently undergoing Phase I clinical trials in solid and hematological cancer populations.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Urea/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Humans , Mice , Protein Kinase Inhibitors/chemistry , Vascular Endothelial Growth Factor A
11.
Blood ; 113(17): 4052-62, 2009 Apr 23.
Article in English | MEDLINE | ID: mdl-19144991

ABSTRACT

To further investigate potential mechanisms of resistance to FLT3 inhibitors, we developed a resistant cell line by long-term culture of MV4-11 cells with ABT-869, designated as MV4-11-R. Gene profiling reveals up-regulation of FLT3LG (FLT3 ligand) and BIRC5 (survivin), but down-regulation of SOCS1, SOCS2, and SOCS3 in MV4-11-R cells. Hypermethylation of these SOCS genes leads to their transcriptional silencing. Survivin is directly regulated by STAT3. Stimulation of the parental MV4-11 cells with FLT3 ligand increases the expression of survivin and phosphorylated protein STAT1, STAT3, STAT5. Targeting survivin by short-hairpin RNA (shRNA) in MV4-11-R cells induces apoptosis and augments ABT-869-mediated cytotoxicity. Overexpression of survivin protects MV4-11 from apoptosis. Subtoxic dose of indirubin derivative (IDR) E804 resensitizes MV4-11-R to ABT-869 treatment by inhibiting STAT signaling activity and abolishing survivin expression. Combining IDR E804 with ABT-869 shows potent in vivo efficacy in the MV4-11-R xenograft model. Taken together, these results demonstrate that enhanced activation of STAT pathways and overexpression of survivin are important mechanisms of resistance to ABT-869, suggesting that the STAT pathways and survivin could be potential targets for reducing resistance developed in patients receiving FLT3 inhibitors.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Microtubule-Associated Proteins/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Epoxy Compounds/pharmacology , Female , Humans , Indazoles/pharmacology , Inhibitor of Apoptosis Proteins , Leukemia, Myeloid, Acute/genetics , Ligands , Mice , Mice, Inbred BALB C , Mice, Nude , Microtubule-Associated Proteins/genetics , Phenylurea Compounds/pharmacology , STAT3 Transcription Factor/genetics , Sesquiterpenes/pharmacology , Substrate Specificity , Survivin , Up-Regulation/genetics , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/metabolism
13.
J Clin Invest ; 117(10): 3107-17, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17823654

ABSTRACT

The effectiveness of IFN-alpha2b for human multiple myeloma has been variable. TRAIL has been proposed to mediate IFN-alpha2b apoptosis in myeloma. In this study we assessed the effects of IFN-alpha2b signaling on the apoptotic activity of TRAIL and human myeloma cell survival. While TRAIL was one of the most potently induced proapoptotic genes in myeloma cells following IFN-alpha2b treatment, less than 20% of myeloma cells underwent apoptosis. Thus, we hypothesized that an IFN-stimulated gene (ISG) with prosurvival activity might suppress TRAIL-mediated apoptosis. Consistent with this, IFN-alpha2b stabilized mitochondria and inhibited caspase-3 activation, which antagonized TRAIL-mediated apoptosis and cytotoxicity after 24 hours of cotreatment in cell lines and in fresh myeloma cells, an effect not evident after 72 hours. Induced expression of G1P3, an ISG with largely unknown function, was correlated with the antiapoptotic activity of IFN-alpha2b. Ectopically expressed G1P3 localized to mitochondria and antagonized TRAIL-mediated mitochondrial potential loss, cytochrome c release, and apoptosis, suggesting specificity of G1P3 for the intrinsic apoptosis pathway. Furthermore, RNAi-mediated downregulation of G1P3 restored IFN-alpha2b-induced apoptosis. Our data identify the direct role of a mitochondria-localized prosurvival ISG in antagonizing the effect of TRAIL. Curtailing G1P3-mediated antiapoptotic signals could improve therapies for myeloma or other malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Interferon-alpha/pharmacology , Mitochondrial Proteins/metabolism , Multiple Myeloma/metabolism , TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Apoptosis/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Gene Expression Regulation , Gene Expression Regulation, Leukemic , Humans , Interferon alpha-2 , Mitochondria/chemistry , Mitochondria/metabolism , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Multiple Myeloma/genetics , Recombinant Proteins , TNF-Related Apoptosis-Inducing Ligand/metabolism
14.
J Med Chem ; 51(5): 1231-41, 2008 Mar 13.
Article in English | MEDLINE | ID: mdl-18260617

