Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
Add more filters










Publication year range
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.
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
4.
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
5.
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
6.
Eur J Med Chem ; 150: 457-478, 2018 Apr 25.
Article in English | MEDLINE | ID: mdl-29547833

ABSTRACT

We have synthesized a wide array of structurally related amphiphilic compounds, containing a functionalized pyrrolidine polar group coupled to different ether-linked hydrocarbon chains, to generate novel structures with antitumor activity. These newly synthesized amphiphilic pyrrolidine-derived compounds were classified in three different sub-libraries regarding the number of hydroxyl groups substituting the pyrrolidine moiety at C3 and C4. Pyrrolidine compounds with one or none hydroxyl groups showed a potent cell killing activity against pancreatic cancer cells, but they lacked selectivity for tumor cells. Pyrrolidine compounds with two hydroxyl groups induced cell death in a wide variety of pancreatic cancer cell lines, and they were somewhat less cytotoxic to normal non-tumor cells. Among these latter compounds, the diol-derived pyrrolidine 20 ((2R,3R,4S)-2-{(9Z)-hexadec-9-en-1-yloxy]methyl}pyrrolidine-3,4-diol) induced autophagy and a potent apoptotic response in pancreatic ductal adenocarcinoma cells, which was inhibited by Bcl-XL overexpression and by caspase inhibition, in a way similar to that of the amphiphilic ether lipid edelfosine, with which it was compared. Pharmacological and genetic inhibition of autophagy potentiated 20-mediated apoptosis. These structure-activity relationship studies point out the importance of the diol polar group and aliphatic side chain of 20 in promoting apoptosis against pancreatic cancer cells in a rather controlled way, and some additional subtle modifications were found to be potential modulators of the cytotoxic activity.


Subject(s)
Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/drug therapy , Pyrrolidines/pharmacology , Surface-Active Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Pancreatic Neoplasms/pathology , Pyrrolidines/chemical synthesis , Pyrrolidines/chemistry , Structure-Activity Relationship , Surface-Active Agents/chemical synthesis , Surface-Active Agents/chemistry
7.
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
8.
Chemistry ; 22(12): 4196-205, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26864218

ABSTRACT

Trimethylsilyl, triethylsilyl, tert-butyldimethylsilyl, and triisopropylsilyl 2-methylprop-2-ene-1-sulfinates were prepared through (CuOTf)2⋅C6H6-catalyzed sila-ene reactions of the corresponding methallylsilanes with SO2 at 50 °C. Sterically hindered, epimerizable, and base-sensitive alcohols gave the corresponding silyl ethers in high yields and purities at room temperature and under neutral conditions. As the byproducts of the silylation reaction (SO2 +isobutylene) are volatile, the workup was simplified to solvent evaporation. The developed method can be employed for the chemo- and regioselective semiprotection of polyols and glycosides and for the silylation of unstable aldols. The high reactivity of the developed reagents is shown by the synthesis of sterically hindered per-O-tert-butyldimethylsilyl-α-D-glucopyranose, the X-ray crystallographic analysis of which is the first for a per-O-silylated hexopyranose. The per-O-silylation of polyols, hydroxy carboxylic acids, and carbohydrates with trimethylsilyl 2-methylprop-2-ene-1-sulfinate was coupled with the GC analysis of nonvolatile polyhydroxy compounds both qualitatively and quantitatively.


Subject(s)
Carbohydrates/chemistry , Polymers/chemistry , Silanes/chemistry , Sulfinic Acids/chemistry , Alcohols/chemistry , Carboxylic Acids/chemistry , Catalysis , Ethers/chemistry , Hexoses/chemistry , Solvents
9.
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
10.
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
11.
Autophagy ; 10(4): 603-17, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24487122

ABSTRACT

APO866, an inhibitor of NAD biosynthesis, exhibits potent antitumor properties in various malignancies. Recently, it has been shown that APO866 induces apoptosis and autophagy in human hematological cancer cells, but the role of autophagy in APO866-induced cell death remains unclear. Here, we report studies on the molecular mechanisms underlying APO866-induced cell death with emphasis on autophagy. Treatment of leukemia and lymphoma cells with APO866 induced both autophagy, as evidenced by an increase in autophagosome formation and in SQSTM1/p62 degradation, but also increased caspase activation as revealed by CASP3/caspase 3 cleavage. As an underlying mechanism, APO866-mediated autophagy was found to deplete CAT/catalase, a reactive oxygen species (ROS) scavenger, thus promoting ROS production and cell death. Inhibition of autophagy by ATG5 or ATG7 silencing prevented CAT degradation, ROS production, caspase activation, and APO866-induced cell death. Finally, supplementation with exogenous CAT also abolished APO866 cytotoxic activity. Altogether, our results indicated that autophagy is essential for APO866 cytotoxic activity on cells from hematological malignancies and also indicate an autophagy-dependent CAT degradation, a novel mechanism for APO866-mediated cell killing. Autophagy-modulating approaches could be a new way to enhance the antitumor activity of APO866 and related agents.


Subject(s)
Acrylamides/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Leukemia/pathology , Lymphoma/pathology , NAD/antagonists & inhibitors , Piperidines/pharmacology , Caspase 3/metabolism , Cell Line, Tumor , Humans , Leukemia/drug therapy , Lymphoma/drug therapy , NAD/biosynthesis , Reactive Oxygen Species/metabolism
12.
Org Lett ; 16(2): 604-7, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24387286

ABSTRACT

An efficient method has been developed for the direct amidification of carboxylic acids via sulfinylamides preformed in situ by the reaction of pure amines with prop-2-ene-1-sulfinyl chloride. The method can be applied to aliphatic acids, including pivalic acid, aromatic acids, and primary and secondary amines. It is compatible with acids bearing unprotected alcohol, phenol, and ketone moieties and applicable to the synthesis of peptides. It does not induce their α-epimerization.


Subject(s)
Amides/chemical synthesis , Amines/chemistry , Carboxylic Acids/chemistry , Peptides/chemical synthesis , Sulfoxides/chemistry , Amides/chemistry , Ketones , Molecular Structure , Peptides/chemistry
13.
Leuk Lymphoma ; 55(9): 2141-50, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24283753

ABSTRACT

APO866 is an inhibitor of nicotinamide adenine dinucleotide (NAD) biosynthesis that exhibits potent anti-lymphoma activity. Rituximab (RTX), an anti-CD20 antibody, kills lymphoma cells by direct apoptosis and antibody- and complement-dependent cell-mediated cytotoxicities, and has clinical efficacy in non-Hodgkin cell lymphomas. In the present study, we evaluated whether RTX could potentiate APO866-induced human B-lymphoma cell death and shed light on death-mediated mechanisms associated with this drug combination. We found that RTX significantly increases APO866-induced death in lymphoma cells from patients and lines. Mechanisms include enhancement of autophagy-mediated cell death, activation of caspase 3 and exacerbation of mitochondrial depolarization, but not increase of reactive oxygen species (ROS) production, when compared with those induced by each drug alone. In vivo, combined administration of APO866 with RTX in a laboratory model of human aggressive lymphoma significantly decreased tumor burden and prolonged survival over single-agent treatment. Our study demonstrates that the combination of RTX and APO866 optimizes B-cell lymphoma apoptosis and therapeutic efficacy over both compounds administered separately.


Subject(s)
Acrylamides/pharmacology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antineoplastic Agents/pharmacology , Piperidines/pharmacology , Animals , Apoptosis/drug effects , Biosynthetic Pathways/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Humans , Lymphoma/drug therapy , Lymphoma/mortality , Lymphoma/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, SCID , NAD/metabolism , Reactive Oxygen Species/metabolism , Rituximab , Xenograft Model Antitumor Assays
14.
Org Lett ; 15(10): 2550-3, 2013 May 17.
Article in English | MEDLINE | ID: mdl-23642170

ABSTRACT

An efficient method has been developed for the preparation of yet unknown acyclic mixed anhydrides of carboxylic and sulfinic acids. Sterically hindered 2-methylbut-3-ene-2-sulfinyl carboxylates add primary and secondary amines preferentially onto the carbonyl moieties realizing a new method for the one-pot preparation of carboxamides. It uses 1:1 mixtures of carboxylic acids and amines without a base, requires no excess of reagents, and liberates only volatile coproducts. Protected di- and tripeptides have been prepared in solution without epimerization by application of this method.


Subject(s)
Amides/chemistry , Amides/chemical synthesis , Amines/chemistry , Anhydrides/chemistry , Carboxylic Acids/chemistry , Sulfonium Compounds/chemistry , Molecular Structure
16.
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
18.
Carbohydr Res ; 356: 110-4, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22129839

ABSTRACT

Four branched tetra- and pentahydroxylated azepanes have been synthesized from a common azepane precursor through dihydroxylation followed by deoxygenation. They have been assayed as glycosidase inhibitors on a panel of 22 glycosidases and one methylated azepane displayed selective, competitive, and moderate inhibition toward bovine kidney α-L-fucosidase.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Imino Sugars/chemical synthesis , alpha-L-Fucosidase/antagonists & inhibitors , Animals , Cattle , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Imino Sugars/pharmacology , Kidney/enzymology , Magnetic Resonance Spectroscopy , Molecular Structure , Oxygen/chemistry , Plants/enzymology , Stereoisomerism , Yeasts/enzymology , alpha-L-Fucosidase/metabolism
19.
Bioorg Med Chem ; 19(24): 7720-7, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079865

ABSTRACT

New derivatives of 1,4-dideoxy-1,4-imino-D-ribitol have been prepared and evaluated for their cytotoxicity on solid and haematological malignancies. 1,4-Dideoxy-5-O-[(9Z)-octadec-9-en-1-yl]-1,4-imino-D-ribitol (13, IC(50) ∼2 µM) and its C(18)-analogues (IC(50) <10 µM) are cytotoxic toward SKBR3 (breast cancer) cells. 13 also inhibits (IC(50) ∼8 µM) growth of JURKAT cells.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Ribitol/analogs & derivatives , Ribitol/pharmacology , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Female , Humans , Jurkat Cells , Neoplasms/drug therapy , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...