Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
PLoS One ; 19(6): e0305162, 2024.
Article in English | MEDLINE | ID: mdl-38861568

ABSTRACT

Using a multilevel modelling approach to analyse a novel dataset of academic publications at all business schools in 11 European countries, this paper finds that the influence of organisational- and country-level contextual factors on researchers varies considerably based on the type of institution and the development level of the country they are located in. At the organisational-level, we find that greater spatial connectivity-operationalised through proximity to nearby business schools, rail stations, and airports-is positively related to scientific research volume and public dissemination (news mentions). While this result is significant only for high-income countries (above EU-average 2018 GDP per capita), this is likely because the low-income countries (below EU-average 2018 GDP per capita) examined here lack a 'critical mass' of well-connected universities to generate observable agglomeration effects. At the country-level, the results indicate that in high-income countries, less prestigious schools benefit from higher rates of recent international immigration from any foreign country, providing a direct policy pathway for increasing research output for universities that aren't already well-known enough to attract the most talented researchers. In low-income countries, recent immigration rates are even stronger predictors of research performance across all levels of institutional prestige; more open immigration policies would likely benefit research performance in these countries to an even greater extent. Finally, the paper's results show that, in low-income countries, a composite measure of a country's quality of life (including self-rated life satisfaction, health, working hours, and housing overcrowding) is positively related to research outcomes through its interaction with school prestige. This suggests that the lower a country's quality of life, the more researchers are incentivised to produce higher levels of research output. While this may in part reflect the greater disparities inherent in these countries' economic systems, it is noteworthy-and perhaps concerning-that we have observed a negative correlation between country-level quality of life and research performance in low-income countries, which is particularly felt by researchers at less prestigious institutions.


Subject(s)
Commerce , Humans , Europe , Universities , Research , Emigration and Immigration
2.
ChemMedChem ; 15(16): 1562-1570, 2020 08 19.
Article in English | MEDLINE | ID: mdl-32613743

ABSTRACT

Loss of ß-cell mass and function can lead to insufficient insulin levels and ultimately to hyperglycemia and diabetes mellitus. The mainstream treatment approach involves regulation of insulin levels; however, approaches intended to increase ß-cell mass are less developed. Promoting ß-cell proliferation with low-molecular-weight inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) offers the potential to treat diabetes with oral therapies by restoring ß-cell mass, insulin content and glycemic control. GNF4877, a potent dual inhibitor of DYRK1A and glycogen synthase kinase 3ß (GSK3ß) was previously reported to induce primary human ß-cell proliferation in vitro and in vivo. Herein, we describe the lead optimization that lead to the identification of GNF4877 from an aminopyrazine hit identified in a phenotypic high-throughput screening campaign measuring ß-cell proliferation.


Subject(s)
Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Mice , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Structure-Activity Relationship , Dyrk Kinases
3.
PLoS One ; 15(3): e0224344, 2020.
Article in English | MEDLINE | ID: mdl-32176701

ABSTRACT

A key event in the development of both major forms of diabetes is the loss of functional pancreatic islet ß-cell mass. Strategies aimed at enhancing ß-cell regeneration have long been pursued, but methods for reliably inducing human ß-cell proliferation with full retention of key functions such as glucose-stimulated insulin secretion (GSIS) are still very limited. We have previously reported that overexpression of the homeobox transcription factor NKX6.1 stimulates ß-cell proliferation, while also enhancing GSIS and providing protection against ß-cell cytotoxicity through induction of the VGF prohormone. We developed an NKX6.1 pathway screen by stably transfecting 832/13 rat insulinoma cells with a VGF promoter-luciferase reporter construct, using the resultant cell line to screen a 630,000 compound chemical library. We isolated three compounds with consistent effects to stimulate human islet cell proliferation, but not expression of NKX6.1 or VGF, suggesting an alternative mechanism of action. Further studies of the most potent of these compounds, GNF-9228, revealed that it selectively activates human ß-cell relative to α-cell proliferation and has no effect on δ-cell replication. In addition, pre-treatment, but not short term exposure of human islets to GNF-9228 enhances GSIS. GNF-9228 also protects 832/13 insulinoma cells against ER stress- and inflammatory cytokine-induced cytotoxicity. GNF-9228 stimulates proliferation via a mechanism distinct from recently emergent DYRK1A inhibitors, as it is unaffected by DYRK1A overexpression and does not activate NFAT translocation. In conclusion, we have identified a small molecule with pleiotropic positive effects on islet biology, including stimulation of human ß-cell proliferation and insulin secretion, and protection against multiple agents of cytotoxic stress.


Subject(s)
Cell Proliferation/drug effects , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Cell Line, Tumor , Glucagon-Secreting Cells/metabolism , Glucagon-Secreting Cells/pathology , Glucose/pharmacology , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/pathology , Insulinoma/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/metabolism , Rats , Dyrk Kinases
4.
J Med Chem ; 63(6): 2958-2973, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32077280

ABSTRACT

Autoimmune deficiency and destruction in either ß-cell mass or function can cause insufficient insulin levels and, as a result, hyperglycemia and diabetes. Thus, promoting ß-cell proliferation could be one approach toward diabetes intervention. In this report we describe the discovery of a potent and selective DYRK1A inhibitor GNF2133, which was identified through optimization of a 6-azaindole screening hit. In vitro, GNF2133 is able to proliferate both rodent and human ß-cells. In vivo, GNF2133 demonstrated significant dose-dependent glucose disposal capacity and insulin secretion in response to glucose-potentiated arginine-induced insulin secretion (GPAIS) challenge in rat insulin promoter and diphtheria toxin A (RIP-DTA) mice. The work described here provides new avenues to disease altering therapeutic interventions in the treatment of type 1 diabetes (T1D).


Subject(s)
Aza Compounds/chemistry , Aza Compounds/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Indoles/chemistry , Indoles/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Aza Compounds/pharmacokinetics , Cell Proliferation/drug effects , Cells, Cultured , Diabetes Mellitus, Type 1/metabolism , Humans , Hypoglycemic Agents/pharmacokinetics , Indoles/pharmacokinetics , Insulin Secretion/drug effects , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Molecular Docking Simulation , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Dyrk Kinases
5.
J Med Chem ; 63(8): 3868-3880, 2020 04 23.
Article in English | MEDLINE | ID: mdl-31940200

ABSTRACT

Farnesoid X receptor (FXR) agonists are emerging as important potential therapeutics for the treatment of nonalcoholic steatohepatitis (NASH) patients, as they exert positive effects on multiple aspects of the disease. FXR agonists reduce lipid accumulation in the liver, hepatocellular inflammation, hepatic injury, and fibrosis. While there are currently no approved therapies for NASH, the bile acid-derived FXR agonist obeticholic acid (OCA; 6-ethyl chenodeoxycholic acid) has shown promise in clinical studies. Previously, we described the discovery of tropifexor (LJN452), the most potent non-bile acid FXR agonist currently in clinical investigation. Here, we report the discovery of a novel chemical series of non-bile acid FXR agonists based on a tricyclic dihydrochromenopyrazole core from which emerged nidufexor (LMB763), a compound with partial FXR agonistic activity in vitro and FXR-dependent gene modulation in vivo. Nidufexor has advanced to Phase 2 human clinical trials in patients with NASH and diabetic nephropathy.


Subject(s)
Benzothiazoles/therapeutic use , Chenodeoxycholic Acid/analogs & derivatives , Diet, High-Fat/adverse effects , Isoxazoles/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Benzothiazoles/chemistry , Chenodeoxycholic Acid/chemistry , Chenodeoxycholic Acid/therapeutic use , Dogs , Humans , Isoxazoles/chemistry , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/etiology , Protein Structure, Tertiary , Rats , Treatment Outcome
6.
Hepatol Commun ; 3(8): 1085-1097, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31388629

ABSTRACT

Farnesoid X receptor (FXR) agonism is emerging as an important potential therapeutic mechanism of action for multiple chronic liver diseases. The bile acid-derived FXR agonist obeticholic acid (OCA) has shown promise in a phase 2 study in patients with nonalcoholic steatohepatitis (NASH). Here, we report efficacy of the novel nonbile acid FXR agonist tropifexor (LJN452) in two distinct preclinical models of NASH. The efficacy of tropifexor at <1 mg/kg doses was superior to that of OCA at 25 mg/kg in the liver in both NASH models. In a chemical and dietary model of NASH (Stelic animal model [STAM]), tropifexor reversed established fibrosis and reduced the nonalcoholic fatty liver disease activity score and hepatic triglycerides. In an insulin-resistant obese NASH model (amylin liver NASH model [AMLN]), tropifexor markedly reduced steatohepatitis, fibrosis, and profibrogenic gene expression. Transcriptome analysis of livers from AMLN mice revealed 461 differentially expressed genes following tropifexor treatment that included a combination of signatures associated with reduction of oxidative stress, fibrogenesis, and inflammation. Conclusion: Based on preclinical validation in animal models, tropifexor is a promising investigational therapy that is currently under phase 2 development for NASH.