ABSTRACT

A series of benzoisoxazoles and benzoisothiazoles have been synthesized and evaluated as inhibitors of receptor tyrosine kinases (RTKs). Structure-activity relationship studies led to the identification of 3-amino benzo[ d]isoxazoles, incorporating a N, N'-diphenyl urea moiety at the 4-position that potently inhibited both the vascular endothelial growth factor receptor (VEGFR) and platelet-derived growth factor receptor families of RTKs. Within this series, orally bioavailable compounds possessing promising pharmacokinetic profiles were identified, and a number of compounds demonstrated in vivo efficacy in models of VEGF-stimulated vascular permeability and tumor growth. In particular, compound 50 exhibited an ED 50 of 2.0 mg/kg in the VEGF-stimulated uterine edema model and 81% inhibition in the human fibrosarcoma (HT1080) tumor growth model when given orally at a dose of 10 mg/kg/day.


Subject(s)
Isoxazoles/chemical synthesis , Models, Molecular , Oxazoles/chemical synthesis , Phenylurea Compounds/chemical synthesis , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Binding Sites , Biological Availability , Capillary Permeability/drug effects , Cell Line , Cell Line, Tumor , Edema/drug therapy , Female , Humans , Isoxazoles/pharmacokinetics , Isoxazoles/pharmacology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Oxazoles/pharmacokinetics , Oxazoles/pharmacology , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/pharmacology , Phosphorylation , Structure-Activity Relationship , Uterus/blood supply , Xenograft Model Antitumor Assays
15.
Leuk Res ; 32(7): 1091-100, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18160102

ABSTRACT

Neoangiogenesis plays an important role in leukemogenesis. We investigated the in vivo anti-leukemic effect of ABT-869 against AML with wild-type FLT3 using RFP transfected HL60 cells with in vivo imaging technology on both the subcutaneous and systemic leukemia xenograft models. ABT-869 showed a five-fold inhibition of tumor growth in comparison with vehicle control. IHC analysis revealed that ABT-869 decreased p-VEGFR1, Ki-67 labeling index, VEGF and remarkably increased apoptotic cells in the xenograft models. ABT-869 also reduced the leukemia burden and prolonged survival. Our study supports the rationale for clinically testing an anti-angiogenesis agent in AML with wild-type FLT3.


Subject(s)
Antineoplastic Agents/therapeutic use , Indazoles/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , fms-Like Tyrosine Kinase 3/metabolism , Bone Marrow Transplantation , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Transplantation, Heterologous
17.
Bioorg Med Chem Lett ; 18(1): 386-90, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-18023347
18.
J Med Chem ; 50(7): 1584-97, 2007 Apr 05.
Article in English | MEDLINE | ID: mdl-17343372

ABSTRACT

In our continued efforts to search for potent and novel receptor tyrosine kinase (RTK) inhibitors as potential anticancer agents, we discovered, through a structure-based design, that 3-aminoindazole could serve as an efficient hinge-binding template for kinase inhibitors. By incorporating an N,N'-diaryl urea moiety at the C4-position of 3-aminodazole, a series of RTK inhibitors were generated, which potently inhibited the tyrosine kinase activity of the vascular endothelial growth factor receptor and the platelet-derived growth factor receptor families. A number of compounds with potent oral activity were identified by utilizing an estradiol-induced mouse uterine edema model and an HT1080 human fibrosarcoma xenograft tumor model. In particular, compound 17p (ABT-869) was found to possess favorable pharmacokinetic profiles across different species and display significant tumor growth inhibition in multiple preclinical animal models.


Subject(s)
Angiogenesis Inhibitors/chemical synthesis , Indazoles/chemical synthesis , Phenylurea Compounds/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Adenosine Triphosphate/chemistry , Administration, Oral , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/pharmacology , Animals , Binding Sites , Edema/chemically induced , Edema/pathology , Estradiol , Female , Humans , Hydrophobic and Hydrophilic Interactions , Indazoles/chemistry , Indazoles/pharmacology , Male , Mice , Models, Molecular , NIH 3T3 Cells , Phenylurea Compounds/chemistry , Phenylurea Compounds/pharmacology , Phosphorylation , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Uterus/drug effects , Uterus/pathology , Xenograft Model Antitumor Assays
19.
Int J Oncol ; 31(6): 1391-402, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17982666

ABSTRACT

We investigated the effect of a novel histone deacetylase inhibitor, A-423378.0, on the colon carcinoma cell line HCT116 and genetically modified derivatives lacking either p21(cip1/waf1) or p53. HCT116 cell lines were incubated with A-423378.0 at different concentrations for 3-120 h. Cell viability, proliferation and apoptosis rates were determined and verified by western blot, detection of mitochondrial membrane potential breakdown DeltaPsi(m), activation of caspases-3, -8 and cytokeratin 18 cleavage. A subcutaneous xenograft model was established in NMRI mice with daily intraperitoneal injections of 10 mg/kg for 14 days. All three HCT116 cell lines responded to A-423378.0 treatment in a dose- and time-dependent manner via induction of apoptosis as measured by breakdown of DeltaPsi(m) and BrdU incorporation. We identified that A-423378.0 induced the expression of TRAIL and TRAIL receptor, especially TRAIL-R2/hDR5, which was up-regulated in HCT116 cells after treatment with A-423378.0. In vivo, a growth inhibitory effect was observed with HDAC-I treatment, which was paralleled by a down-regulation of PCNA and a concomitant induction of apoptosis. Treatment of wild-type or knock-out HCT116 cells with A-423378.0 exerts potent anti-proliferative and pro-apoptotic effects in vitro and in vivo. A-423378.0 was able to induce apoptosis in both p21(WAF1) and p53 deficient tumour cells, which appeared to be mediated by the intrinsic cell death pathway. Interestingly, the effects of A-423378.0 on the extrinsic cell death pathway through activation of TRAIL and its signalling pathway indicate that A-423378.0 may be a potent new therapeutic compound for the treatment of advanced colorectal cancer.


Subject(s)
Apoptosis/drug effects , Colonic Neoplasms/drug therapy , Cyclin-Dependent Kinase Inhibitor p21/physiology , Enzyme Inhibitors/pharmacology , Histone Deacetylase Inhibitors , Thiazoles/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , Caspases/metabolism , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Male , Membrane Potentials/drug effects , Mice , RNA, Messenger/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/analysis , bcl-2-Associated X Protein/physiology
20.
Mol Cancer Ther ; 5(4): 1007-13, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16648572

ABSTRACT

The properties of several multitargeted receptor tyrosine kinase inhibitors have been studied for their inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling. A structurally novel, multitargeted tyrosine kinase inhibitor (ABT-869), imatinib (STI571), and four compounds currently in clinical development (AG013736, BAY 43-9006, CHIR258, and SU11248) were tested for inhibition of CSF-1R signaling in both the enzymatic and cellular assays. ABT-869 showed potent CSF-1R inhibition in both the enzyme and cell-based assays (IC50s < 20 nmol/L). In contrast to a previous report, we have found that imatinib has activity against human CSF-1R in both assays at submicromolar concentrations. In enzyme assays, we have found that the inhibition of CSF-1R by both ABT-869 and imatinib are competitive with ATP, with Ki values of 3 and 120 nmol/L, respectively. SU11248 is a potent inhibitor of CSF-1R in the enzyme assay (IC50 = 7 nmol/L) and inhibits receptor phosphorylation in the cellular assay (IC50 = 61 nmol/L). AG013736 was also a potent inhibitor of CSF-1R in both assays (enzyme, IC50 = 16 nmol/L; cellular, IC50 = 21 nmol/L), whereas BAY 43-9006 is less potent in the enzyme assay (IC50 = 107 nmol/L) than in the cellular system (IC50 = 20 nmol/L). In contrast, we found that CHIR258 had less activity in the cellular assay (IC50 = 535 nmol/L) relative to its enzymatic potency (IC50 = 26 nmol/L). These results show the use of a cell-based assay to confirm the inhibitory activity of lead compounds and drug candidates, such as ABT-869, against the CSF-1R protein in situ.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indazoles/pharmacology , Phenylurea Compounds/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , 3T3 Cells , Adenosine Triphosphate/metabolism , Animals , Benzamides , Binding Sites , Humans , Imatinib Mesylate , Kinetics , Lead/pharmacology , Mice , Phosphorylation , Piperazines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL