Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Bioorg Med Chem Lett ; 80: 129084, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36423823

ABSTRACT

In the treatment of non-small cell lung cancer (NSCLC), patients harboring exon 20 insertion mutations in the epidermal growth factor receptor (EGFR) gene (EGFR) have few effective therapies because this subset of mutants is generally resistant to most currently approved EGFR inhibitors. This report describes the structure-guided design of a novel series of potent, irreversible inhibitors of EGFR exon 20 insertion mutations, including the V769_D770insASV and D770_N771insSVD mutants. Extensive structure-activity relationship (SAR) studies led to the discovery of mobocertinib (compound 21c), which inhibited growth of Ba/F3 cells expressing the ASV insertion with a half-maximal inhibitory concentration of 11 nM and with selectivity over wild-type EGFR. Daily oral administration of mobocertinib induced tumor regression in a Ba/F3 ASV xenograft mouse model at well-tolerated doses. Mobocertinib was approved in September 2021 for the treatment of adult patients with advanced NSCLC with EGFR exon 20 insertion mutations with progression on or after platinum-based chemotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Mice , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mutagenesis, Insertional , Mutation , ErbB Receptors , Exons , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Cancer Discov ; 11(7): 1672-1687, 2021 07.
Article in English | MEDLINE | ID: mdl-33632773

ABSTRACT

Most EGFR exon 20 insertion (EGFRex20ins) driver mutations in non-small cell lung cancer (NSCLC) are insensitive to approved EGFR tyrosine kinase inhibitors (TKI). To address the limitations of existing therapies targeting EGFR-mutated NSCLC, mobocertinib (TAK-788), a novel irreversible EGFR TKI, was specifically designed to potently inhibit oncogenic variants containing activating EGFRex20ins mutations with selectivity over wild-type EGFR. The in vitro and in vivo activity of mobocertinib was evaluated in engineered and patient-derived models harboring diverse EGFRex20ins mutations. Mobocertinib inhibited viability of various EGFRex20ins-driven cell lines more potently than approved EGFR TKIs and demonstrated in vivo antitumor efficacy in patient-derived xenografts and murine orthotopic models. These findings support the ongoing clinical development of mobocertinib for the treatment of EGFRex20ins-mutated NSCLC. SIGNIFICANCE: No oral EGFR-targeted therapies are approved for EGFR exon 20 insertion (EGFRex20ins) mutation-driven NSCLC. Mobocertinib is a novel small-molecule EGFR inhibitor specifically designed to target EGFRex20ins mutants. Preclinical data reported here support the clinical development of mobocertinib in patients with NSCLC with EGFR exon 20 insertion mutations.See related commentary by Pacheco, p. 1617.This article is highlighted in the In This Issue feature, p. 1601.


Subject(s)
Aniline Compounds/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Exons , Indoles/therapeutic use , Lung Neoplasms/drug therapy , Pyrimidines/therapeutic use , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor/drug effects , ErbB Receptors , Humans , Indoles/pharmacology , Lung Neoplasms/genetics , Mice , Mutagenesis, Insertional , Pyrimidines/pharmacology , Xenograft Model Antitumor Assays
3.
Clin Cancer Res ; 22(22): 5527-5538, 2016 Nov 15.
Article in English | MEDLINE | ID: mdl-27780853

ABSTRACT

PURPOSE: Non-small cell lung cancers (NSCLCs) harboring ALK gene rearrangements (ALK+) typically become resistant to the first-generation anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor (TKI) crizotinib through development of secondary resistance mutations in ALK or disease progression in the brain. Mutations that confer resistance to second-generation ALK TKIs ceritinib and alectinib have also been identified. Here, we report the structure and first comprehensive preclinical evaluation of the next-generation ALK TKI brigatinib. EXPERIMENTAL DESIGN: A kinase screen was performed to evaluate the selectivity profile of brigatinib. The cellular and in vivo activities of ALK TKIs were compared using engineered and cancer-derived cell lines. The brigatinib-ALK co-structure was determined. RESULTS: Brigatinib potently inhibits ALK and ROS1, with a high degree of selectivity over more than 250 kinases. Across a panel of ALK+ cell lines, brigatinib inhibited native ALK (IC50, 10 nmol/L) with 12-fold greater potency than crizotinib. Superior efficacy of brigatinib was also observed in mice with ALK+ tumors implanted subcutaneously or intracranially. Brigatinib maintained substantial activity against all 17 secondary ALK mutants tested in cellular assays and exhibited a superior inhibitory profile compared with crizotinib, ceritinib, and alectinib at clinically achievable concentrations. Brigatinib was the only TKI to maintain substantial activity against the most recalcitrant ALK resistance mutation, G1202R. The unique, potent, and pan-ALK mutant activity of brigatinib could be rationalized by structural analyses. CONCLUSIONS: Brigatinib is a highly potent and selective ALK inhibitor. These findings provide the molecular basis for the promising activity being observed in ALK+, crizotinib-resistant patients with NSCLC being treated with brigatinib in clinical trials. Clin Cancer Res; 22(22); 5527-38. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/drug effects , Lung Neoplasms/drug therapy , Organophosphorus Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Crizotinib , Hep G2 Cells , Humans , Lung Neoplasms/metabolism , Mutation/drug effects , Pyrazoles/pharmacology , Pyridines/pharmacology , Sulfones/pharmacology , U937 Cells
4.
J Med Chem ; 59(10): 4948-64, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27144831

ABSTRACT

In the treatment of echinoderm microtubule-associated protein-like 4 (EML4)-anaplastic lymphoma kinase positive (ALK+) non-small-cell lung cancer (NSCLC), secondary mutations within the ALK kinase domain have emerged as a major resistance mechanism to both first- and second-generation ALK inhibitors. This report describes the design and synthesis of a series of 2,4-diarylaminopyrimidine-based potent and selective ALK inhibitors culminating in identification of the investigational clinical candidate brigatinib. A unique structural feature of brigatinib is a phosphine oxide, an overlooked but novel hydrogen-bond acceptor that drives potency and selectivity in addition to favorable ADME properties. Brigatinib displayed low nanomolar IC50s against native ALK and all tested clinically relevant ALK mutants in both enzyme-based biochemical and cell-based viability assays and demonstrated efficacy in multiple ALK+ xenografts in mice, including Karpas-299 (anaplastic large-cell lymphomas [ALCL]) and H3122 (NSCLC). Brigatinib represents the most clinically advanced phosphine oxide-containing drug candidate to date and is currently being evaluated in a global phase 2 registration trial.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Lung Neoplasms/drug therapy , Organophosphorus Compounds/pharmacology , Phosphines/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Administration, Oral , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , Mice, SCID , Molecular Conformation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Phosphines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship
5.
Clin Cancer Res ; 20(22): 5745-5755, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25239608

ABSTRACT

PURPOSE: KIT is the major oncogenic driver of gastrointestinal stromal tumors (GIST). Imatinib, sunitinib, and regorafenib are approved therapies; however, efficacy is often limited by the acquisition of polyclonal secondary resistance mutations in KIT, with those located in the activation (A) loop (exons 17/18) being particularly problematic. Here, we explore the KIT-inhibitory activity of ponatinib in preclinical models and describe initial characterization of its activity in patients with GIST. EXPERIMENTAL DESIGN: The cellular and in vivo activities of ponatinib, imatinib, sunitinib, and regorafenib against mutant KIT were evaluated using an accelerated mutagenesis assay and a panel of engineered and GIST-derived cell lines. The ponatinib-KIT costructure was also determined. The clinical activity of ponatinib was examined in three patients with GIST previously treated with all three FDA-approved agents. RESULTS: In engineered and GIST-derived cell lines, ponatinib potently inhibited KIT exon 11 primary mutants and a range of secondary mutants, including those within the A-loop. Ponatinib also induced regression in engineered and GIST-derived tumor models containing these secondary mutations. In a mutagenesis screen, 40 nmol/L ponatinib was sufficient to suppress outgrowth of all secondary mutants except V654A, which was suppressed at 80 nmol/L. This inhibitory profile could be rationalized on the basis of structural analyses. Ponatinib (30 mg daily) displayed encouraging clinical activity in two of three patients with GIST. CONCLUSION: Ponatinib possesses potent activity against most major clinically relevant KIT mutants and has demonstrated preliminary evidence of activity in patients with refractory GIST. These data strongly support further evaluation of ponatinib in patients with GIST.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/genetics , Imidazoles/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/genetics , Pyridazines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Benzamides/pharmacology , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Exons , Female , Gastrointestinal Stromal Tumors/diagnosis , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Imidazoles/chemistry , Imidazoles/therapeutic use , Indoles/pharmacology , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Mutation , Neoplasm Recurrence, Local , Piperazines/pharmacology , Protein Binding , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/chemistry , Pyridazines/chemistry , Pyridazines/therapeutic use , Pyrimidines/pharmacology , Pyrroles/pharmacology , Sunitinib , Tomography, X-Ray Computed , Tumor Burden/drug effects , Tumor Burden/genetics , Xenograft Model Antitumor Assays
6.
Chem Biol Drug Des ; 78(6): 999-1005, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22034911

ABSTRACT

Activating gene rearrangements of anaplastic lymphoma kinase (ALK) have been identified as driver mutations in non-small-cell lung cancer, inflammatory myofibroblastic tumors, and other cancers. Crizotinib, a dual MET/ALK inhibitor, has demonstrated promising clinical activity in patients with non-small-cell lung cancer and inflammatory myofibroblastic tumors harboring ALK translocations. Inhibitors of driver kinases often elicit kinase domain mutations that confer resistance, and such mutations have been successfully predicted using in vitro mutagenesis screens. Here, this approach was used to discover an extensive set of ALK mutations that can confer resistance to crizotinib. Mutations at 16 residues were identified, structurally clustered into five regions around the kinase active site, which conferred varying degrees of resistance. The screen successfully predicted the L1196M, C1156Y, and F1174L mutations, recently identified in crizotinib-resistant patients. In separate studies, we demonstrated that crizotinib has relatively modest potency in ALK-positive non-small-cell lung cancer cell lines. A more potent ALK inhibitor, TAE684, maintained substantial activity against mutations that conferred resistance to crizotinib. Our study identifies multiple novel mutations in ALK that may confer clinical resistance to crizotinib, suggests that crizotinib's narrow selectivity window may underlie its susceptibility to such resistance and demonstrates that a more potent ALK inhibitor may be effective at overcoming resistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Anaplastic Lymphoma Kinase , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Crizotinib , Humans , Lung Neoplasms , Mutation , Pyrimidines/pharmacology , Receptor Protein-Tyrosine Kinases/metabolism
7.
Bioorg Med Chem Lett ; 21(12): 3743-8, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21561767

ABSTRACT

Ponatinib (AP24534) was previously identified as a pan-BCR-ABL inhibitor that potently inhibits the T315I gatekeeper mutant, and has advanced into clinical development for the treatment of refractory or resistant CML. In this study, we explored a novel series of five and six membered monocycles as alternate hinge-binding templates to replace the 6,5-fused imidazopyridazine core of ponatinib. Like ponatinib, these monocycles are tethered to pendant toluanilides via an ethynyl linker. Several compounds in this series displayed excellent in vitro potency against both native BCR-ABL and the T315I mutant. Notably, a subset of inhibitors exhibited desirable PK and were orally active in a mouse model of T315I-driven CML.


Subject(s)
Alkynes/chemical synthesis , Alkynes/pharmacology , Aniline Compounds/chemical synthesis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Toluene/chemical synthesis , Administration, Oral , Alkynes/chemistry , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Cyclization , Disease Models, Animal , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Models, Molecular , Molecular Structure , Mutation , Rats , Structure-Activity Relationship , Toluene/chemistry , Toluene/pharmacology
8.
Mol Cancer Ther ; 10(6): 1059-71, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21482695

ABSTRACT

The mTOR pathway is hyperactivated through oncogenic transformation in many human malignancies. Ridaforolimus (AP23573; MK-8669) is a novel rapamycin analogue that selectively targets mTOR and is currently under clinical evaluation. In this study, we investigated the mechanistic basis for the antitumor activity of ridaforolimus in a range of human tumor types, exploring potential markers of response, and determining optimal dosing regimens to guide clinical studies. Administration of ridaforolimus to tumor cells in vitro elicited dose-dependent inhibition of mTOR activity with concomitant effects on cell growth and division. We showed that ridaforolimus exhibits a predominantly cytostatic mode of action, consistent with the findings for other mTOR inhibitors. Potent inhibitory effects on vascular endothelial growth factor secretion, endothelial cell growth, and glucose metabolism were also observed. Although PTEN and/or phosphorylated AKT status have been proposed as potential mTOR pathway biomarkers, neither was predictive for ridaforolimus responsiveness in the heterogeneous panel of cancer cell lines examined. In mouse models, robust antitumor activity was observed in human tumor xenografts using a series of intermittent dosing schedules, consistent with pharmacodynamic observations of mTOR pathway inhibition for at least 72 hours following dosing. Parallel skin-graft rejection studies established that intermittent dosing schedules lack the immunosuppressive effects seen with daily dosing. Overall these findings show the broad inhibitory effects of ridaforolimus on cell growth, division, metabolism, and angiogenesis, and support the use of intermittent dosing as a means to optimize antitumor activity while minimizing systemic effects.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Sirolimus/analogs & derivatives , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antibiotics, Antineoplastic/administration & dosage , Cell Growth Processes/drug effects , Cell Line, Tumor , Endothelial Cells/drug effects , Female , Glucose/metabolism , HCT116 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Oncogene Protein v-akt/metabolism , PTEN Phosphohydrolase/metabolism , Phosphorylation/drug effects , Sirolimus/administration & dosage , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/metabolism , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
9.
J Med Chem ; 53(12): 4701-19, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20513156

ABSTRACT

In the treatment of chronic myeloid leukemia (CML) with BCR-ABL kinase inhibitors, the T315I gatekeeper mutant has emerged as resistant to all currently approved agents. This report describes the structure-guided design of a novel series of potent pan-inhibitors of BCR-ABL, including the T315I mutation. A key structural feature is the carbon-carbon triple bond linker which skirts the increased bulk of Ile315 side chain. Extensive SAR studies led to the discovery of development candidate 20g (AP24534), which inhibited the kinase activity of both native BCR-ABL and the T315I mutant with low nM IC(50)s, and potently inhibited proliferation of corresponding Ba/F3-derived cell lines. Daily oral administration of 20g significantly prolonged survival of mice injected intravenously with BCR-ABL(T315I) expressing Ba/F3 cells. These data, coupled with a favorable ADME profile, support the potential of 20g to be an effective treatment for CML, including patients refractory to all currently approved therapies.


Subject(s)
Antineoplastic Agents/chemical synthesis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Pyridazines/chemical synthesis , Administration, Oral , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Blood-Brain Barrier/metabolism , Cell Line, Tumor , Crystallography, X-Ray , Drug Screening Assays, Antitumor , Fusion Proteins, bcr-abl/genetics , Imidazoles/pharmacokinetics , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality , Mice , Mice, SCID , Models, Molecular , Mutation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacokinetics , Pyridazines/pharmacology , Rats
10.
Cancer Cell ; 16(5): 401-12, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19878872

ABSTRACT

Inhibition of BCR-ABL by imatinib induces durable responses in many patients with chronic myeloid leukemia (CML), but resistance attributable to kinase domain mutations can lead to relapse and a switch to second-line therapy with nilotinib or dasatinib. Despite three approved therapeutic options, the cross-resistant BCR-ABL(T315I) mutation and compound mutants selected on sequential inhibitor therapy remain major clinical challenges. We report design and preclinical evaluation of AP24534, a potent, orally available multitargeted kinase inhibitor active against T315I and other BCR-ABL mutants. AP24534 inhibited all tested BCR-ABL mutants in cellular and biochemical assays, suppressed BCR-ABL(T315I)-driven tumor growth in mice, and completely abrogated resistance in cell-based mutagenesis screens. Our work supports clinical evaluation of AP24534 as a pan-BCR-ABL inhibitor for treatment of CML.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Pyridazines/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Growth Processes/drug effects , Cell Line, Tumor , Crystallography, X-Ray , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imidazoles/chemistry , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/enzymology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, SCID , Models, Molecular , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Proto-Oncogene Proteins c-abl/chemistry , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Pyridazines/chemistry , Signal Transduction/drug effects
11.
J Med Chem ; 52(15): 4743-56, 2009 Aug 13.
Article in English | MEDLINE | ID: mdl-19572547

ABSTRACT

A novel series of potent dual Src/Abl kinase inhibitors based on a 9-(arenethenyl)purine core has been identified. Unlike traditional dual Src/Abl inhibitors targeting the active enzyme conformation, these inhibitors bind to the inactive, DFG-out conformation of both kinases. Extensive SAR studies led to the discovery of potent and orally bioavailable inhibitors, some of which demonstrated in vivo efficacy. Once-daily oral administration of inhibitor 9i (AP24226) significantly prolonged the survival of mice injected intravenously with wild type Bcr-Abl expressing Ba/F3 cells at a dose of 10 mg/kg. In a separate model, oral administration of 9i to mice bearing subcutaneous xenografts of Src Y527F expressing NIH 3T3 cells elicited dose-dependent tumor shrinkage with complete tumor regression observed at the highest dose. Notably, several inhibitors (e.g., 14a, AP24163) exhibited modest cellular potency (IC50 = 300-400 nM) against the Bcr-Abl mutant T315I, a variant resistant to all currently marketed therapies for chronic myeloid leukemia.


Subject(s)
Drug Design , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Purines/chemical synthesis , src-Family Kinases/antagonists & inhibitors , Animals , Female , Humans , K562 Cells , Mice , NIH 3T3 Cells , Protein Conformation , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/chemistry , Purines/pharmacology , Rats , Structure-Activity Relationship , src-Family Kinases/chemistry
12.
Bioorg Med Chem Lett ; 18(17): 4907-12, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18691885

ABSTRACT

Novel N(9)-arenethenyl purines, optimized potent dual Src/Abl tyrosine kinase inhibitors, are described. The key structural feature is a trans vinyl linkage at N(9) on the purine core which projects hydrophobic substituents into the selectivity pocket at the rear of the ATP site. Their synthesis was achieved through a Horner-Wadsworth-Emmons reaction of N(9)-phosphorylmethylpurines and substituted benzaldehydes or Heck reactions between 9-vinyl purines and aryl halides. Most compounds are potent inhibitors of both Src and Abl kinase, and several possess good oral bioavailability.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-abl/antagonists & inhibitors , Purines/chemistry , Purines/pharmacology , src-Family Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Growth Inhibitors/chemistry , Growth Inhibitors/pharmacology , Humans , K562 Cells , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-abl/physiology , Rats
13.
Toxicol Appl Pharmacol ; 218(3): 256-64, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17222881

ABSTRACT

Alveolar macrophages (AMs) primed with LPS and treated with concentrated ambient air particles (CAPs) showed enhanced release of tumor necrosis factor (TNF) and provide an in vitro model for the amplified effects of air pollution particles seen in people with preexisting lung disease. To investigate the mechanism(s) by which CAPs mediate TNF release in primed rat AMs, we first tested the effect of a panel of antioxidants. N-Acetyl-l-cysteine (20 mM), dimethyl thiourea (20 mM) and catalase (5 microM) significantly inhibited TNF release by primed AMs incubated with CAPs. Conversely, when LPS-primed AMs were treated with CAPs in the presence of exogenous oxidants (H(2)O(2) generated by glucose oxidase, 10 microM/h), TNF release and cell toxicity was significantly increased. The soluble fraction of CAPs suspensions caused most of the increased bioactivity in the presence of exogenous H(2)O(2). The metal chelator deferoxamine (DFO) strongly inhibited the interaction of the soluble fraction with H(2)O(2) but had no effect on the bioactivity of the insoluble CAPs fraction. We conclude that CAPs can mediate their effects in primed AMs by acting on oxidant-sensitive cytokine release in at least two distinct ways. In the primed cell, insoluble components of PM mediate enhanced TNF production that is H(2)O(2)-dependent (catalase-sensitive) yet independent of iron (DFO-insensitive). In the presence of exogenous H(2)O(2) released by AMs, PMNs, or other lung cells within an inflamed alveolar milieu, soluble iron released from air particles can also mediate cytokine release and cell toxicity.


Subject(s)
Air Pollutants/toxicity , Macrophages, Alveolar/drug effects , Particulate Matter/toxicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Antioxidants/pharmacology , Catalase/pharmacology , Cell Survival/drug effects , Cells, Cultured , Deferoxamine/pharmacology , Drug Combinations , Female , Hydrogen Peroxide/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Oxidative Stress/drug effects , Particle Size , Rats , Rats, Inbred Strains , Tumor Necrosis Factor-alpha/antagonists & inhibitors
14.
Am J Respir Cell Mol Biol ; 35(4): 474-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16675784

ABSTRACT

The class A macrophage scavenger receptor SR-AI/II is implicated as a pattern recognition receptor for innate immunity, but its functional role in lung defense has not been studied. We used mice genetically deficient in SR-AI/II and their wild-type C57BL/6 counterparts to investigate the contribution of this receptor to defense against pneumococcal infection and inhaled particles. SR-AI/II deficiency caused impaired phagocytosis of fluorescent bacteria in vivo, diminished clearance of live bacteria from the lungs, and substantially increased pneumonic inflammation. Survival studies also showed increased mortality in SR-AI/II-deficient mice with pneumococcal lung infection. Similarly, after challenge of the airways with TiO(2) particles, SR-AI/II-deficient mice showed increased proinflammatory cytokine levels in lung lavage fluid and a more pronounced neutrophilic inflammation. The data indicate that the lung macrophage class A scavenger receptor SR-AI/II contributes to innate defense against bacteria and inhaled particles.


Subject(s)
Immunity, Innate , Lung/immunology , Pneumonia, Pneumococcal/immunology , Scavenger Receptors, Class A/physiology , Animals , Disease Susceptibility , Female , Lung/metabolism , Lung/microbiology , Macrophages, Alveolar/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis , Pneumonia, Pneumococcal/mortality , Scavenger Receptors, Class A/genetics , Titanium/pharmacology
15.
J Exp Med ; 200(2): 267-72, 2004 Jul 19.
Article in English | MEDLINE | ID: mdl-15263032

ABSTRACT

Alveolar macrophages (AMs) express the class A scavenger receptor macrophage receptor with collagenous structure (MARCO), but its role in vivo in lung defense against bacteria and environmental particles has not been studied. We used MARCO-deficient mice to directly test the in vivo role of AM MARCO in innate defense against pneumococcal infection and environmental particles. In a murine model of pneumococcal pneumonia, MARCO(-/-) mice displayed an impaired ability to clear bacteria from the lungs, increased pulmonary inflammation and cytokine release, and diminished survival. In vitro binding of Streptococcus pneumoniae and in vivo uptake of unopsonized particles by MARCO(-/-) AMs were dramatically impaired. MARCO(-/-) mice treated with the "inert" environmental particle TiO(2) showed enhanced inflammation and chemokine expression, indicating that MARCO-mediated clearance of inert particles by AMs prevents inflammatory responses otherwise initiated by other lung cells. Our findings point to an important role of MARCO in mounting an efficient and appropriately regulated innate immune response against inhaled particles and airborne pathogens.


Subject(s)
Lung/immunology , Macrophages, Alveolar/immunology , Pneumonia, Pneumococcal/immunology , Receptors, Immunologic/metabolism , Animals , Bronchoalveolar Lavage , Bronchoalveolar Lavage Fluid , Cell Survival , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Immunity, Innate , Inflammation , Lung/cytology , Lung/microbiology , Lung/pathology , Macrophages/pathology , Macrophages, Alveolar/microbiology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phagocytosis , Pneumonia, Pneumococcal/microbiology , Protein Binding , Streptococcus pneumoniae/metabolism , Time Factors , Titanium/pharmacology
16.
Am J Respir Cell Mol Biol ; 30(5): 744-50, 2004 May.
Article in English | MEDLINE | ID: mdl-14630611

ABSTRACT

Exposure to ambient air pollution particles causes greater health effects in individuals with preexisting inflammatory lung diseases. To model inflammatory priming in vitro, HTB54 lung epithelial cells were pretreated with tumor necrosis factor-alpha (TNF-alpha) and then exposed to a panel of environmental particles, including concentrated ambient particles (CAPs). TNF-alpha priming significantly enhanced interleukin (IL)-8 secretion in response to CAPs and other urban air particles in HTB54 cells. Enhancement was seen with whole CAP suspensions as well as with its separate water-soluble and -insoluble components. Treating CAP suspensions with 20 microM deferoxamine or 2 mM dimethylthiourea attenuated the enhancement, indicating that transition metals and oxidative stress participate in the CAPs-dependent IL-8 response of primed cells. Because activated neutrophils are also present in diseased lungs and are sources of additional oxidative stress on epithelial cells, primed HTB54 cells were cocultured with activated neutrophils. Wild-type neutrophils markedly enhanced IL-8 release to CAPs in primed HTB54 cells, an effect substantially diminished when neutrophils from NADPH knockout mice were used. Cytokine priming and interactions with activated neutrophils can amplify lung epithelial inflammatory responses to ambient air particles.


Subject(s)
Air Pollutants/pharmacology , Epithelial Cells/drug effects , Interleukin-8/metabolism , Neutrophil Activation , Neutrophils/metabolism , Tumor Necrosis Factor-alpha/metabolism , Air Pollutants/metabolism , Animals , Antioxidants/pharmacology , Cell Line, Tumor , Cells, Cultured , Coculture Techniques , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Inhalation Exposure , Interleukin-8/immunology , Macrophages, Alveolar/metabolism , Metals/pharmacology , Mice , Mice, Knockout , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophil Activation/immunology , Neutrophils/cytology , Neutrophils/immunology , Oxidants/pharmacology , Oxidative Stress , Particle Size , Rats , Tumor Necrosis Factor-alpha/immunology
17.
Inhal Toxicol ; 14(4): 325-47, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12028808

ABSTRACT

We investigated whether coexposure to inhaled ambient particles and ozone affects airway responsiveness (AR, measured as enhanced pause, Penh) and allergic inflammation (AI) in a murine model of asthma. Ovalbumin-sensitized mice were challenged with either ovalbumin ("asthmatic") or phosphate-buffered saline (PBS) aerosols for 3 successive days. Immediately after daily challenge, mice were exposed for 5 h to concentrated ambient particles (CAPs), or 0.3 ppm ozone, or both, or neither (n > or = 61/group, 12 experiments). Exposure to CAPs alone or coexposure to CAPs + O(3) caused an increase in Penh in both normal and "asthmatic" mice. These responses were transient and small, increasing approximately 0.9% per 100-microg/m(3) increase in CAPs. Analysis of the effects of particle composition on AR revealed an association between the AlSi particle fraction and increased AR in "asthmatic" mice exposed to ozone and particles. No effects of pollutants on AI were noted. We conclude that (1) particle exposure causes an immediate, short-lived (<24 h) increase in AR in mice; (2) these responses are small; and (3) changes in AR may be correlated with specific elements within the particle mixture.


Subject(s)
Air Pollutants/adverse effects , Asthma/physiopathology , Environmental Exposure , Lung/pathology , Oxidants, Photochemical/adverse effects , Ozone/adverse effects , Airway Resistance/drug effects , Animals , Disease Models, Animal , Drug Interactions , Inflammation , Particle Size
SELECTION OF CITATIONS
SEARCH DETAIL
...