Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Sci Transl Med ; 13(609): eabb3738, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34516823

ABSTRACT

The clinical efficacy of epidermal growth factor receptor (EGFR)­targeted therapy in EGFR-mutant non­small cell lung cancer is limited by the development of drug resistance. One mechanism of EGFR inhibitor resistance occurs through amplification of the human growth factor receptor (MET) proto-oncogene, which bypasses EGFR to reactivate downstream signaling. Tumors exhibiting concurrent EGFR mutation and MET amplification are historically thought to be codependent on the activation of both oncogenes. Hence, patients whose tumors harbor both alterations are commonly treated with a combination of EGFR and MET tyrosine kinase inhibitors (TKIs). Here, we identify and characterize six patient-derived models of EGFR-mutant, MET-amplified lung cancer that have switched oncogene dependence to rely exclusively on MET activation for survival. We demonstrate in this MET-driven subset of EGFR TKI-refractory cancers that canonical EGFR downstream signaling was governed by MET, even in the presence of sustained mutant EGFR expression and activation. In these models, combined EGFR and MET inhibition did not result in greater efficacy in vitro or in vivo compared to single-agent MET inhibition. We further identified a reduced EGFR:MET mRNA expression stoichiometry as associated with MET oncogene dependence and single-agent MET TKI sensitivity. Tumors from 10 of 11 EGFR inhibitor­resistant EGFR-mutant, MET-amplified patients also exhibited a reduced EGFR:MET mRNA ratio. Our findings reveal that a subset of EGFR-mutant, MET-amplified lung cancers develop dependence on MET activation alone, suggesting that such patients could be treated with a single-agent MET TKI rather than the current standard-of-care EGFR and MET inhibitor combination regimens.


Subject(s)
ErbB Receptors , Lung Neoplasms , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , ErbB Receptors/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mutation/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
2.
Br J Pharmacol ; 178(3): 600-613, 2021 02.
Article in English | MEDLINE | ID: mdl-33125717

ABSTRACT

BACKGROUND AND PURPOSE: Savolitinib (AZD6094, HMPL-504, volitinib) is an oral, potent, and highly MET receptor TK inhibitor. This series of studies aimed to develop a pharmacokinetic-pharmacodynamic (PK/PD) model to link inhibition of MET phosphorylation (pMET) by savolitinib with anti-tumour activity. EXPERIMENTAL APPROACH: Cell line-derived xenograft (CDX) experiments using human lung cancer (EBC-1) and gastric cancer (MKN-45) cells were conducted in athymic nude mice using a variety of doses and schedules of savolitinib. Tumour pMET changes and growth inhibition were calculated after 28 days. Population PK/PD techniques were used to construct a PK/PD model for savolitinib. KEY RESULTS: Savolitinib showed dose- and dose frequency-dependent anti-tumour activity in the CDX models, with more frequent, lower dosing schedules (e.g., twice daily) being more effective than intermittent, higher dosing schedules (e.g., 4 days on/3 days off or 2 days on/5 days off). There was a clear exposure-response relationship, with maximal suppression of pMET of >90%. Data from additional CDX and patient-derived xenograft (PDX) models overlapped, allowing calculation of a single EC50 of 0.38 ng·ml-1 . Tumour growth modelling demonstrated that prolonged, high levels of pMET inhibition (>90%) were required for tumour stasis and regression in the models. CONCLUSION AND IMPLICATIONS: High and persistent levels of MET inhibition by savolitinib were needed for optimal monotherapy anti-tumour activity in preclinical models. The modelling framework developed here can be used to translate tumour growth inhibition from the mouse to human and thus guide choice of clinical dose and schedule.