7.
Nat Med ; 24(11): 1732-1742, 2018 11.
Article in English | MEDLINE | ID: mdl-30297908

ABSTRACT

Available corrector drugs are unable to effectively rescue the folding defects of CFTR-ΔF508 (or CFTR-F508del), the most common disease-causing mutation of the cystic fibrosis transmembrane conductance regulator, a plasma membrane (PM) anion channel, and thus to substantially ameliorate clinical phenotypes of cystic fibrosis (CF). To overcome the corrector efficacy ceiling, here we show that compounds targeting distinct structural defects of CFTR can synergistically rescue mutant expression and function at the PM. High-throughput cell-based screens and mechanistic analysis identified three small-molecule series that target defects at nucleotide-binding domain (NBD1), NBD2 and their membrane-spanning domain (MSD) interfaces. Although individually these compounds marginally improve ΔF508-CFTR folding efficiency, function and stability, their combinations lead to ~50-100% of wild-type-level correction in immortalized and primary human airway epithelia and in mouse nasal epithelia. Likewise, corrector combinations were effective against rare missense mutations in various CFTR domains, probably acting via structural allostery, suggesting a mechanistic framework for their broad application.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/chemistry , Cystic Fibrosis/drug therapy , Protein Folding/drug effects , Small Molecule Libraries/chemistry , Allosteric Regulation/drug effects , Bronchi/cytology , Bronchi/drug effects , Cell Membrane/drug effects , Cell Membrane/genetics , Cystic Fibrosis/genetics , Cystic Fibrosis/pathology , Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , Mutation , Nasal Mucosa/cytology , Nasal Mucosa/drug effects , Protein Domains/drug effects , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
8.
Nat Methods ; 15(11): 941-946, 2018 11.
Article in English | MEDLINE | ID: mdl-30297964

ABSTRACT

CRISPR-Cas9 screening allows genome-wide interrogation of gene function. Currently, to achieve the high and uniform Cas9 expression desirable for screening, one needs to engineer stable and clonal Cas9-expressing cells-an approach that is not applicable in human primary cells. Guide Swap permits genome-scale pooled CRISPR-Cas9 screening in human primary cells by exploiting the unexpected finding that editing by lentivirally delivered, targeted guide RNAs (gRNAs) occurs efficiently when Cas9 is introduced in complex with nontargeting gRNA. We validated Guide Swap in depletion and enrichment screens in CD4+ T cells. Next, we implemented Guide Swap in a model of ex vivo hematopoiesis, and identified known and previously unknown regulators of CD34+ hematopoietic stem and progenitor cell (HSPC) expansion. We anticipate that this platform will be broadly applicable to other challenging cell types, and thus will enable discovery in previously inaccessible but biologically relevant human primary cell systems.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Gene Editing , Genome, Human , Hematopoietic Stem Cells/metabolism , RNA, Guide, Kinetoplastida/genetics , CD8-Positive T-Lymphocytes/cytology , Cells, Cultured , HEK293 Cells , Hematopoietic Stem Cells/cytology , Humans
9.
J Med Chem ; 60(24): 9960-9973, 2017 12 28.
Article in English | MEDLINE | ID: mdl-29148806

ABSTRACT

The farnesoid X receptor (FXR) is a nuclear receptor that acts as a master regulator of bile acid metabolism and signaling. Activation of FXR inhibits bile acid synthesis and increases bile acid conjugation, transport, and excretion, thereby protecting the liver from the harmful effects of bile accumulation, leading to considerable interest in FXR as a therapeutic target for the treatment of cholestasis and nonalcoholic steatohepatitis. We identified a novel series of highly potent non-bile acid FXR agonists that introduce a bicyclic nortropine-substituted benzothiazole carboxylic acid moiety onto a trisubstituted isoxazole scaffold. Herein, we report the discovery of 1 (tropifexor, LJN452), a novel and highly potent agonist of FXR. Potent in vivo activity was demonstrated in rodent PD models by measuring the induction of FXR target genes in various tissues. Tropifexor has advanced into phase 2 human clinical trials in patients with NASH and PBC.


Subject(s)
Benzothiazoles/pharmacology , Cholestasis/drug therapy , Isoxazoles/pharmacology , Non-alcoholic Fatty Liver Disease/drug therapy , Receptors, Cytoplasmic and Nuclear/agonists , Administration, Oral , Animals , Benzothiazoles/therapeutic use , Biological Availability , Dogs , Drug Evaluation, Preclinical/methods , Fibroblast Growth Factors/genetics , Gene Expression Regulation/drug effects , Humans , Isoxazoles/therapeutic use , Male , Microsomes, Liver/drug effects , Piperidines/chemistry , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Structure-Activity Relationship , Triglycerides/blood
10.
Cancer Res ; 76(6): 1591-602, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26825170

ABSTRACT

Non-small cell lung cancer patients carrying oncogenic EGFR mutations initially respond to EGFR-targeted therapy, but later elicit minimal response due to dose-limiting toxicities and acquired resistance. EGF816 is a novel, irreversible mutant-selective EGFR inhibitor that specifically targets EGFR-activating mutations arising de novo and upon resistance acquisition, while sparing wild-type (WT) EGFR. EGF816 potently inhibited the most common EGFR mutations L858R, Ex19del, and T790M in vitro, which translated into strong tumor regressions in vivo in several patient-derived xenograft models. Notably, EGF816 also demonstrated antitumor activity in an exon 20 insertion mutant model. At levels above efficacious doses, EGF816 treatment led to minimal inhibition of WT EGFR and was well tolerated. In single-dose studies, EGF816 provided sustained inhibition of EGFR phosphorylation, consistent with its ability for irreversible binding. Furthermore, combined treatment with EGF816 and INC280, a cMET inhibitor, resulted in durable antitumor efficacy in a xenograft model that initially developed resistance to first-generation EGFR inhibitors via cMET activation. Thus, we report the first preclinical characterization of EGF816 and provide the groundwork for its current evaluation in phase I/II clinical trials in patients harboring EGFR mutations, including T790M.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Mutation/drug effects , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Lung Neoplasms/metabolism , Mice , Mice, Nude , Phosphorylation/drug effects , Rats , Xenograft Model Antitumor Assays/methods
11.
Nat Commun ; 6: 8372, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26496802

ABSTRACT

Insufficient pancreatic ß-cell mass or function results in diabetes mellitus. While significant progress has been made in regulating insulin secretion from ß-cells in diabetic patients, no pharmacological agents have been described that increase ß-cell replication in humans. Here we report aminopyrazine compounds that stimulate robust ß-cell proliferation in adult primary islets, most likely as a result of combined inhibition of DYRK1A and GSK3B. Aminopyrazine-treated human islets retain functionality in vitro and after transplantation into diabetic mice. Oral dosing of these compounds in diabetic mice induces ß-cell proliferation, increases ß-cell mass and insulin content, and improves glycaemic control. Biochemical, genetic and cell biology data point to Dyrk1a as the key molecular target. This study supports the feasibility of treating diabetes with an oral therapy to restore ß-cell mass, and highlights a tractable pathway for future drug discovery efforts.


Subject(s)
Cell Proliferation , Glycogen Synthase Kinase 3/genetics , Insulin-Secreting Cells/cytology , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Animals , Cell Division/drug effects , Cell Proliferation/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Down-Regulation/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/enzymology , Male , Mice , Mice, Transgenic , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Pyridazines/pharmacology , Dyrk Kinases
12.
Bioorg Med Chem Lett ; 24(23): 5478-83, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25455488

ABSTRACT

Systematic SAR optimization of the GPR119 agonist lead 1, derived from an internal HTS campaign, led to compound 29. Compound 29 displays significantly improved in vitro activity and oral exposure, leading to GLP1 elevation in acutely dosed mice and reduced glucose excursion in an OGTT study in rats at doses ⩾10 mg/kg.


Subject(s)
Pyrimidines/chemical synthesis , Receptors, G-Protein-Coupled/drug effects , Animals , Drug Discovery , Mice , Molecular Structure , Rats , Structure-Activity Relationship
13.
Bioorg Med Chem Lett ; 24(10): 2383-7, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24751443

ABSTRACT

Screening hit 5 was identified in a biochemical screen for GPR119 agonists. Compound 5 was structurally novel, displayed modest biochemical activity and no oral exposure, but was structurally distinct from typical GPR119 agonist scaffolds. Systematic optimization led to compound 36 with significantly improved in vitro activity and oral exposure, to elevate GLP1 acutely in an in vivo mouse model at a dose of 10mg/kg.


Subject(s)
Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Diabetes Mellitus, Type 2/drug therapy , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacology , Mice , Mice, Inbred C57BL , Pyrazoles/chemistry , Pyrimidines/chemistry , Structure-Activity Relationship
14.
J Med Chem ; 57(8): 3263-82, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24666203

ABSTRACT

Activation of the G-protein coupled receptor (GPCR) Takeda G-protein receptor 5 (TGR5), also known as G-protein bile acid receptor 1 (GPBAR1), has been shown to play a key role in pathways associated with diabetes, metabolic syndrome, and autoimmune disease. Nipecotamide 5 was identified as an attractive starting point after a high-throughput screen (HTS) for receptor agonists. A comprehensive hit-to-lead effort culminated in the discovery of 45h as a potent, selective, and bioavailable TGR5 agonist to test in preclinical metabolic disease models. In genetically obese mice (ob/ob), 45h was as effective as a dipeptidyl peptidase-4 (DPP-4) inhibitor at reducing peak glucose levels in an acute oral glucose tolerance test (OGTT), but this effect was lost upon chronic dosing.


Subject(s)
Hypoglycemic Agents/chemical synthesis , Piperazines/chemical synthesis , Receptors, G-Protein-Coupled/agonists , Administration, Oral , Animals , Biological Availability , Drug Discovery , Glucagon-Like Peptide 1/analysis , Humans , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Piperazines/pharmacology , Structure-Activity Relationship
15.
Cancer Discov ; 4(6): 662-673, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24675041

ABSTRACT

UNLABELLED: Non-small cell lung cancers (NSCLC) harboring anaplastic lymphoma kinase (ALK) gene rearrangements invariably develop resistance to the ALK tyrosine kinase inhibitor (TKI) crizotinib. Herein, we report the first preclinical evaluation of the next-generation ALK TKI, ceritinib (LDK378), in the setting of crizotinib resistance. An interrogation of in vitro and in vivo models of acquired resistance to crizotinib, including cell lines established from biopsies of patients with crizotinib-resistant NSCLC, revealed that ceritinib potently overcomes crizotinib-resistant mutations. In particular, ceritinib effectively inhibits ALK harboring L1196M, G1269A, I1171T, and S1206Y mutations, and a cocrystal structure of ceritinib bound to ALK provides structural bases for this increased potency. However, we observed that ceritinib did not overcome two crizotinib-resistant ALK mutations, G1202R and F1174C, and one of these mutations was identified in 5 of 11 biopsies from patients with acquired resistance to ceritinib. Altogether, our results demonstrate that ceritinib can overcome crizotinib resistance, consistent with clinical data showing marked efficacy of ceritinib in patients with crizotinib-resistant disease. SIGNIFICANCE: The second-generation ALK inhibitor ceritinib can overcome several crizotinib-resistant mutations and is potent against several in vitro and in vivo laboratory models of acquired resistance to crizotinib. These findings provide the molecular basis for the marked clinical activity of ceritinib in patients with ALK-positive NSCLC with crizotinib-resistant disease. Cancer Discov; 4(6); 662-73. ©2014 AACR. See related commentary by Ramalingam and Khuri, p. 634 This article is highlighted in the In This Issue feature, p. 621.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Sulfones/therapeutic use , Anaplastic Lymphoma Kinase , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Crizotinib , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, SCID , Mutation , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Tumor Burden
16.
PLoS One ; 8(10): e78483, 2013.
Article in English | MEDLINE | ID: mdl-24205242

ABSTRACT

Type-1 diabetes (T1D) is an autoimmune disease targeting insulin-producing beta cells, resulting in dependence on exogenous insulin. To date, significant efforts have been invested to develop immune-modulatory therapies for T1D treatment. Previously, IL-2 immunotherapy was demonstrated to prevent and reverse T1D at onset in the non-obese diabetic (NOD) mouse model, revealing potential as a therapy in early disease stage in humans. In the NOD model, IL-2 deficiency contributes to a loss of regulatory T cell function. This deficiency can be augmented with IL-2 or antibody bound to IL-2 (Ab/IL-2) therapy, resulting in regulatory T cell expansion and potentiation. However, an understanding of the mechanism by which reconstituted regulatory T cell function allows for reversal of diabetes after onset is not clearly understood. Here, we describe that Ab/IL-2 immunotherapy treatment, given at the time of diabetes onset in NOD mice, not only correlated with reversal of diabetes and expansion of Treg cells, but also demonstrated the ability to significantly increase beta cell proliferation. Proliferation appeared specific to Ab/IL-2 immunotherapy, as anti-CD3 therapy did not have a similar effect. Furthermore, to assess the effect of Ab/IL-2 immunotherapy well after the development of diabetes, we tested the effect of delaying treatment for 4 weeks after diabetes onset, when beta cells were virtually absent. At this late stage after diabetes onset, Ab/IL-2 treatment was not sufficient to reverse hyperglycemia. However, it did promote survival in the absence of exogenous insulin. Proliferation of beta cells could not account for this improvement as few beta cells remained. Rather, abnormal insulin and glucagon dual-expressing cells were the only insulin-expressing cells observed in islets from mice with established disease. Thus, these data suggest that in diabetic NOD mice, beta cells have an innate capacity for regeneration both early and late in disease, which is revealed through IL-2 immunotherapy.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/therapy , Insulin-Secreting Cells/immunology , Interleukin-2/immunology , Animals , Diabetes Mellitus, Type 1/metabolism , Disease Models, Animal , Female , Glucagon/metabolism , Immunotherapy/methods , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Interleukin-2/metabolism , Mice , Mice, Inbred NOD , Regeneration/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
17.
Proc Natl Acad Sci U S A ; 110(50): 20224-9, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277854

ABSTRACT

Wnt signaling is one of the key oncogenic pathways in multiple cancers, and targeting this pathway is an attractive therapeutic approach. However, therapeutic success has been limited because of the lack of therapeutic agents for targets in the Wnt pathway and the lack of a defined patient population that would be sensitive to a Wnt inhibitor. We developed a screen for small molecules that block Wnt secretion. This effort led to the discovery of LGK974, a potent and specific small-molecule Porcupine (PORCN) inhibitor. PORCN is a membrane-bound O-acyltransferase that is required for and dedicated to palmitoylation of Wnt ligands, a necessary step in the processing of Wnt ligand secretion. We show that LGK974 potently inhibits Wnt signaling in vitro and in vivo, including reduction of the Wnt-dependent LRP6 phosphorylation and the expression of Wnt target genes, such as AXIN2. LGK974 is potent and efficacious in multiple tumor models at well-tolerated doses in vivo, including murine and rat mechanistic breast cancer models driven by MMTV-Wnt1 and a human head and neck squamous cell carcinoma model (HN30). We also show that head and neck cancer cell lines with loss-of-function mutations in the Notch signaling pathway have a high response rate to LGK974. Together, these findings provide both a strategy and tools for targeting Wnt-driven cancers through the inhibition of PORCN.


Subject(s)
Membrane Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Pyrazines/pharmacology , Pyridines/pharmacology , Wnt Signaling Pathway/drug effects , Acyltransferases , Animals , Axin Protein/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Cloning, Molecular , High-Throughput Screening Assays , Humans , Mice , Mutagenesis , Phosphorylation/drug effects , Pyrazines/therapeutic use , Pyridines/therapeutic use , Radioligand Assay , Rats , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
J Am Chem Soc ; 135(5): 1669-72, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23330637

ABSTRACT

The identification of factors that promote ß cell proliferation could ultimately move type 1 diabetes treatment away from insulin injection therapy and toward a cure. We have performed high-throughput, cell-based screens using rodent ß cell lines to identify molecules that induce proliferation of ß cells. Herein we report the discovery and characterization of WS6, a novel small molecule that promotes ß cell proliferation in rodent and human primary islets. In the RIP-DTA mouse model of ß cell ablation, WS6 normalized blood glucose and induced concomitant increases in ß cell proliferation and ß cell number. Affinity pulldown and kinase profiling studies implicate Erb3 binding protein-1 and the IκB kinase pathway in the mechanism of action of WS6.


Subject(s)
High-Throughput Screening Assays , Islets of Langerhans/drug effects , Urea/pharmacology , Animals , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Islets of Langerhans/cytology , Mice , Molecular Structure , Molecular Weight , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
19.
Naunyn Schmiedebergs Arch Pharmacol ; 384(6): 565-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21947251

ABSTRACT

Pharmacologic antagonism of cannabinoid 1 receptors (CB1 receptors) in the central nervous system (CNS) suppresses food intake, promotes weight loss, and improves the metabolic profile. Since the CB1 receptor is expressed both in the CNS and in peripheral tissues, therapeutic value may be gained with CB1 receptor inverse agonists acting on receptors in both domains. The present report examines the metabolic and CNS actions of a novel CB1 receptor inverse agonist, compound 64, a 1,5,6-trisubstituted pyrazolopyrimidinone. Compound 64 showed similar or superior binding affinity, in vitro potency, and pharmacokinetic profile compared to rimonabant. Both compounds improved the metabolic profile in diet-induced obese (DIO) rats and obese cynomolgus monkeys. Weight loss tended to be greater in compound 64-treated DIO rats compared to pair-fed counterparts, suggesting that compound 64 may have metabolic effects beyond those elicited by weight loss alone. In the CNS, reversal of agonist-induced hypothermia and hypolocomotion indicated that compound 64 possessed an antagonist activity in vivo. Dosed alone, compound 64 suppressed extinction of conditioned freezing (10 mg/kg) and rapid eye movement (REM) sleep (30 mg/kg), consistent with previous reports for rimonabant, although for REM sleep, compound 64 was greater than threefold less potent than for metabolic effects. Together, these data suggested that (1) impairment of extinction learning and REM sleep suppression are classic, centrally mediated responses to CB1 receptor inverse agonists, and (2) some separation may be achievable between central and peripheral effects with brain-penetrating CB1 receptor inverse agonists while maintaining metabolic efficacy. Furthermore, chronic treatment with compound 64 contributes to evidence that peripheral CB1 receptor blockade may yield beneficial outcomes that exceed those elicited by weight loss alone.


Subject(s)
Obesity/drug therapy , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Animals , Brain/metabolism , Dose-Response Relationship, Drug , Drug Inverse Agonism , Extinction, Psychological/drug effects , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Obesity/metabolism , Piperidines/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Pyrimidinones/administration & dosage , Pyrimidinones/pharmacokinetics , Rats , Rats, Sprague-Dawley , Rimonabant , Sleep, REM/drug effects , Tissue Distribution
20.
Antimicrob Agents Chemother ; 53(5): 1898-906, 2009 May.
Article in English | MEDLINE | ID: mdl-19223628

ABSTRACT

Menstrual toxic shock syndrome is a rare but potentially life-threatening illness manifest through the actions of Staphylococcus aureus toxic shock syndrome toxin 1 (TSST-1). Previous studies have shown that tampon additives can influence staphylococcal TSST-1 production. We report here on the TSST-1-suppressing activity of 34 compounds that are commonly used additives in the pharmaceutical, food, and perfume industries. Many of the tested chemicals had a minimal impact on the growth of S. aureus and yet were potent inhibitors of TSST-1 production. The TSST-1-reducing compounds included surfactants with an ether, amide, or amine linkage to their fatty acid moiety (e.g., myreth-3-myristate, Laureth-3, disodium lauroamphodiacetate, disodium lauramido monoethanolamido, sodium lauriminodipropionic acid, and triethanolamine laureth sulfate); aromatic compounds (e.g. phenylethyl and benzyl alcohols); and several isoprenoids and related compounds (e.g., terpineol and menthol). The membrane-targeting and -altering effects of the TSST-1-suppressing compounds led us to assess the activity of molecules that are known to inhibit fatty acid biosynthesis (e.g., cerulenin, triclosan, and hexachlorophene). These compounds also reduced S. aureus TSST-1 production. This study suggests that more additives than previously recognized inhibit the production of TSST-1.


Subject(s)
Bacterial Toxins , Benzyl Alcohols/pharmacology , Enterotoxins , Fatty Acid Synthesis Inhibitors/pharmacology , Staphylococcus aureus , Superantigens , Surface-Active Agents/pharmacology , Terpenes/pharmacology , Bacterial Toxins/biosynthesis , Benzyl Alcohols/chemistry , Culture Media , Enterotoxins/biosynthesis , Fatty Acid Synthesis Inhibitors/chemistry , Female , Humans , Shock, Septic/microbiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development , Staphylococcus aureus/metabolism , Superantigens/biosynthesis , Superantigens/drug effects , Surface-Active Agents/chemistry , Terpenes/chemistry , Vagina/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL