Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 23(23): 6363-9, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24138939

ABSTRACT

Structure-activity relationship (SAR) studies on a highly potent series of arylamide FMS inhibitors were carried out with the aim of improving FMS kinase selectivity, particularly over KIT. Potent compound 17r (FMS IC50 0.7 nM, FMS cell IC50 6.1 nM) was discovered that had good PK properties and a greater than fivefold improvement in selectivity for FMS over KIT kinase in a cellular assay relative to the previously reported clinical candidate 4. This improved selectivity was manifested in vivo by no observed decrease in circulating reticulocytes, a measure of bone safety, at the highest studied dose. Compound 17r was highly active in a mouse pharmacodynamic model and demonstrated disease-modifying effects in a dose-dependent manner in a strep cell wall-induced arthritis model of rheumatoid arthritis in rats.


Subject(s)
Amides/pharmacology , Heterocyclic Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Amides/chemical synthesis , Amides/chemistry , Animals , Heterocyclic Compounds/chemical synthesis , Heterocyclic Compounds/chemistry , Male , Mice , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Rats , Rats, Sprague-Dawley , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Stereoisomerism , Structure-Activity Relationship
2.
J Med Chem ; 54(22): 7860-83, 2011 Nov 24.
Article in English | MEDLINE | ID: mdl-22039836

ABSTRACT

A class of potent inhibitors of colony-stimulating factor-1 receptor (CSF-1R or FMS), as exemplified by 8 and 21, was optimized to improve pharmacokinetic and pharmacodynamic properties and potential toxicological liabilities. Early stage absorption, distribution, metabolism, and excretion assays were employed to ensure the incorporation of druglike properties resulting in the selection of several compounds with good activity in a pharmacodynamic screening assay in mice. Further investigation, utilizing the type II collagen-induced arthritis model in mice, culminated in the selection of anti-inflammatory development candidate JNJ-28312141 (23, FMS IC(50) = 0.69 nM, cell assay IC(50) = 2.6 nM). Compound 23 also demonstrated efficacy in rat adjuvant and streptococcal cell wall-induced models of arthritis and has entered phase I clinical trials.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Imidazoles/chemical synthesis , Piperidines/chemical synthesis , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Arthritis, Experimental/drug therapy , Arthritis, Experimental/etiology , Arthritis, Experimental/pathology , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation/drug effects , Crystallography, X-Ray , Female , Humans , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , In Vitro Techniques , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Microsomes, Liver/metabolism , Models, Molecular , Piperidines/pharmacokinetics , Piperidines/pharmacology , Protein Conformation , Rats , Rats, Inbred Lew , Receptor, Macrophage Colony-Stimulating Factor/chemistry , Solubility , Stereoisomerism , Structure-Activity Relationship
4.
Mol Cancer Ther ; 8(11): 3151-61, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19887542

ABSTRACT

There is increasing evidence that tumor-associated macrophages promote the malignancy of some cancers. Colony-stimulating factor-1 (CSF-1) is expressed by many tumors and is a growth factor for macrophages and mediates osteoclast differentiation. Herein, we report the efficacy of a novel orally active CSF-1 receptor (CSF-1R) kinase inhibitor, JNJ-28312141, in proof of concept studies of solid tumor growth and tumor-induced bone erosion. H460 lung adenocarcinoma cells did not express CSF-1R and were not growth inhibited by JNJ-28312141 in vitro. Nevertheless, daily p.o. administration of JNJ-28312141 caused dose-dependent suppression of H460 tumor growth in nude mice that correlated with marked reductions in F4/80(+) tumor-associated macrophages and with increased plasma CSF-1, a possible biomarker of CSF-1R inhibition. Furthermore, the tumor microvasculature was reduced in JNJ-28312141-treated mice, consistent with a role for macrophages in tumor angiogenesis. In separate studies, JNJ-28312141 was compared with zoledronate in a model in which MRMT-1 mammary carcinoma cells inoculated into the tibias of rats led to severe cortical and trabecular bone lesions. Both agents reduced tumor growth and preserved bone. However, JNJ-28312141 reduced the number of tumor-associated osteoclasts superior to zoledronate. JNJ-28312141 exhibited additional activity against FMS-related receptor tyrosine kinase-3 (FLT3). To more fully define the therapeutic potential of this new agent, JNJ-28312141 was evaluated in a FLT3-dependent acute myeloid leukemia tumor xenograft model and caused tumor regression. In summary, this novel CSF-1R/FLT3 inhibitor represents a new agent with potential therapeutic activity in acute myeloid leukemia and in settings where CSF-1-dependent macrophages and osteoclasts contribute to tumor growth and skeletal events.


Subject(s)
Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Imidazoles/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Bone Neoplasms/enzymology , Bone Neoplasms/pathology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Growth Processes/drug effects , Cell Line, Tumor , Female , Humans , Immunohistochemistry , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/enzymology , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Nude , Osteoclasts/drug effects , Osteoclasts/pathology , Rats , Rats, Sprague-Dawley , Receptor, Macrophage Colony-Stimulating Factor/blood , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Substrate Specificity , Xenograft Model Antitumor Assays , fms-Like Tyrosine Kinase 3/metabolism
5.
J Med Chem ; 52(4): 1081-99, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19193011

ABSTRACT

A series of pyrido[2,3-d]pyrimidin-5-ones has been synthesized and evaluated as inhibitors of the kinase domain of macrophage colony-stimulating factor-1 receptor (FMS). FMS inhibitors may be useful in treating rheumatoid arthritis and other chronic inflammatory diseases. Structure-based optimization of the lead amide analogue 10 led to hydroxamate analogue 37, which possessed excellent potency and an improved pharmacokinetic profile. During the chronic phase of streptococcal cell wall-induced arthritis in rats, compound 37 (10, 3, and 1 mg/kg) was highly effective at reversing established joint swelling. In an adjuvant-induced arthritis model in rats, 37 prevented joint swelling partially at 10 mg/kg. In this model, osteoclastogenesis and bone erosion were prevented by low doses (1 or 0.33 mg/kg) that had minimal impact on inflammation. These data underscore the potential of FMS inhibitors to prevent erosions and reduce symptoms in rheumatoid arthritis.


Subject(s)
Anti-Inflammatory Agents/chemistry , Arthritis, Rheumatoid/drug therapy , Pyrimidinones/chemistry , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Bone Resorption/prevention & control , Disease Models, Animal , Inflammation/drug therapy , Osteoclasts/drug effects , Pharmacokinetics , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Structure, Tertiary , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Rats , Structure-Activity Relationship
6.
Bioorg Med Chem Lett ; 18(12): 3632-7, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18495479

ABSTRACT

An anti-inflammatory 1,2,4-phenylenetriamine-containing series of FMS inhibitors with a potential to form reactive metabolites was transformed into a series with equivalent potency by incorporation of carbon-based replacement groups. Structure-based modeling provided the framework to efficiently effect this transformation and restore potencies to previous levels. This optimization removed a risk factor for potential idiosyncratic drug reactions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drug Design , Enzyme Inhibitors/pharmacology , Phenylenediamines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Computer Simulation , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Hydrogen Bonding , Inhibitory Concentration 50 , Models, Molecular , Molecular Structure , Phenylenediamines/chemical synthesis , Phenylenediamines/chemistry , Stereoisomerism , Structure-Activity Relationship
9.
Bioorg Med Chem Lett ; 18(6): 2097-102, 2008 Mar 15.
Article in English | MEDLINE | ID: mdl-18289848

ABSTRACT

A series of 3,4,6-substituted 2-quinolones has been synthesized and evaluated as inhibitors of the kinase domain of macrophage colony-stimulating factor-1 receptor (FMS). The fully optimized compound, 4-(4-ethyl-phenyl)-3-(2-methyl-3H-imidazol-4-yl)-2-quinolone-6-carbonitrile 21b, has an IC(50) of 2.5 nM in an in vitro assay and 5.0 nM in a bone marrow-derived macrophage cellular assay. Inhibition of FMS signaling in vivo was also demonstrated in a mouse pharmacodynamic model.


Subject(s)
Macrophages/drug effects , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Quinolones/chemical synthesis , Quinolones/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Administration, Oral , Animals , Biological Availability , Cell Proliferation/drug effects , Fluorescence Polarization , Genes, fos/genetics , Macrophage Colony-Stimulating Factor/metabolism , Mice , Mice, Inbred C57BL , Molecular Structure , Quinolones/pharmacokinetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Spleen/metabolism , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 17(22): 6070-4, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17904845

ABSTRACT

A series of 2'-aminoanilides have been identified which exhibit potent and selective inhibitory activity against the cFMS tyrosine kinase. Initial SAR studies within this series are described which examine aroyl and amino group substitutions, as well as the introduction of hydrophilic substituents on the benzene core. Compound 47 inhibits the isolated enzyme (IC(50)=0.027 microM) and blocks CSF-1-induced proliferation of bone marrow-derived macrophages (IC(50)=0.11 microM) and as such, serves as a lead candidate for further optimization studies.


Subject(s)
Anilides/chemical synthesis , Anilides/pharmacology , Anti-Inflammatory Agents/pharmacology , Piperidines/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Anilides/chemistry , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Macrophages/drug effects , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Structure-Activity Relationship
11.
J Biochem Biophys Methods ; 65(2-3): 107-20, 2005 Dec 31.
Article in English | MEDLINE | ID: mdl-16325916

ABSTRACT

Development of alphavbeta3-integrin inhibitors has been hampered by a lack of pharmacodynamic endpoints to identify doses that inhibit alphavbeta3 in vivo. To address this need, we developed an alphavbeta3 radioreceptor assay (RRA) that could be performed in 100% plasma. The RRA was based on 125I-echistatin binding to plate-immobilized alphavbeta3. Small molecule alphavbeta3 inhibitors efficiently competed echistatin binding to alphavbeta3 when the assay was carried out in buffer. However, when carried out in 100% plasma, the RRA revealed a 45 to >3000-fold loss in compound potencies. The losses in potency reflected, in part, the high plasma protein binding by the compounds examined. The RRA was adapted as an ex vivo pharmacodynamic model. Echistatin binding was measured in the presence of plasma harvested at timed intervals from rats dosed with select compounds. Using this pharmacodynamic model, compound and dose selection was optimized for further testing in models of corneal angiogenesis. Moderate anti-angiogenic activity was achieved when rats were dosed sufficient to achieve sustained (>50%) plasma inhibition through the trough interval. Thus, the RRA provided a simple technique to rank order compound potency in plasma, and could find general use as an ex vivo pharmacodynamic assay to select compounds and doses for preclinical and clinical proof-of-principle studies.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/blood , Radioligand Assay/methods , Angiogenesis Inhibitors/pharmacokinetics , Angiogenesis Inhibitors/pharmacology , Animals , Blood Platelets/drug effects , Blood Platelets/metabolism , Blood Proteins/metabolism , Cornea/blood supply , Cornea/drug effects , Drug Evaluation, Preclinical , In Vitro Techniques , Intercellular Signaling Peptides and Proteins , Male , Neovascularization, Pathologic/prevention & control , Peptides/blood , Peptides/pharmacokinetics , Protein Binding , Rats , Rats, Sprague-Dawley
12.
Virology ; 332(2): 491-7, 2005 Feb 20.
Article in English | MEDLINE | ID: mdl-15680414

ABSTRACT

Natural killer (NK) cells play an important role in both innate and adaptive antiviral immune responses. The adaptive response typically requires that virus-specific antibodies decorate infected cells which then direct NK cell lysis through a CD16 mediated process termed antibody-dependent cellular cytotoxicity (ADCC). In this report, we employ a highly polymerized chimeric IgG1/IgA immunoglobulin (Ig) fusion protein that, by virtue of its capacity to extensively crosslink CD16, activates NK cells while directing the lysis of infected target cells. We employ HIV as a model system, and demonstrate that freshly isolated NK cells preloaded with an HIV gp120-specific chimeric IgG1/IgA fusion protein efficiently lyse HIV-infected target cells at picomolar concentrations. NK cells pre-armed in this manner retain the capacity to kill targets over an extended period of time. This strategy may have application to other disease states including various viral infections and cancers.


Subject(s)
HIV Infections/immunology , Killer Cells, Natural/immunology , Antibody-Dependent Cell Cytotoxicity , Antigens, CD/immunology , Calcium Signaling/physiology , Cell Line, Tumor , Flow Cytometry , Humans , Immunoglobulin G/immunology , Immunotherapy/methods , Receptors, IgG/immunology
13.
J Biol Chem ; 277(13): 11456-64, 2002 Mar 29.
Article in English | MEDLINE | ID: mdl-11805109

ABSTRACT

Drug toxicities associated with HAART lend urgency to the development of new anti-HIV therapies. Inhibition of viral replication at the entry stage of the viral life cycle is an attractive strategy because it prevents de novo infection. Soluble CD4 (sCD4), the first drug in this class, failed to suppress viral replication in vivo. At least three factors contributed to this failure: sCD4 demonstrated poor neutralizing activity against most primary isolates of HIV in vitro; it demonstrated an intrinsic capacity to enhance viral replication at low concentrations; and it exhibited a relatively short half-life in vivo. Many anti-gp120 monoclonal antibodies, including neutralizing monoclonal antibodies also enhance viral replication at suboptimal concentrations. Advances in our understanding of the events leading up to viral entry suggest strategies by which this activity can be diminished. We hypothesized that by constructing a sCD4-based molecule that is large, binds multiple gp120s simultaneously, and is highly avid toward gp120, we could remove its capacity to enhance viral entry. Here we describe the construction of a polymeric CD4-IgG1 fusion protein. The hydrodynamic radius of this molecule is approximately 12 nm. It can bind at least 10 gp120 subunits with binding kinetics that suggest a highly avid interaction toward virion-associated envelope. This protein does not enhance viral replication at suboptimal concentrations. These observations may aid in the design of new therapeutics and vaccines.


Subject(s)
AIDS Vaccines/metabolism , CD4 Immunoadhesins/metabolism , AIDS Vaccines/immunology , AIDS Vaccines/therapeutic use , Antibodies, Monoclonal/immunology , Biopolymers , CD4 Immunoadhesins/immunology , CD4-Positive T-Lymphocytes/virology , Cells, Cultured , Chromatography, Gel , Coculture Techniques , HIV Envelope Protein gp120/immunology , HIV-1/immunology , HIV-1/physiology , Humans , Membrane Fusion/immunology , Neutralization Tests , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...