Subject(s)
Antineoplastic Agents , Proto-Oncogene Proteins c-met , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Mice , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Pyrazines , Triazines , Xenograft Model Antitumor Assays
3.
Bioorg Med Chem ; 28(23): 115815, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33091850

ABSTRACT

In this article, we report our efforts towards improving in vitro human clearance in a series of 5-azaquinazolines through a series of C4 truncations and C2 expansions. Extensive DMPK studies enabled us to tackle high Aldehyde Oxidase (AO) metabolism and unexpected discrepancies in human hepatocyte and liver microsomal intrinsic clearance. Our efforts culminated with the discovery of 5-azaquinazoline 35, which also displayed exquisite selectivity for IRAK4, and showed synergistic in vitro activity against MyD88/CD79 double mutant ABC-DLBCL in combination with the covalent BTK inhibitor acalabrutinib.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/metabolism , Quinazolines/chemistry , Aldehyde Oxidase/metabolism , Animals , Binding Sites , Cell Line, Tumor , Cell Survival/drug effects , Crystallography, X-Ray , Dogs , Drug Stability , Half-Life , Hepatocytes/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Mice , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Quinazolines/metabolism , Quinazolines/pharmacology , Rats , Structure-Activity Relationship
4.
PLoS One ; 15(7): e0236192, 2020.
Article in English | MEDLINE | ID: mdl-32692756

ABSTRACT

Breast cancer (BC) is the foremost cause of cancer related deaths in women globally. Currently there is a scarcity of reliable biomarkers for its early stage diagnosis and theranostics monitoring. Altered DNA methylation patterns leading to the silencing of tumor suppressor genes are considered as an important mechanism underlying tumor development and progression in various cancer types, including BC. Very recently, epigenetic silencing of SHISA3, an antagonist of ß-catenin, has been reported in various types of tumor. However, the role of SHISA3 in BC has not been investigated yet. Therefore, we aimed at evaluating the contribution of SHISA3 in BC causation by analyzing its expression and methylation levels in BC cell lines (MDA-MB231, MCF-7 and BT-474) and in 103 paired BC tissue samples. The SHISA3 expression and methylation status was determined by qPCR and methylation specific PCR (MSP) respectively. The role of SHISA3 in BC tumorigenesis was evaluated by proliferation and migration assays after ectopic expression of SHISA3. The association between SHISA3 hypermethylation and clinicopathological parameters of BC patients was also studied. The downregulation of SHISA3 expression was found in three BC cell lines used and in all BC tissue samples. However, SHISA3 promoter region was hypermethylated in 61% (63/103) tumorous tissues in comparison to the 18% of their matched normal tissues. The 5-aza-2'-deoxycytidine treatment restored SHISA3 expression by reversing promoter hypermethylation in both MDA-MB231 and MCF-7 cells. Furthermore, ectopic expression of SHISA3 significantly reduced the proliferation and migration ability of these cells. Taken together, our findings for the first time reveal epigenetic silencing and tumor suppressing role of SHISA3 in BC. Henceforth, this study has identified SHISA3 as potentially powerful target for the development of new therapies against BC, as well as novel diagnostic and therapy response monitoring approaches.


Subject(s)
Breast Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Wnt Signaling Pathway/genetics , Azacitidine/pharmacology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , DNA Methylation/genetics , Female , Humans , Membrane Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
Sci Transl Med ; 12(541)2020 04 29.
Article in English | MEDLINE | ID: mdl-32350132

ABSTRACT

Gastrointestinal stromal tumor (GIST) is the most common human sarcoma driven by mutations in KIT or platelet-derived growth factor α (PDGFRα). Although first-line treatment, imatinib, has revolutionized GIST treatment, drug resistance due to acquisition of secondary KIT/PDGFRα mutations develops in a majority of patients. Second- and third-line treatments, sunitinib and regorafenib, lack activity against a plethora of mutations in KIT/PDGFRα in GIST, with median time to disease progression of 4 to 6 months and inhibition of vascular endothelial growth factor receptor 2 (VEGFR2) causing high-grade hypertension. Patients with GIST have an unmet need for a well-tolerated drug that robustly inhibits a range of KIT/PDGFRα mutations. Here, we report the discovery and pharmacological characterization of AZD3229, a potent and selective small-molecule inhibitor of KIT and PDGFRα designed to inhibit a broad range of primary and imatinib-resistant secondary mutations seen in GIST. In engineered and GIST-derived cell lines, AZD3229 is 15 to 60 times more potent than imatinib in inhibiting KIT primary mutations and has low nanomolar activity against a wide spectrum of secondary mutations. AZD3229 causes durable inhibition of KIT signaling in patient-derived xenograft (PDX) models of GIST, leading to tumor regressions at doses that showed no changes in arterial blood pressure (BP) in rat telemetry studies. AZD3229 has a superior potency and selectivity profile to standard of care (SoC) agents-imatinib, sunitinib, and regorafenib, as well as investigational agents, avapritinib (BLU-285) and ripretinib (DCC-2618). AZD3229 has the potential to be a best-in-class inhibitor for clinically relevant KIT/PDGFRα mutations in GIST.


Subject(s)
Antineoplastic Agents , Gastrointestinal Stromal Tumors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/genetics , Humans , Mutation , Naphthyridines , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/genetics , Pyrazoles , Pyrroles , Rats , Receptor, Platelet-Derived Growth Factor alpha/genetics , Triazines , Urea/analogs & derivatives , Vascular Endothelial Growth Factor A
6.
Clin Cancer Res ; 26(14): 3751-3759, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32220888

ABSTRACT

PURPOSE: The emergence of secondary mutations is a cause of resistance to current KIT inhibitors used in the treatment of patients with gastrointestinal stromal tumors (GIST). AZD3229 is a selective inhibitor of wild-type KIT and a wide spectrum of primary and secondary mutations seen in patients with GIST. The objective of this analysis is to establish the pharmacokinetic-pharmacodynamic (PKPD) relationship of AZD3229 in a range of mouse GIST tumor models harboring primary and secondary KIT mutations, and to benchmark AZD3229 against other KIT inhibitors. EXPERIMENTAL DESIGN: A PKPD model was developed for AZD3229 linking plasma concentrations to inhibition of phosphorylated KIT using data generated from several in vivo preclinical tumor models, and in vitro data generated in a panel of Ba/F3 cell lines. RESULTS: AZD3229 drives inhibition of phosphorylated KIT in an exposure-dependent manner, and optimal efficacy is observed when >90% inhibition of KIT phosphorylation is sustained over the dosing interval. Integrating the predicted human pharmacokinetics into the mouse PKPD model predicts that an oral twice daily human dose greater than 34 mg is required to ensure adequate coverage across the mutations investigated. Benchmarking shows that compared with standard-of-care KIT inhibitors, AZD3229 has the potential to deliver the required target coverage across a wider spectrum of primary or secondary mutations. CONCLUSIONS: We demonstrate that AZD3229 warrants clinical investigation as a new treatment for patients with GIST based on its ability to inhibit both ATP-binding and A-loop mutations of KIT at clinically relevant exposures.


Subject(s)
Gastrointestinal Stromal Tumors/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Quinazolines/pharmacology , Triazoles/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Gastrointestinal Stromal Tumors/pathology , Humans , Mice , Models, Biological , Mutation , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Quinazolines/therapeutic use , Receptor, Platelet-Derived Growth Factor alpha/genetics , Triazoles/therapeutic use , Xenograft Model Antitumor Assays
7.
J Med Chem ; 62(21): 9918-9930, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31622099

ABSTRACT

In this article, we report the discovery of a series of 5-azaquinazolines as selective IRAK4 inhibitors. From modestly potent quinazoline 4, we introduced a 5-aza substitution to mask the 4-NH hydrogen bond donor (HBD). This allowed us to substitute the core with a 2-aminopyrazole, which showed large gains in cellular potency despite the additional formal HBD. Further optimization led to 6-cyanomethyl-5-azaquinazoline 13, a selective IRAK4 inhibitor, which proved efficacious in combination with ibrutinib, while showing very little activity as a single agent up to 100 mg/kg. This contrasted to previously reported IRAK4 inhibitors that exhibited efficacy in the same model as single agents and was attributed to the enhanced specificity of 13 toward IRAK4.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Molecular Targeted Therapy , Myeloid Differentiation Factor 88/genetics , Quinazolines/chemistry , Quinazolines/pharmacology , Administration, Oral , Animals , Cell Line, Tumor , Drug Design , Female , Humans , Interleukin-1 Receptor-Associated Kinases/chemistry , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Mice , Models, Molecular , Mutation , Protein Conformation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Quinazolines/administration & dosage , Quinazolines/pharmacokinetics , Rats , Rats, Wistar , Structure-Activity Relationship , Tissue Distribution , Xenograft Model Antitumor Assays
8.
J Med Chem ; 61(19): 8797-8810, 2018 10 11.
Article in English | MEDLINE | ID: mdl-30204441

ABSTRACT

While the treatment of gastrointestinal stromal tumors (GISTs) has been revolutionized by the application of targeted tyrosine kinase inhibitors capable of inhibiting KIT-driven proliferation, diverse mutations to this kinase drive resistance to established therapies. Here we describe the identification of potent pan-KIT mutant kinase inhibitors that can be dosed without being limited by the tolerability issues seen with multitargeted agents. This effort focused on identification and optimization of an existing kinase scaffold through the use of structure-based design. Starting from a series of previously reported phenoxyquinazoline and quinoline based inhibitors of the tyrosine kinase PDGFRα, potency against a diverse panel of mutant KIT driven Ba/F3 cell lines was optimized, with a particular focus on reducing activity against a KDR driven cell model in order to limit the potential for hypertension commonly seen in second and third line GIST therapies. AZD3229 demonstrates potent single digit nM growth inhibition across a broad cell panel, with good margin to KDR-driven effects. Selectivity over KDR can be rationalized predominantly by the interaction of water molecules with the protein and ligand in the active site, and its kinome selectivity is similar to the best of the approved GIST agents. This compound demonstrates excellent cross-species pharmacokinetics, shows strong pharmacodynamic inhibition of target, and is active in several in vivo models of GIST.


Subject(s)
Drug Discovery , Gastrointestinal Stromal Tumors/drug therapy , Mutant Proteins/antagonists & inhibitors , Mutation , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Quinazolines/chemistry , Quinazolines/pharmacology , Triazoles/chemistry , Triazoles/pharmacology , Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/metabolism , Gastrointestinal Stromal Tumors/pathology , Humans , Models, Molecular , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Conformation , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , Quinazolines/pharmacokinetics , Tissue Distribution , Triazoles/pharmacokinetics , Tumor Cells, Cultured
9.
Bioorg Med Chem ; 26(4): 913-924, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29398441

ABSTRACT

We have developed a series of orally efficacious IRAK4 inhibitors, based on a scaffold hopping strategy and using rational structure based design. Efforts to tackle low permeability and high efflux in our previously reported pyrrolopyrimidine series (Scott et al., 2017) led to the identification of pyrrolotriazines which contained one less formal hydrogen bond donor and were intrinsically more lipophilic. Further optimisation of substituents on this pyrrolotriazine core culminated with the discovery of 30 as a promising in vivo probe to assess the potential of IRAK4 inhibition for the treatment of MyD88 mutant DLBCL in combination with a BTK inhibitor. When tested in an ABC-DLBCL model with a dual MyD88/CD79 mutation (OCI-LY10), 30 demonstrated tumour regressions in combination with ibrutinib.


Subject(s)
Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Pyrroles/chemistry , Thiazines/chemistry , Animals , Binding Sites , Caco-2 Cells , Dogs , Drug Design , Half-Life , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Interleukin-1 Receptor-Associated Kinases/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/pathology , Molecular Dynamics Simulation , Mutation , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Permeability/drug effects , Protein Kinases/chemistry , Protein Kinases/metabolism , Pyrroles/pharmacokinetics , Pyrroles/pharmacology , Rats , Structure-Activity Relationship , Thiazines/pharmacokinetics , Thiazines/pharmacology
10.
J Med Chem ; 60(24): 10071-10091, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29172502

ABSTRACT

Herein we report the optimization of a series of pyrrolopyrimidine inhibitors of interleukin-1 receptor associated kinase 4 (IRAK4) using X-ray crystal structures and structure based design to identify and optimize our scaffold. Compound 28 demonstrated a favorable physicochemical and kinase selectivity profile and was identified as a promising in vivo tool with which to explore the role of IRAK4 inhibition in the treatment of mutant MYD88L265P diffuse large B-cell lymphoma (DLBCL). Compound 28 was shown to be capable of demonstrating inhibition of NF-κB activation and growth of the ABC subtype of DLBCL cell lines in vitro at high concentrations but showed greater effects in combination with a BTK inhibitor at lower concentrations. In vivo, the combination of compound 28 and ibrutinib led to tumor regression in an ABC-DLBCL mouse model.


Subject(s)
Antineoplastic Agents/pharmacology , Interleukin-1 Receptor-Associated Kinases/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/drug therapy , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Crystallography, X-Ray , Dogs , Female , Humans , Interleukin-1 Receptor-Associated Kinases/chemistry , Lymphoma, Large B-Cell, Diffuse/genetics , Magnetic Resonance Spectroscopy , Male , Mice, SCID , Mutation , Myeloid Differentiation Factor 88/genetics , Protein Kinase Inhibitors/administration & dosage , Pyrimidines/chemistry , Pyrroles/chemistry , Rats, Wistar , Structure-Activity Relationship , Xenograft Model Antitumor Assays
11.
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
12.
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
13.
Nat Commun ; 6: 8163, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26348592

ABSTRACT

In eukaryotes, the covalent attachment of ubiquitin chains directs substrates to the proteasome for degradation. Recently, ubiquitin-like modifications have also been described in the archaeal domain of life. It has subsequently been hypothesized that ubiquitin-like proteasomal degradation might also operate in these microbes, since all archaeal species utilize homologues of the eukaryotic proteasome. Here we perform a structural and biochemical analysis of a ubiquitin-like modification pathway in the archaeon Sulfolobus acidocaldarius. We reveal that this modifier is homologous to the eukaryotic ubiquitin-related modifier Urm1, considered to be a close evolutionary relative of the progenitor of all ubiquitin-like proteins. Furthermore we demonstrate that urmylated substrates are recognized and processed by the archaeal proteasome, by virtue of a direct interaction with the modifier. Thus, the regulation of protein stability by Urm1 and the proteasome in archaea is likely representative of an ancient pathway from which eukaryotic ubiquitin-mediated proteolysis has evolved.


Subject(s)
Archaeal Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Sulfolobus acidocaldarius/genetics , Ubiquitins/genetics , Archaeal Proteins/metabolism , Chromatography, Gel , Chromatography, Liquid , Circular Dichroism , Crystallography, X-Ray , Mass Spectrometry , Microscopy, Electron , Proteasome Endopeptidase Complex/ultrastructure , Proteolysis , Sulfolobus acidocaldarius/metabolism , Ubiquitins/metabolism
14.
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
15.
PLoS Genet ; 9(9): e1003749, 2013.
Article in English | MEDLINE | ID: mdl-24086144

ABSTRACT

Circadian rhythms in Drosophila rely on cyclic regulation of the period (per) and timeless (tim) clock genes. The molecular cycle requires rhythmic phosphorylation of PER and TIM proteins, which is mediated by several kinases and phosphatases such as Protein Phosphatase-2A (PP2A) and Protein Phosphatase-1 (PP1). Here, we used mass spectrometry to identify 35 "phospho-occupied" serine/threonine residues within PER, 24 of which are specifically regulated by PP1/PP2A. We found that cell culture assays were not good predictors of protein function in flies and so we generated per transgenes carrying phosphorylation site mutations and tested for rescue of the per(01) arrhythmic phenotype. Surprisingly, most transgenes restore wild type rhythms despite carrying mutations in several phosphorylation sites. One particular transgene, in which T610 and S613 are mutated to alanine, restores daily rhythmicity, but dramatically lengthens the period to ~ 30 hrs. Interestingly, the single S613A mutation extends the period by 2-3 hours, while the single T610A mutation has a minimal effect, suggesting these phospho-residues cooperate to control period length. Conservation of S613 from flies to humans suggests that it possesses a critical clock function, and mutational analysis of residues surrounding T610/S613 implicates the entire region in determining circadian period. Biochemical and immunohistochemical data indicate defects in overall phosphorylation and altered timely degradation of PER carrying the double or single S613A mutation(s). The PER-T610A/S613A mutant also alters CLK phosphorylation and CLK-mediated output. Lastly, we show that a mutation at a previously identified site, S596, is largely epistatic to S613A, suggesting that S613 negatively regulates phosphorylation at S596. Together these data establish functional significance for a new domain of PER, demonstrate that cooperativity between phosphorylation sites maintains PER function, and support a model in which specific phosphorylated regions regulate others to control circadian period.


Subject(s)
Circadian Rhythm/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Period Circadian Proteins/genetics , Phosphorylation/genetics , Animals , Drosophila Proteins/physiology , Drosophila melanogaster/physiology , Humans , Mutation , Period Circadian Proteins/physiology , Phenotype , Protein Phosphatase 1/genetics , Protein Phosphatase 1/metabolism , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism
16.
FEBS Lett ; 586(19): 3471-6, 2012 Sep 21.
Article in English | MEDLINE | ID: mdl-22967899

ABSTRACT

N-alpha-tosyl-L-phenylalanyl chloromethyl ketone (TPCK) has anti-tumorigenic properties, but its direct cellular targets are unknown. Previously, we showed TPCK inhibited the PDKl-dependent AGC kinases RSK, Akt and S6K1 without inhibiting PKA, ERK1/2, PI3K, and PDK1 itself. Here we show TPCK-inhibition of the RSK-related kinases MSK1 and 2, which can be activated independently of PDK1. Mass spectrometry analysis of RSK1, Aktl, S6K1 and MSK1 immunopurified from TPCK-treated cells identified TPCK adducts on cysteines located in conserved activation loop Phenylalanine-Cysteine (Phe-Cys) motifs. Mutational analysis of the Phe-Cys residues conferred partial TPCK resistance. These studies elucidate a primary mechanism by which TPCK inhibits several AGC kinases, inviting consideration of TPCK-like compounds in chemotherapy given their potential for broad control of cellular growth, proliferation and survival.


Subject(s)
Protein Kinase Inhibitors/pharmacology , Protein Kinases/chemistry , Tosylphenylalanyl Chloromethyl Ketone/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Amino Acid Substitution , Base Sequence , Conserved Sequence , Cysteine/chemistry , DNA Primers/genetics , HEK293 Cells , Humans , Molecular Sequence Data , Mutagenesis, Site-Directed , Phenylalanine/chemistry , Protein Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/genetics , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Sequence Homology, Amino Acid
17.
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
18.
Mol Biol Cell ; 21(15): 2661-73, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20519438

ABSTRACT

Cytoplasmic linker protein (CLIP)-170 is a microtubule (MT) plus-end-tracking protein that regulates MT dynamics and links MT plus ends to different intracellular structures. We have shown previously that intramolecular association between the N and C termini results in autoinhibition of CLIP-170, thus altering its binding to MTs and the dynactin subunit p150(Glued) (J. Cell Biol. 2004: 166, 1003-1014). In this study, we demonstrate that conformational changes in CLIP-170 are regulated by phosphorylation that enhances the affinity between the N- and C-terminal domains. By using site-directed mutagenesis and phosphoproteomic analysis, we mapped the phosphorylation sites in the third serine-rich region of CLIP-170. A phosphorylation-deficient mutant of CLIP-170 displays an "open" conformation and a higher binding affinity for growing MT ends and p150(Glued) as compared with nonmutated protein, whereas a phosphomimetic mutant confined to the "folded back" conformation shows decreased MT association and does not interact with p150(Glued). We conclude that phosphorylation regulates CLIP-170 conformational changes resulting in its autoinhibition.


Subject(s)
Microtubule-Associated Proteins/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Dyneins/metabolism , Mice , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Microtubules/drug effects , Microtubules/metabolism , Models, Biological , Molecular Sequence Data , Mutation/genetics , Neoplasm Proteins/chemistry , Neoplasm Proteins/metabolism , Okadaic Acid/pharmacology , Phosphorylation/drug effects , Phosphoserine/metabolism , Protein Folding/drug effects , Protein Transport/drug effects , Proteome/metabolism
19.
Nat Biotechnol ; 27(10): 933-40, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19801977

ABSTRACT

Constitutive activation of one or more kinase signaling pathways is a hallmark of many cancers. Here we extend the previously described mass spectrometry-based KAYAK approach by monitoring kinase activities from multiple signaling pathways simultaneously. This improved single-reaction strategy, which quantifies the phosphorylation of 90 synthetic peptides in a single mass spectrometry run, is compatible with nanogram to microgram amounts of cell lysate. Furthermore, the approach enhances kinase monospecificity through substrate competition effects, faithfully reporting the signatures of many signaling pathways after mitogen stimulation or of basal pathway activation differences across a panel of well-studied cancer cell lines. Hierarchical clustering of activities from related experiments groups peptides phosphorylated by similar kinases together and, when combined with pathway alteration using pharmacological inhibitors, distinguishes underlying differences in potency, off-target effects and genetic backgrounds. Finally, we introduce a strategy to identify the kinase, and even associated protein complex members, responsible for phosphorylation events of interest.


Subject(s)
Computational Biology/methods , Mass Spectrometry/methods , Protein Kinases/metabolism , CDC2 Protein Kinase , Cell Cycle/drug effects , Cells, Cultured , Cluster Analysis , Cyclin B/metabolism , Cyclin-Dependent Kinases , Epidermal Growth Factor/pharmacology , HeLa Cells , Humans , Insulin/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Reproducibility of Results , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology
20.
Proc Natl Acad Sci U S A ; 106(28): 11606-11, 2009 Jul 14.
Article in English | MEDLINE | ID: mdl-19564600

ABSTRACT

Most kinases are capable of recognizing and phosphorylating peptides containing short, linear sequence motifs. To measure the activation state of many kinases from the same cell lysate, we created a multiplexed, mass-spectrometry-based in vitro kinase assay. Ninety chemically synthesized peptides derived from well-characterized peptide substrates and in vivo phosphorylation sites with either known or previously unidentified upstream kinases were reacted individually in a plate format with crude cell lysates and ATP. Phosphorylation rates were directly measured based on the addition of 90 same-sequence, site-specific phosphopeptides enriched in stable isotopes to act as ideal quantitative internal standards for analysis by liquid chromatography coupled to tandem mass spectrometry. This approach concurrently measured up to 90 site-specific peptide phosphorylation rates, reporting a diagnostic fingerprint for activated kinase pathways. We applied this unique kinome-activity profiling strategy in a variety of cellular settings, including mitogen stimulation, cell cycle, pharmacological inhibition of pathways, and to a panel of breast cancer cell lines. Finally, we identified the source of activity for a peptide (derived from a PI3K regulatory subunit) from our library. This peptide substrate demonstrated mitotic and tyrosine-specific phosphorylation, which was confirmed to be a novel Src family kinase site in vivo.


Subject(s)
Cell Cycle/physiology , Phosphopeptides/metabolism , Phosphotransferases/metabolism , Signal Transduction/physiology , Adenosine Triphosphate/metabolism , Cell Line, Tumor , Humans , In Vitro Techniques , Isotope Labeling , Mass Spectrometry , Phosphorylation
SELECTION OF CITATIONS
SEARCH DETAIL
...