Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Molecules ; 28(4)2023 Feb 16.
Article in English | MEDLINE | ID: mdl-36838885

ABSTRACT

Targeting cancer cells that are highly dependent on the nicotinamide adenine dinucleotide (NAD+) metabolite is a promising therapeutic strategy. Nicotinamide phosphoribosyltransferase (NAMPT) is the rate-limiting enzyme catalyzing NAD+ production. Despite the high efficacy of several developed NAMPT inhibitors (i.e., FK866 (APO866)) in preclinical studies, their clinical activity was proven to be limited. Here, we report the synthesis of new NAMPT Inhibitors, JJ08, FEI191 and FEI199, which exhibit a broad anticancer activity in vitro. Results show that these compounds are potent NAMPT inhibitors that deplete NAD+ and NADP(H) after 24 h of drug treatment, followed by an increase in reactive oxygen species (ROS) accumulation. The latter event leads to ATP loss and mitochondrial depolarization with induction of apoptosis and necrosis. Supplementation with exogenous NAD+ precursors or catalase (ROS scavenger) abrogates the cell death induced by the new compounds. Finally, in vivo administration of the new NAMPT inhibitors in a mouse xenograft model of human Burkitt lymphoma delays tumor growth and significantly prolongs mouse survival. The most promising results are collected with JJ08, which completely eradicates tumor growth. Collectively, our findings demonstrate the efficient anticancer activity of the new NAMPT inhibitor JJ08 and highlight a strong interest for further evaluation of this compound in hematological malignancies.


Subject(s)
Enzyme Inhibitors , Hematologic Neoplasms , Nicotinamide Phosphoribosyltransferase , Animals , Humans , Mice , Cell Line, Tumor , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Hematologic Neoplasms/drug therapy , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Reactive Oxygen Species
2.
J Enzyme Inhib Med Chem ; 37(1): 1773-1811, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35758198

ABSTRACT

The haem enzyme indoleamine 2,3-dioxygenase 1 (IDO1) catalyses the rate-limiting step in the kynurenine pathway of tryptophan metabolism and plays an essential role in immunity, neuronal function, and ageing. Expression of IDO1 in cancer cells results in the suppression of an immune response, and therefore IDO1 inhibitors have been developed for use in anti-cancer immunotherapy. Here, we report an extension of our previously described highly efficient haem-binding 1,2,3-triazole and 1,2,4-triazole inhibitor series, the best compound having both enzymatic and cellular IC50 values of 34 nM. We provide enzymatic inhibition data for almost 100 new compounds and X-ray diffraction data for one compound in complex with IDO1. Structural and computational studies explain the dramatic drop in activity upon extension to pocket B, which has been observed in diverse haem-binding inhibitor scaffolds. Our data provides important insights for future IDO1 inhibitor design.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Triazoles , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Heme , Triazoles/chemistry , Triazoles/pharmacology
3.
Bioorg Med Chem Lett ; 26(17): 4330-3, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27469130

ABSTRACT

Indoleamine 2,3-dioxygenase 2 (IDO2) is a potential therapeutic target for the treatment of diseases that involve immune escape such as cancer. In contrast to IDO1, only a very limited number of inhibitors have been described for IDO2 due to inherent difficulties in expressing and purifying a functionally active, soluble form of the enzyme. Starting from our previously discovered highly efficient 4-aryl-1,2,3-triazole IDO1 inhibitor scaffold, we used computational structure-based methods to design inhibitors of IDO2 which we then tested in cellular assays. Our approach yielded low molecular weight inhibitors of IDO2, the most active displaying an IC50 value of 51µM for mIDO2, and twofold selectivity over hIDO1. These compounds could be useful as molecular probes to investigate the biological role of IDO2, and could inspire the design of new IDO2 inhibitors.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Triazoles/chemical synthesis , Catalytic Domain , Drug Design , Enzyme Activation/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Molecular Weight , Triazoles/chemistry , Triazoles/pharmacology
4.
Eur J Med Chem ; 239: 114504, 2022 Sep 05.
Article in English | MEDLINE | ID: mdl-35724566

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal diseases for which chemotherapy has not been very successful yet. FK866 ((E)-N-(4-(1-benzoylpiperidin-4-yl)butyl)-3-(pyridin-3-yl)acrylamide) is a well-known NAMPT (nicotinamide phosphoribosyltransferase) inhibitor with anti-cancer activities, but it failed in phase II clinical trials. We found that FK866 shows anti-proliferative activity in three PDAC cell lines, as well as in Jurkat T-cell leukemia cells. More than 50 FK866 analogues were synthesized that introduce substituents on the phenyl ring of the piperidine benzamide group of FK866 and exchange its buta-1,4-diyl tether for 1-oxyprop-3-yl, (E)-but-2-en-1,4-diyl and 2- and 3-carbon tethers. The pyridin-3-yl moiety of FK866 was exchanged for chlorinated and fluorinated analogues and for pyrazin-2-yl and pyridazin-4-yl groups. Several compounds showed low nanomolar or sub-nanomolar cell growth inhibitory activity. Our best cell anti-proliferative compounds were the 2,4,6-trimethoxybenzamide analogue of FK866 ((E)-N-(4-(1-(2,4,6-trimethoxybenzoyl)piperidin-4-yl)butyl)-3-(pyridin-3-yl)acrylamide) (9), the 2,6-dimethoxybenzamide (8) and 2-methoxybenzamide (4), which exhibited an IC50 of 0.16 nM, 0.004 nM and 0.08 nM toward PDAC cells, respectively.


Subject(s)
Acrylamides , Antineoplastic Agents , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Piperidines , Acrylamides/chemistry , Acrylamides/pharmacology , Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Cytokines , Humans , Pancreatic Neoplasms/drug therapy , Piperidines/chemistry , Piperidines/pharmacology , Pancreatic Neoplasms
5.
J Med Chem ; 64(4): 2205-2227, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33557523

ABSTRACT

The heme enzyme indoleamine 2,3-dioxygenase 1 (IDO1) plays an essential role in immunity, neuronal function, and aging through catalysis of the rate-limiting step in the kynurenine pathway of tryptophan metabolism. Many IDO1 inhibitors with different chemotypes have been developed, mainly targeted for use in anti-cancer immunotherapy. Lead optimization of direct heme iron-binding inhibitors has proven difficult due to the remarkable selectivity and sensitivity of the heme-ligand interactions. Here, we present experimental data for a set of closely related small azole compounds with more than 4 orders of magnitude differences in their inhibitory activities, ranging from millimolar to nanomolar levels. We investigate and rationalize their activities based on structural data, molecular dynamics simulations, and density functional theory calculations. Our results not only expand the presently known four confirmed chemotypes of sub-micromolar heme binding IDO1 inhibitors by two additional scaffolds but also provide a model to predict the activities of novel scaffolds.


Subject(s)
Azoles/pharmacology , Enzyme Inhibitors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Azoles/chemical synthesis , Azoles/metabolism , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/metabolism , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Molecular Dynamics Simulation , Molecular Structure , Protein Binding , Quantitative Structure-Activity Relationship
6.
J Med Chem ; 62(19): 8784-8795, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31525930

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) catalyzes the rate-limiting step in the kynurenine pathway of tryptophan metabolism, which is involved in immunity, neuronal function, and aging. Its implication in pathologies such as cancer and neurodegenerative diseases has stimulated the development of IDO1 inhibitors. However, negative phase III clinical trial results of the IDO1 inhibitor epacadostat in cancer immunotherapy call for a better understanding of the role and the mechanisms of IDO1 inhibition. In this work, we investigate the molecular inhibition mechanisms of four known IDO1 inhibitors and of two quinones in detail, using different experimental and computational approaches. We also determine for the first time the X-ray structure of the highly efficient 1,2,3-triazole inhibitor MMG-0358. Based on our results and a comprehensive literature overview, we propose a classification scheme for IDO1 inhibitors according to their inhibition mechanism, which will be useful for further developments in the field.


Subject(s)
Enzyme Inhibitors/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Crystallography, X-Ray , Density Functional Theory , Enzyme Inhibitors/chemistry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Molecular Conformation , Oximes/chemistry , Oximes/metabolism , Protein Binding , Quinones/chemistry , Quinones/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sulfonamides/chemistry , Sulfonamides/metabolism , Temperature , Triazoles/chemistry , Triazoles/metabolism
7.
J Med Chem ; 58(24): 9421-37, 2015 Dec 24.
Article in English | MEDLINE | ID: mdl-25970480

ABSTRACT

Since the discovery of indoleamine 2,3-dioxygenase 1 (IDO1) as an attractive target for anticancer therapy in 2003, the search for inhibitors has been intensely pursued both in academia and in pharmaceutical companies. Many novel IDO1 inhibitor scaffolds have been described, and a few potent compounds have entered clinical trials. However, a significant number of the reported compounds contain problematic functional groups, suggesting that enzyme inhibition could be the result of undesirable side reactions instead of selective binding to IDO1. Here, we describe issues in the employed experimental protocols, review and classify reported IDO1 inhibitors, and suggest different approaches for confirming viable inhibitor scaffolds.


Subject(s)
Enzyme Inhibitors/chemistry , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Animals , Cell Survival/drug effects , Drug Discovery , Enzyme Assays , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Structure-Activity Relationship , Tryptophan/analogs & derivatives , Tryptophan/chemistry , Tryptophan/metabolism , Tryptophan/pharmacology
8.
Eur J Med Chem ; 84: 284-301, 2014 Sep 12.
Article in English | MEDLINE | ID: mdl-25036789

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is a key regulator of immune responses and therefore an important therapeutic target for the treatment of diseases that involve pathological immune escape, such as cancer. Here, we describe a robust and sensitive high-throughput screen (HTS) for IDO1 inhibitors using the Prestwick Chemical Library of 1200 FDA-approved drugs and the Maybridge HitFinder Collection of 14,000 small molecules. Of the 60 hits selected for follow-up studies, 14 displayed IC50 values below 20 µM under the secondary assay conditions, and 4 showed an activity in cellular tests. In view of the high attrition rate we used both experimental and computational techniques to identify and to characterize compounds inhibiting IDO1 through unspecific inhibition mechanisms such as chemical reactivity, redox cycling, or aggregation. One specific IDO1 inhibitor scaffold, the imidazole antifungal agents, was chosen for rational structure-based lead optimization, which led to more soluble and smaller compounds with micromolar activity.


Subject(s)
Antifungal Agents/pharmacology , Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Animals , Antifungal Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mice , Molecular Dynamics Simulation , Molecular Structure , Structure-Activity Relationship
9.
J Med Chem ; 55(11): 5270-90, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22616902

ABSTRACT

Indoleamine 2,3-dioxygenase 1 (IDO1) is an important therapeutic target for the treatment of diseases such as cancer that involve pathological immune escape. Starting from the scaffold of our previously discovered IDO1 inhibitor 4-phenyl-1,2,3-triazole, we used computational structure-based methods to design more potent ligands. This approach yielded highly efficient low molecular weight inhibitors, the most active being of nanomolar potency both in an enzymatic and in a cellular assay, while showing no cellular toxicity and a high selectivity for IDO1 over tryptophan 2,3-dioxygenase (TDO). A quantitative structure-activity relationship based on the electrostatic ligand-protein interactions in the docked binding modes and on the quantum chemically derived charges of the triazole ring demonstrated a good explanatory power for the observed activities.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Triazoles/chemical synthesis , Animals , Catalytic Domain , Cell Line , Drug Design , Enzyme Assays , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Imidazoles/chemical synthesis , Imidazoles/chemistry , Imidazoles/pharmacology , Ligands , Mice , Models, Molecular , Protein Binding , Protein Conformation , Quantitative Structure-Activity Relationship , Quantum Theory , Static Electricity , Triazoles/chemistry , Triazoles/pharmacology , Tryptophan Oxygenase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL