Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Pharmaceutics ; 15(4)2023 Apr 05.
Article in English | MEDLINE | ID: mdl-37111634

ABSTRACT

The ROR1 receptor tyrosine kinase is expressed in embryonic tissues but is absent in normal adult tissues. ROR1 is of importance in oncogenesis and is overexpressed in several cancers, such as NSCLC. In this study, we evaluated ROR1 expression in NSCLC patients (N = 287) and the cytotoxic effects of a small molecule ROR1 inhibitor (KAN0441571C) in NSCLC cell lines. ROR1 expression in tumor cells was more frequent in non-squamous (87%) than in squamous (57%) carcinomas patients, while 21% of neuroendocrine tumors expressed ROR1 (p = 0.0001). A significantly higher proportion of p53 negative patients in the ROR1+ group than in the p53 positive non-squamous NSCLC patients (p = 0.03) was noted. KAN0441571C dephosphorylated ROR1 and induced apoptosis (Annexin V/PI) in a time- and dose-dependent manner in five ROR1+ NSCLC cell lines and was superior compared to erlotinib (EGFR inhibitor). Apoptosis was confirmed by the downregulation of MCL-1 and BCL-2, as well as PARP and caspase 3 cleavage. The non-canonical Wnt pathway was involved. The combination of KAN0441571C and erlotinib showed a synergistic apoptotic effect. KAN0441571C also inhibited proliferative (cell cycle analyses, colony formation assay) and migratory (scratch wound healing assay) functions. Targeting NSCLC cells by a combination of ROR1 and EGFR inhibitors may represent a novel promising approach for the treatment of NSCLC patients.

2.
Pharmaceutics ; 14(10)2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36297673

ABSTRACT

The receptor tyrosine kinase orphan receptor 1 (ROR1) is absent in most normal adult tissues but overexpressed in various malignancies and is of importance for tumor cell survival, proliferation, and metastasis. In this study, we evaluated the apoptotic effects of a novel small molecule inhibitor of ROR1 (KAN0441571C) as well as venetoclax (BCL-2 inhibitor), bendamustine, idelalisib (PI3Kδ inhibitor), everolimus (mTOR inhibitor), and ibrutinib (BTK inhibitor) alone or in combination in human MCL primary cells and cell lines. ROR1 expression was evaluated by flow cytometry and Western blot (WB). Cytotoxicity was analyzed by MTT and apoptosis by Annexin V/PI staining as well as signaling and apoptotic proteins (WB). ROR1 was expressed both in patient-derived MCL cells and human MCL cell lines. KAN0441571C alone induced significant time- and dose-dependent apoptosis of MCL cells. Apoptosis was accompanied by decreased expression of MCL-1 and BCL-2 and cleavage of PARP and caspase 3. ROR1 was dephosphorylated as well as ROR1-associated signaling pathway molecules, including the non-canonical WNT signaling pathway (PI3Kδ/AKT/mTOR). The combination of KAN0441571C and ibrutinib, venetoclax, idelalisib, everolimus, or bendamustine had a synergistic apoptotic effect and significantly prevented phosphorylation of ROR1-associated signaling molecules as compared to KAN0441571C alone. Our results suggest that targeting ROR1 by a small molecule inhibitor, KAN0441571C, should be further evaluated particularly in combination with other targeting drugs as a new therapeutic approach for MCL.

3.
Cell Rep ; 37(12): 110129, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34936867

ABSTRACT

Writing and erasing of posttranslational modifications are crucial to phenotypic plasticity and antigenic variation of eukaryotic pathogens. Targeting pathogens' modification machineries, thus, represents a valid approach to fighting parasitic diseases. However, identification of parasitic targets and the development of selective anti-parasitic drugs still represent major bottlenecks. Here, we show that the zinc-dependent histone deacetylases (HDACs) of the protozoan parasite Trypanosoma cruzi are key regulators that have significantly diverged from their human counterparts. Depletion of T. cruzi class I HDACs tcDAC1 and tcDAC2 compromises cell-cycle progression and division, leading to cell death. Notably, tcDAC2 displays a deacetylase activity essential to the parasite and shows major structural differences with human HDACs. Specifically, tcDAC2 harbors a modular active site with a unique subpocket targeted by inhibitors showing substantial anti-parasitic effects in cellulo and in vivo. Thus, the targeting of the many atypical HDACs in pathogens can enable anti-parasitic selective chemical impairment.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Trypanosoma cruzi/enzymology , Trypanosoma cruzi/genetics , Trypanosoma cruzi/metabolism , Animals , Catalytic Domain , Cell Cycle , Cell Division/drug effects , Cell Line , Cell Proliferation/drug effects , Chagas Disease/drug therapy , Chagas Disease/parasitology , Chlorocebus aethiops , DNA, Protozoan , Female , Genetic Complementation Test , Histone Deacetylase Inhibitors/chemistry , Histone Deacetylases/chemistry , Host-Parasite Interactions , Humans , Male , Mice , Mice, Inbred BALB C , Models, Molecular , Phylogeny , Protein Conformation , Protein Processing, Post-Translational , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Sequence Deletion , Trypanosoma cruzi/drug effects , Vero Cells
4.
Handb Exp Pharmacol ; 269: 75-99, 2021.
Article in English | MEDLINE | ID: mdl-34490515

ABSTRACT

Receptor tyrosine kinases (RTKs) are frequently dysregulated in malignancies and important for the malignant characteristics of tumor cells. RTKs are attractive structures for drug targeting of cancer. The RTK ROR1 is of significance during embryogenesis but downregulated in post-partum tissues. However, ROR1 is overexpressed in several hematological and solid tumors and important for tumor cell proliferation, survival, migration, and metastasis. WNT5a is a main ligand for ROR1. Several clinical trials are ongoing using anti-ROR1 antibody based drugs directed against the external domain (monoclonal antibodies, BiTE, CAR-T). We have produced small molecules (KAN834/1571c) fitting to the ATP pocket of the intracellular tyrosine kinase (TK) domain of ROR1 (TK inhibitor, TKI). These inhibitors of ROR1 prevented ROR1 phosphorylation and inactivated the WNT/ß-catenin independent as well as WNT/ß-catenin dependent pathways. ROR1-TKI induced apoptosis of ROR1 positive fresh patient derived tumor cells and appropriate cell lines and a dose and time dependent tumor reduction in animal models. In combination with other clinically relevant targeting drugs as venetoclax a synergistic apoptotic effect was seen. Two other small molecules (ARI-1 and strictinin) bound also to ROR1 and inhibited tumor growth. Development of small molecule ROR1 inhibitors is warranted to include this novel therapeutic approach for cancer therapy.


Subject(s)
Protein Kinase Inhibitors , Receptor Tyrosine Kinase-like Orphan Receptors , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Humans , Protein Kinase Inhibitors/pharmacology
5.
Cell Death Dis ; 12(6): 577, 2021 06 04.
Article in English | MEDLINE | ID: mdl-34088900

ABSTRACT

Small cell lung cancer (SCLC) remains a deadly form of cancer, with a 5-year survival rate of less than 10 percent, necessitating novel therapies. Receptor tyrosine kinase-like orphan receptor 1 (ROR1) is an oncofetal protein that is emerging as a therapeutic target and is co-expressed with BCL2 in multiple tumor types due to microRNA coregulation. We hypothesize that ROR1-targeted therapy is effective in small cell lung cancer and synergizes with therapeutic BCL2 inhibition. Tissue microarrays (TMAs) and formalin-fixed paraffin-embedded (FFPE) SCLC patient samples were utilized to determine the prevalence of ROR1 and BCL2 expression in SCLC. Eight SCLC-derived cell lines were used to determine the antitumor activity of a small molecule ROR1 inhibitor (KAN0441571C) alone and in combination with the BCL2 inhibitor venetoclax. The Chou-Talalay method was utilized to determine synergy with the drug combination. ROR1 and BCL2 protein expression was identified in 93% (52/56) and 86% (48/56) of SCLC patient samples, respectively. Similarly, ROR1 and BCL2 were shown by qRT-PCR to have elevated expression in 79% (22/28) and 100% (28/28) of SCLC patient samples, respectively. KAN0441571C displayed efficacy in 8 SCLC cell lines, with an IC50 of 500 nM or less. Synergy as defined by a combination index of <1 via the Chou-Talalay method between KAN0441571C and venetoclax was demonstrated in 8 SCLC cell lines. We have shown that ROR1 inhibition is synergistic with BCL2 inhibition in SCLC models and shows promise as a novel therapeutic target in SCLC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Lung Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors , Small Cell Lung Carcinoma/drug therapy , Sulfonamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cell Line, Tumor , Drug Synergism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Molecular Targeted Therapy , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/biosynthesis , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Small Cell Lung Carcinoma/genetics , Small Cell Lung Carcinoma/metabolism , Sulfonamides/administration & dosage , Survival Analysis
6.
EJHaem ; 2(3): 498-502, 2021 Aug.
Article in English | MEDLINE | ID: mdl-35844694

ABSTRACT

ROR1 - a receptor tyrosine kinase - is overexpressed in CLL. Ibrutinib, a Bruton's tyrosine kinase inhibitor, is clinically effective in CLL but patients may develop resistance. We evaluated the effect of an ROR1 inhibitor, KAN0441571C, in CLL cells from six patients obtained before and after developing resistance to ibrutinib. The ROR1 inhibitor induced apoptosis in ibrutinib-resistant CLL cells to the same degree as in ibrutinib-sensitive cells and dephosphorylated ROR1. This was also noted in one patient who became resistant to both ibrutinib and the Bcl-2 inhibitor venetoclax. The combination of ROR1 inhibitor and venetoclax had a synergistic apoptotic effect on ibrutinib-resistant cells.

7.
Biomedicines ; 8(6)2020 Jun 23.
Article in English | MEDLINE | ID: mdl-32586008

ABSTRACT

The receptor tyrosine kinase ROR1 is absent in most normal adult tissues, but overexpressed in several malignancies. In this study, we explored clinical and functional inhibitory aspects of ROR1 in diffuse large B-cell lymphoma (DLBCL). ROR1 expression in tumor cells was more often observed in primary refractory DLBCL, Richter's syndrome and transformed follicular lymphoma than in relapsed and non-relapsed DLBCL patients (p < 0.001). A survival effect of ROR1 expression was preliminarily observed in relapsed/refractory patients independent of gender and stage but not of age, cell of origin and international prognostic index. A second generation small molecule ROR1 inhibitor (KAN0441571C) induced apoptosis of ROR1+ DLBCL cell lines, similar to venetoclax (BCL-2 inhibitor) but superior to ibrutinib (BTK inhibitor). The combination of KAN0441571C and venetoclax at EC50 concentrations induced almost complete killing of DLBCL cell lines. Apoptosis was accompanied by the downregulation of BCL-2 and MCL-1 and confirmed by the cleavage of PARP and caspases 3, 8, 9. PI3Kδ/AKT/mTOR (non-canonical Wnt pathway) as well as ß-catenin and CK1δ (canonical pathway) were inactivated. In zebra fishes transplanted with a ROR1+ DLBCL cell line, KAN0441571C induced a significant tumor reduction. New drugs with mechanisms of action other than those available for DLBCL are warranted. ROR1 inhibitors might represent a novel promising approach.

8.
Cancers (Basel) ; 11(12)2019 Dec 16.
Article in English | MEDLINE | ID: mdl-31888209

ABSTRACT

In breast cancer, Lysine-specific demethylase-1 (LSD1) and other lysine demethylases (KDMs), such as Lysine-specific demethylase 6A also known as Ubiquitously transcribed tetratricopeptide repeat, X chromosome (UTX), are co-expressed and co-localize with estrogen receptors (ERs), suggesting the potential use of hybrid (epi)molecules to target histone methylation and therefore regulate/redirect hormone receptor signaling. Here, we report on the biological activity of a dual-KDM inhibitor (MC3324), obtained by coupling the chemical properties of tranylcypromine, a known LSD1 inhibitor, with the 2OG competitive moiety developed for JmjC inhibition. MC3324 displays unique features not exhibited by the single moieties and well-characterized mono-pharmacological inhibitors. Inhibiting LSD1 and UTX, MC3324 induces significant growth arrest and apoptosis in hormone-responsive breast cancer model accompanied by a robust increase in H3K4me2 and H3K27me3. MC3324 down-regulates ERα in breast cancer at both transcriptional and non-transcriptional levels, mimicking the action of a selective endocrine receptor disruptor. MC3324 alters the histone methylation of ERα-regulated promoters, thereby affecting the transcription of genes involved in cell surveillance, hormone response, and death. MC3324 reduces cell proliferation in ex vivo breast cancers, as well as in breast models with acquired resistance to endocrine therapies. Similarly, MC3324 displays tumor-selective potential in vivo, in both xenograft mice and chicken embryo models, with no toxicity and good oral efficacy. This epigenetic multi-target approach is effective and may overcome potential mechanism(s) of resistance in breast cancer.

9.
Eur J Med Chem ; 161: 277-291, 2019 Jan 01.
Article in English | MEDLINE | ID: mdl-30366254

ABSTRACT

Despite the recent reductions in the global burden of malaria, this disease remains a devastating cause of death in tropical and subtropical regions. As there is no broadly effective vaccine for malaria, prevention and treatment still rely on chemotherapy. Unfortunately, emerging resistance to the gold standard artemisinin combination therapies means that new drugs with novel modes of action are urgently needed. In this context, Plasmodium histone modifying enzymes have emerged as potential drug targets, prompting us to develop and optimize compounds directed against such epigenetic targets. A panel of 51 compounds designed to target different epigenetic enzymes were screened for activity against Plasmodium falciparum parasites. Based on in vitro activity against drug susceptible and drug-resistant P. falciparum lines, selectivity index criterion and favorable pharmacokinetic properties, four compounds, one HDAC inhibitor (1) and three DNMT inhibitors (37, 43 and 45), were selected for preclinical studies in a mouse model of malaria. In vivo data showed that 37, 43 and 45 exhibited oral efficacy in the mouse model of Plasmodium berghei infection. These compounds represent promising starting points for the development of novel antimalarial drugs.


Subject(s)
Antimalarials/pharmacology , Plasmodium berghei/drug effects , Plasmodium falciparum/drug effects , Quinazolines/pharmacology , Animals , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Fibroblasts/drug effects , Fibroblasts/microbiology , Humans , Mice , Mice, Inbred BALB C , Molecular Conformation , Parasitic Sensitivity Tests , Plasmodium falciparum/growth & development , Quinazolines/chemical synthesis , Quinazolines/chemistry , Structure-Activity Relationship
10.
Nat Commun ; 9(1): 3872, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250201

ABSTRACT

The glycolytic PFKFB3 enzyme is widely overexpressed in cancer cells and an emerging anti-cancer target. Here, we identify PFKFB3 as a critical factor in homologous recombination (HR) repair of DNA double-strand breaks. PFKFB3 rapidly relocates into ionizing radiation (IR)-induced nuclear foci in an MRN-ATM-γH2AX-MDC1-dependent manner and co-localizes with DNA damage and HR repair proteins. PFKFB3 relocalization is critical for recruitment of HR proteins, HR activity, and cell survival upon IR. We develop KAN0438757, a small molecule inhibitor that potently targets PFKFB3. Pharmacological PFKFB3 inhibition impairs recruitment of ribonucleotide reductase M2 and deoxynucleotide incorporation upon DNA repair, and reduces dNTP levels. Importantly, KAN0438757 induces radiosensitization in transformed cells while leaving non-transformed cells unaffected. In summary, we identify a key role for PFKFB3 enzymatic activity in HR repair and present KAN0438757, a selective PFKFB3 inhibitor that could potentially be used as a strategy for the treatment of cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Hydroxybenzoates/pharmacology , Neoplasms/therapy , Phosphofructokinase-2/antagonists & inhibitors , Sulfones/pharmacology , Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Chemoradiotherapy/methods , DNA Breaks, Double-Stranded/radiation effects , Dideoxynucleotides/metabolism , Enzyme Inhibitors/therapeutic use , Humans , Hydroxybenzoates/therapeutic use , Phosphofructokinase-2/genetics , Phosphofructokinase-2/metabolism , RNA, Small Interfering/metabolism , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Radiation, Ionizing , Recombinational DNA Repair/drug effects , Recombinational DNA Repair/radiation effects , Sulfones/therapeutic use
11.
PLoS One ; 13(6): e0198038, 2018.
Article in English | MEDLINE | ID: mdl-29856777

ABSTRACT

There is a great unmet medical need in pancreatic carcinoma (PC) for novel drugs with other mechanisms of action than existing. PC cells express the onco-fetal RTK ROR1, absent on most normal post-partem cells. ROR1 is involved in proliferation, survival, EMT and metastasis of tumor cells in various malignancies. A small molecule inhibitor (KAN0439834) (530 Da) targeting the TK domain of ROR1 was developed and the activity in ROR1 expressing human PC cell lines (n = 8) evaluated. The effects were compared to a murine mAb against the external part of ROR1, gemcitabine, erlotinib and ibrutinib. KAN0439834 induced significant apoptosis of the tumor cells. EC50 values for KAN0439834 varied between 250-650 nM depending on the cell line. The corresponding values for erlotinib and ibrutinib were 10-40 folds higher. KAN0439834 was much more effective in inducing tumor cell death than the ROR1 mAb although both inhibited ROR1 phosphorylation and downstream non-canonical Wnt pathway molecules. Combination of KAN0439834 with erlotinib or ibrutinib had significant additive effects on tumor cell death. A first-in-class small molecule ROR1 inhibitor (KAN0439834) showed promising in vitro activity against a number of human PC cell lines. Interesting is the additive effects of erlotinib and ibrutinib which warrants further studies as both these agents are in clinical trials for pancreatic carcinoma.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Erlotinib Hydrochloride/pharmacology , Organic Chemicals/pharmacology , Pancreas/pathology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors , Adenine/analogs & derivatives , Cell Line, Tumor , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Low Density Lipoprotein Receptor-Related Protein-6/chemistry , Pancreas/drug effects , Pancreatic Neoplasms/pathology , Phosphorylation/drug effects , Piperidines , Protein Multimerization/drug effects , Protein Structure, Quaternary , Receptor Tyrosine Kinase-like Orphan Receptors/chemistry , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism
12.
Future Med Chem ; 7(6): 765-82, 2015.
Article in English | MEDLINE | ID: mdl-25996069

ABSTRACT

The sirtuins form a superfamily of evolutionarily conserved NAD(+)-dependent protein N-ϵ-acyl-lysine (AcK) deacylases with roles in a variety of key cellular processes. Sirtuins have a broadly conserved overall structure with a catalytic site formed by a hydrophobic channel between the NAD(+)-binding Rossmann fold domain and a smaller Zn(2+)-binding domain. Schistosomes express five members of the sirtuin family and generic sirtuin inhibitors induce apoptosis and death in schistosome larvae, the disruption of adult worm pairs, inhibition of egg laying and damage to the male and female worm reproductive systems. Sirtuins in schistosomes and other parasitic flatworms present structural differences from their human orthologues that should allow the development of selective inhibitors that can be developed as drug leads.


Subject(s)
Anthelmintics/pharmacology , Drug Discovery/methods , Helminth Proteins/antagonists & inhibitors , Schistosoma/drug effects , Schistosomiasis/drug therapy , Sirtuins/antagonists & inhibitors , Amino Acid Sequence , Animals , Anthelmintics/chemistry , Helminth Proteins/chemistry , Helminth Proteins/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Molecular Targeted Therapy/methods , Schistosoma/chemistry , Schistosoma/metabolism , Schistosomiasis/parasitology , Sequence Alignment , Sirtuins/chemistry , Sirtuins/metabolism
13.
J Biomol Screen ; 20(1): 112-21, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25325257

ABSTRACT

Sirtuins are NAD(+)-dependent histone deacetylases (HDACs) that cleave off acetyl but also other acyl groups from the ϵ-amino group of lysines in histones and other substrate proteins. Five sirtuin isoforms are encoded in the genome of the parasitic pathogen Schistosoma mansoni. During its life cycle, S. mansoni undergoes drastic changes in phenotype that are associated with epigenetic modifications. Previous work showed strong effects of hSirt2 inhibitors on both worm life span and reproduction. Thus, we postulate smSirt2 as a new antiparasite target. We report both the optimization of a homogeneous fluorescence-based assay and the development of a new heterogeneous fluorescence-based assay to determine smSirt2 activity. The homogeneous assay uses a coumarin-labeled acetyl lysine derivative, and the heterogeneous version is using a biotinylated and fluorescence-labeled oligopeptide. Magnetic streptavidin-coated beads allow higher substrate loading per well than streptavidin-coated microtiter plates and make it possible to screen for inhibitors of either smSirt2 or its human isoform (hSirt2) for selectivity studies. We also present hits from a pilot screen with inhibitors showing an IC50 lower than 50 µM. Binding of the hits to their targets is rationalized by docking studies using a homology model of smSirt2.


Subject(s)
Fluorometry/methods , Schistosoma mansoni/enzymology , Sirtuin 2/metabolism , Animals , Binding Sites , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Epigenesis, Genetic/drug effects , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Niacinamide/chemistry , Niacinamide/pharmacology , Protein Binding , Schistosoma mansoni/drug effects , Schistosoma mansoni/genetics , Sirtuin 2/antagonists & inhibitors , Sirtuin 2/genetics , Structure-Activity Relationship , Tyrphostins/chemistry , Tyrphostins/pharmacology
14.
J Biomol Screen ; 17(7): 933-45, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22644269

ABSTRACT

The TWEAK-Fn14 pathway is upregulated in models of inflammation, autoimmune diseases, and cancer. Both TWEAK and Fn14 show increased expression also in the CNS in response to different stimuli, particularly astrocytes, microglia, and neurons, leading to activation of NF-κB and release of proinflammatory cytokines. Although neutralizing antibodies against these proteins have been shown to have therapeutic efficacy in animal models of inflammation, no small-molecule therapeutics are yet available. Here, we describe the development of a novel homogeneous time-resolved fluorescence (HTRF)-based screening assay together with several counterassays for the identification of small-molecule inhibitors of this protein-protein interaction. Recombinant HIS-TWEAK and Fn14-Fc proteins as well as FLAG-TWEAK and Fn14-FLAG proteins and an anti-Fn14 antibody were used to establish and validate these assays and to screen a library of 60 000 compounds. Two HTRF counterassays with unrelated proteins in the same assay format, an antiaggregation assay and a redox assay, were applied to filter out potential false-positive compounds. The novel assay and associated screening cascade should be useful for the discovery of small-molecule inhibitors of the TWEAK-Fn14 protein interaction.


Subject(s)
Drug Screening Assays, Antitumor/methods , High-Throughput Screening Assays/methods , Receptors, Tumor Necrosis Factor/antagonists & inhibitors , Tumor Necrosis Factor Inhibitors , Autoimmune Diseases/metabolism , Cell Line , Cytokine TWEAK , HEK293 Cells , Humans , Inflammation/metabolism , Neoplasms/metabolism , Oligopeptides , Peptides/metabolism , Receptors, Tumor Necrosis Factor/metabolism , TWEAK Receptor , Tumor Necrosis Factors/metabolism
15.
J Med Chem ; 51(9): 2777-86, 2008 May 08.
Article in English | MEDLINE | ID: mdl-18410081

ABSTRACT

Seven novel binders, binding in the active site of Plasmodium falciparum spermidine synthase, were identified by structure-based virtual screening. The binding of these compounds was experimentally verified by NMR techniques. Spermidine synthase, an enzyme involved in the polyamine pathway, has been suggested as a target for treating malaria. The virtual screening protocol combined 3D pharmacophore filtering, docking, and scoring, focusing on finding compounds predicted to form interactions mimicking those of a previously known binder. The virtual screen resulted in the selection of 28 compounds that were acquired and tested from 2.6 million starting structures. Two of the seven binders were predicted to bind in the amino substrate binding pocket. Both of these showed stronger binding upon addition of methylthioadenosine, one of the two products of the enzyme, and a known binder and inhibitor. The five other compounds were predicted to bind in the part of the active site where the other substrate, decarboxylated S-adenosylmethionine, binds. These five compounds all competed for binding with methylthioadenosine.


Subject(s)
Models, Molecular , Plasmodium falciparum/enzymology , Quantitative Structure-Activity Relationship , Spermidine Synthase/chemistry , Animals , Binding Sites , Databases, Factual , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 2-Ring/chemistry , Hydrogen Bonding , Magnetic Resonance Spectroscopy , Protein Binding , Spermidine Synthase/antagonists & inhibitors , Spermidine Synthase/metabolism , Structure-Activity Relationship
16.
Bioorg Med Chem Lett ; 14(4): 891-5, 2004 Feb 23.
Article in English | MEDLINE | ID: mdl-15012988

ABSTRACT

The inhibitory effect on PTP1B caused by the addition of pyridazine analogues has been investigated. Biophysical techniques, that is, mass spectrometry (MS), nuclear magnetic resonance (NMR), and isothermal titration calorimetry (ITC) were used for the characterization. Pyridazine analogues cause catalytic oxidation of the reducing agent, generating hydrogen peroxide that oxidizes the active site cysteine on the enzyme, leading to enzyme inactivation. Two additional compound classes show the same effect. We found one common structural feature in these molecules that allows the reaction with triplet molecular oxygen to be less endothermic. A proposed mechanism for the catalytic redox cycle is described.


Subject(s)
Protein Tyrosine Phosphatases/antagonists & inhibitors , Pyridazines/pharmacology , Catalysis , Hydrogen Peroxide/chemistry , Hydrogen Peroxide/metabolism , Molecular Structure , Oxidation-Reduction , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/chemistry , Pyridazines/classification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
Comb Chem High Throughput Screen ; 5(8): 623-30, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12470259

ABSTRACT

NMR based screening has become an important tool in the pharmaceutical industry. Methods that provide information on the location of small molecule binding sites on the surface of a drug target (e. g. SAR-by-NMR and related techniques) are of particular interest. In order to extend the applicability of such techniques to drug targets of higher molecular weight, selective labeling strategies may be employed. Dual-amino acid selective labeling and site directed non-native amino acid replacement (SNAAR) allow for the selective detection of NMR resonances of a specific amino acid residue. This results in significantly reduced spectral complexity, which not only enables application to higher molecular weight systems, but also eliminates the need for sequential resonance assignment in order to identify the binding site. Regio-selective (or segmental) labeling of an entire protein domain of a multi domain protein may also be achieved. Labeling only a selected part of a multi domain protein (e. g. a catalytic or ligand binding domain) is an attractive way to simplify the spectral interpretation without disturbing the system under study.


Subject(s)
Amino Acids/analysis , Isotope Labeling/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Amino Acid Sequence , Amino Acids/chemical synthesis , Binding Sites , Carbon Isotopes , Ligands , Molecular Sequence Data , Mutagenesis, Site-Directed , Nitrogen Isotopes , Protein Structure, Tertiary , Structure-Activity Relationship , Substrate Specificity
18.
J Am Chem Soc ; 124(40): 11874-80, 2002 Oct 09.
Article in English | MEDLINE | ID: mdl-12358531

ABSTRACT

The time-limiting step in HTS often is the development of an appropriate assay. In addition, hits from HTS fairly often turn out to be false positives and generally display unfavorable properties for further development. Here we describe an alternative process for hit generation, applied to the human adipocyte fatty acid binding protein FABP4. A small molecular ligand for FABP4 that blocks the binding of endogenous ligands may be developed into a drug for the treatment of type-2 diabetes. Using NMR spectroscopy, we screened FABP4 for low-affinity binders in a diversity library consisting of small soluble scaffolds, which yielded 52 initial hits in total. The potencies of these hits were ranked, and crystal structures of FABP4 complexes for two of the hits were obtained. The structural data were subsequently used to direct similarity searches for available analogues, as well as chemical synthesis of 12 novel analogues. In this way, a series of three selective FABP4 ligands with attractive pharmacochemical profiles and potencies of 10 microM or better was obtained.


Subject(s)
Carrier Proteins/metabolism , Drug Evaluation, Preclinical/methods , Neoplasm Proteins , Tumor Suppressor Proteins , Amino Acid Sequence , Carrier Proteins/chemistry , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Kinetics , Ligands , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Sequence Homology, Amino Acid , Structure-Activity Relationship
19.
Drug Discov Today ; 7(8): 471-8, 2002 Apr 15.
Article in English | MEDLINE | ID: mdl-11965396

ABSTRACT

Structure-based screening represents an integrated approach for the identification and optimization of hits by the combined use of nuclear magnetic resonance (NMR) spectroscopy, homology modeling and X-ray crystallography. A general feature of the methodology is the introduction of structure-based methods (NMR, modeling and X-ray) early in the drug discovery process to optimize hits in terms of their affinities and specificities. This approach promises to deliver leads with improved physicochemical properties as compared with leads generated from a traditional HTS program. This review presents examples of structure-based screening from published and in-house drug discovery projects.


Subject(s)
Drug Design , Drug Evaluation, Preclinical/methods , Pharmaceutical Preparations/chemistry , Crystallography, X-Ray , Ligands , Magnetic Resonance Spectroscopy , Pharmaceutical Preparations/chemical synthesis , Structure-Activity Relationship
20.
J Am Chem Soc ; 124(11): 2446-7, 2002 Mar 20.
Article in English | MEDLINE | ID: mdl-11890787

ABSTRACT

A new method for site-selective screening by NMR is presented. The core of the new method is the dual amino acid sequence specific labeling technique. Amino acid X is labeled with (13)C and amino acid Y is labeled with (15)N. Provided only one XY pair occurs in the amino acid sequence, only one signal in the 1D carbonyl (13)C spectrum will display a splitting due to the (1)J(C'N) coupling. Using this labeling strategy it is possible to screen selectively for binding to a selected epitope without the need for sequence specific assignments. An HNCO spectrum (1D or 2D) can be used either directly as a screening experiment or indirectly to identify what signals to monitor in a 2D (1)H-(15)N correlation spectrum. Chemical shift perturbations upon addition of a potential ligand are easily detected even for large proteins due to the reduced spectral complexity resulting from the use of a selectively labeled sample. The new technique is demonstrated on the human adipocyte fatty acid binding protein FABP-4. Due to the reduced spectral complexity, the method should be applicable to larger proteins than are conventional methods.


Subject(s)
Amino Acids/analysis , Nuclear Magnetic Resonance, Biomolecular/methods , Binding Sites , Carbon Isotopes , Nitrogen Isotopes , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...