Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Hemasphere ; 8(5): e77, 2024 May.
Article in English | MEDLINE | ID: mdl-38716146

ABSTRACT

The mainstay of acute myeloid leukemia (AML) treatment still relies on traditional chemotherapy, with a survival rate of approximately 30% for patients under 65 years of age and as low as 5% for those beyond. This unfavorable prognosis primarily stems from frequent relapses, resistance to chemotherapy, and limited approved targeted therapies for specific AML subtypes. Around 70% of all AML cases show overexpression of the transcription factor HOXA9, which is associated with a poor prognosis, increased chemoresistance, and higher relapse rates. However, direct targeting of HOXA9 in a clinical setting has not been achieved yet. The dysregulation caused by the leukemic HOXA9 transcription factor primarily results from its binding activity to DNA, leading to differentiation blockade. Our previous investigations have identified two HOXA9/DNA binding competitors, namely DB1055 and DB818. We assessed their antileukemic effects in comparison to HOXA9 knockdown or cytarabine treatment. Using human AML cell models, DB1055 and DB818 induced in vitro cell growth reduction, death, differentiation, and common transcriptomic deregulation but did not impact human CD34+ bone marrow cells. Furthermore, DB1055 and DB818 exhibited potent antileukemic activities in a human THP-1 AML in vivo model, leading to the differentiation of monocytes into macrophages. In vitro assays also demonstrated the efficacy of DB1055 and DB818 against AML blasts from patients, with DB1055 successfully reducing leukemia burden in patient-derived xenografts in NSG immunodeficient mice. Our findings indicate that inhibiting HOXA9/DNA interaction using DNA ligands may offer a novel differentiation therapy for the future treatment of AML patients dependent on HOXA9.

2.
Cancers (Basel) ; 11(6)2019 Jun 17.
Article in English | MEDLINE | ID: mdl-31213012

ABSTRACT

HOXA9 (Homeobox A9) is a homeotic transcription factor known for more than two decades to be associated with leukemia. The expression of HOXA9 homeoprotein is associated with anterior-posterior patterning during embryonic development, and its expression is then abolished in most adult cells, with the exception of hematopoietic progenitor cells. The oncogenic function of HOXA9 was first assessed in human acute myeloid leukemia (AML), particularly in the mixed-phenotype associated lineage leukemia (MPAL) subtype. HOXA9 expression in AML is associated with aggressiveness and a poor prognosis. Since then, HOXA9 has been involved in other hematopoietic malignancies and an increasing number of solid tumors. Despite this, HOXA9 was for a long time not targeted to treat cancer, mainly since, as a transcription factor, it belongs to a class of protein long considered to be an "undruggable" target; however, things have now evolved. The aim of the present review is to focus on the different aspects of HOXA9 targeting that could be achieved through multiple ways: (1) indirectly, through the inhibition of its expression, a strategy acting principally at the epigenetic level; or (2) directly, through the inhibition of its transcription factor function by acting at either the protein/protein interaction or the protein/DNA interaction interfaces.

3.
J Med Chem ; 62(3): 1306-1329, 2019 02 14.
Article in English | MEDLINE | ID: mdl-30645099

ABSTRACT

Most transcription factors were for a long time considered as undruggable targets because of the absence of binding pockets for direct targeting. HOXA9, implicated in acute myeloid leukemia, is one of them. To date, only indirect targeting of HOXA9 expression or multitarget HOX/PBX protein/protein interaction inhibitors has been developed. As an attractive alternative by inhibiting the DNA binding, we selected a series of heterocyclic diamidines as efficient competitors for the HOXA9/DNA interaction through binding as minor groove DNA ligands on the HOXA9 cognate sequence. Selected DB818 and DB1055 compounds altered HOXA9-mediated transcription in luciferase assays, cell survival, and cell cycle, but increased cell death and granulocyte/monocyte differentiation, two main HOXA9 functions also highlighted using transcriptomic analysis of DB818-treated murine Hoxa9-transformed hematopoietic cells. Altogether, these data demonstrate for the first time the propensity of sequence-selective DNA ligands to inhibit HOXA9/DNA binding both in vitro and in a murine Hoxa9-dependent leukemic cell model.


Subject(s)
DNA/drug effects , Heterocyclic Compounds/pharmacology , Homeodomain Proteins/antagonists & inhibitors , Leukemia/pathology , Models, Biological , Cell Death/drug effects , Cell Proliferation/drug effects , DNA/chemistry , Drug Design , Gene Expression/drug effects , Heterocyclic Compounds/chemistry , Leukemia/genetics , Ligands
4.
Molecules ; 23(6)2018 06 19.
Article in English | MEDLINE | ID: mdl-29921764

ABSTRACT

Transcription factors are involved in a large number of human diseases such as cancers for which they account for about 20% of all oncogenes identified so far. For long time, with the exception of ligand-inducible nuclear receptors, transcription factors were considered as "undruggable" targets. Advances knowledge of these transcription factors, in terms of structure, function (expression, degradation, interaction with co-factors and other proteins) and the dynamics of their mode of binding to DNA has changed this postulate and paved the way for new therapies targeted against transcription factors. Here, we discuss various ways to target transcription factors in cancer models: by modulating their expression or degradation, by blocking protein/protein interactions, by targeting the transcription factor itself to prevent its DNA binding either through a binding pocket or at the DNA-interacting site, some of these inhibitors being currently used or evaluated for cancer treatment. Such different targeting of transcription factors by small molecules is facilitated by modern chemistry developing a wide variety of original molecules designed to specifically abort transcription factor and by an increased knowledge of their pathological implication through the use of new technologies in order to make it possible to improve therapeutic control of transcription factor oncogenic functions.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Delivery Systems/methods , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/drug effects , Animals , Humans , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Transcription Factors/metabolism
5.
Mol Divers ; 22(3): 637-646, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29557543

ABSTRACT

Novel nitro (3a-3f)- and amino (4a-4f and 5a-5f)-substituted 2-benzimidazolyl and 2-benzothiazolyl benzo[b]thieno-2-carboxamides were designed and synthesized as potential antibacterial agents. The antibacterial activity of these compounds has been evaluated against Gram-positive (Staphylococcus aureus and Enterococcus faecalis) and Gram-negative bacteria (Escherichia coli and Moraxella catarrhalis). The most promising antibacterial activity was observed for the nitro- and amino-substituted benzimidazole derivatives 3a, 4a, 5a and 5b with MICs 2-8 [Formula: see text]. Additionally, compounds with inferior antibacterial activity were further tested for their antiproliferative activity in vitro against three human cancer cell lines. Amino-substituted benzothiazole hydrochloride salt 5d displayed the most pronounced and selective activity against the MCF-7 cell line with an [Formula: see text] of 40 nM. Furthermore, DNA binding experiments of selected derivatives indicated that DNA cannot be considered as a primary biological target for this type of compounds.


Subject(s)
Anti-Bacterial Agents , Antineoplastic Agents , Benzimidazoles , Benzothiazoles , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Benzothiazoles/chemistry , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/metabolism , Enterococcus faecalis/drug effects , Enterococcus faecalis/growth & development , Escherichia coli/drug effects , Escherichia coli/growth & development , Humans , Microbial Sensitivity Tests , Moraxella catarrhalis/drug effects , Moraxella catarrhalis/growth & development , Staphylococcus aureus/drug effects , Staphylococcus aureus/growth & development
6.
Bioorg Med Chem ; 26(8): 1950-1960, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29519603

ABSTRACT

In this paper novel isoindolines substituted with cyano and amidino benzimidazoles and benzothiazoles were synthesized as new potential anti-cancer agents. The new structures were evaluated for antiproliferative activity, cell cycle changes, cell death, as well as DNA binding and topoisomerase inhibition properties on selected compounds. Results showed that all tested compounds exerted antitumor activity, especially amidinobenzothiazole and amidinobenzimidazole substituted isoindolin-1-ones and benzimidazole substituted 1-iminoisoindoline that showed antiproliferative effect in the submicromolar range. Moreover, the DNA-binding properties of selected compounds were evaluated by biophysical and biochemical approaches including thermal denaturation studies, circular dichroism spectra analyses and topoisomerase I/II inhibition assays and results identified some of them as strong DNA ligands, harboring or not additional topoisomerase II inhibition and able to locate in the nucleus as determined by fluorescence microscopy. In conclusion, we evidenced novel cyano- and amidino-substituted isoindolines coupled with benzimidazoles and benzothiazoles as topoisomerase inhibitors and/or DNA binding compounds with potent antitumor activities.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzimidazoles/chemistry , Benzothiazoles/chemistry , DNA/metabolism , Isoindoles/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Circular Dichroism , DNA/chemistry , DNA Topoisomerases, Type I/chemistry , DNA Topoisomerases, Type I/metabolism , DNA Topoisomerases, Type II/chemistry , DNA Topoisomerases, Type II/metabolism , Drug Screening Assays, Antitumor , Humans , Isoindoles/metabolism , Isoindoles/pharmacology , MCF-7 Cells , Microscopy, Fluorescence , Structure-Activity Relationship
7.
J Med Chem ; 60(11): 4665-4679, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28463515

ABSTRACT

Aberrant DNA hypermethylation of promoter of tumor suppressor genes is commonly observed in cancer, and its inhibition by small molecules is promising for their reactivation. Here we designed bisubstrate analogues-based inhibitors, by mimicking each substrate, the S-adenosyl-l-methionine and the deoxycytidine, and linking them together. This approach resulted in quinazoline-quinoline derivatives as potent inhibitors of DNMT3A and DNMT1, some showing certain isoform selectivity. We highlighted the importance of (i) the nature and rigidity of the linker between the two moieties for inhibition, as (ii) the presence of the nitrogen on the quinoline group, and (iii) of a hydrophobic group on the quinazoline. The most potent inhibitors induced demethylation of CDKN2A promoter in colon carcinoma HCT116 cells and its reactivation after 7 days of treatment. Furthermore, in a leukemia cell model system, we found a correlation between demethylation of the promoter induced by the treatment, chromatin opening at the promoter, and the reactivation of a reporter gene.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Neoplasms/enzymology , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1 , DNA Methylation , DNA Methyltransferase 3A , Genes, Tumor Suppressor , Humans , Neoplasms/pathology , Substrate Specificity
8.
Eur J Med Chem ; 136: 468-479, 2017 Aug 18.
Article in English | MEDLINE | ID: mdl-28525845

ABSTRACT

Within this manuscript design, synthesis of novel 2-imidazolinyl substituted benzo[b]thieno-2-carboxamides bearing either benzimidazole or benzothiazole subunit and biological activity are presented and described. The antiproliferative activities were assessed in vitro on a panel of human cancer cell lines. Tested compounds showed moderate activity while cytotoxicity on normal fibroblasts was lower in comparison with 5-fluorouracile. The variations of 2-imidazolinyl substituent at heteroaromatic subunits in different positions led to different cytotoxic properties. The strongest selective activity against HeLa cells was observed for the benzothiazole derivative 4d with 2-imidazolinyl group at the benzo[b]thiophene subunit with a corresponding IC50 = 1.16 µM. Additionally, several biological experiments were performed to explain the mode of biological action. Fluorescence microscopy evidenced nuclear subcellular localization of compounds 3a, 4a and 4c. Additionally, detailed DNA binding studies confirmed a strong DNA groove binding for derivatives 4a and 4c while DNase I footprinting experiments evidenced sequence-selective binding of compound 4c in the A-T rich side. Furthermore, topoisomerase suppressive effect was for compounds 4a-4c.


Subject(s)
Amidines/pharmacology , Antineoplastic Agents/pharmacology , Benzimidazoles/pharmacology , Benzothiazoles/pharmacology , DNA, Neoplasm/drug effects , Thiophenes/pharmacology , Amidines/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Benzimidazoles/chemical synthesis , Benzimidazoles/chemistry , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Binding Sites/drug effects , Cell Proliferation/drug effects , DNA, Neoplasm/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Molecular Structure , Structure-Activity Relationship , Thiophenes/chemical synthesis , Thiophenes/chemistry , Tumor Cells, Cultured
9.
Eur J Med Chem ; 122: 530-545, 2016 Oct 21.
Article in English | MEDLINE | ID: mdl-27448912

ABSTRACT

We describe the synthesis, 3D-derived quantitative structure-activity relationship (QSAR), antiproliferative activity and DNA binding properties of a series of 2-amino, 5-amino and 2,5-diamino substituted benzimidazo[1,2-a]quinolines prepared by environmentally friendly uncatalyzed microwave assisted amination. The antiproliferative activities were assessed in vitro against colon, lung and breast carcinoma cell lines; activities ranged from submicromolar to micromolar. The strongest antiproliferative activity was demonstrated by 2-amino-substituted analogues, whereas 5-amino and or 2,5-diamino substituted derivatives resulted in much less activity. Derivatives bearing 4-methyl- or 3,5-dimethyl-1-piperazinyl substituents emerged as the most active. DNA binding properties and the mode of interaction of chosen substituted benzimidazo[1,2-a]quinolines prepared herein were studied using melting temperature studies, a series of spectroscopic studies (UV/Visible, fluorescence, and circular dichroism), and biochemical experiments (topoisomerase I-mediated DNA relaxation and DNase I footprinting experiments). Both compound 36 and its bis-quaternary iodide salt 37 intercalate between adjacent base pairs of the DNA helix while compound 33 presented a very weak topoisomerase I poisoning activity. A 3D-QSAR analysis was performed to identify hydrogen bonding properties, hydrophobicity, molecular flexibility and distribution of hydrophobic regions as these molecular properties had the highest impact on the antiproliferative activity against the three cell lines.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Benzimidazoles/chemistry , DNA/metabolism , Quantitative Structure-Activity Relationship , Quinolines/metabolism , Quinolines/pharmacology , Antineoplastic Agents/chemistry , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Quinolines/chemistry
10.
Int J Mol Sci ; 16(11): 26555-81, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26556350

ABSTRACT

DNA alkylating drugs have been used in clinics for more than seventy years. The diversity of their mechanism of action (major/minor groove; mono-/bis-alkylation; intra-/inter-strand crosslinks; DNA stabilization/destabilization, etc.) has undoubtedly major consequences on the cellular response to treatment. The aim of this review is to highlight the variety of established protein recognition of DNA adducts to then particularly focus on glyceraldehyde-3-phosphate dehydrogenase (GAPDH) function in DNA adduct interaction with illustration using original experiments performed with S23906-1/DNA adduct. The introduction of this review is a state of the art of protein/DNA adducts recognition, depending on the major or minor groove orientation of the DNA bonding as well as on the molecular consequences in terms of double-stranded DNA maintenance. It reviews the implication of proteins from both DNA repair, transcription, replication and chromatin maintenance in selective DNA adduct recognition. The main section of the manuscript is focusing on the implication of the moonlighting protein GAPDH in DNA adduct recognition with the model of the peculiar DNA minor groove alkylating and destabilizing drug S23906-1. The mechanism of action of S23906-1 alkylating drug and the large variety of GAPDH cellular functions are presented prior to focus on GAPDH direct binding to S23906-1 adducts.


Subject(s)
Alkylating Agents/pharmacology , DNA Adducts/metabolism , DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/metabolism , Alkylation , Cell Nucleus , Cytoplasm , DNA Damage , DNA Replication , Glyceraldehyde-3-Phosphate Dehydrogenase (Phosphorylating)/metabolism , Nucleic Acid Conformation , Protein Binding , Transcription Factors/metabolism
11.
Bioorg Med Chem Lett ; 25(21): 4927-4932, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26051649

ABSTRACT

DNA minor-groove-binding compounds have limited biological applications, in part due to problems with sequence specificity that cause off-target effects. A model to enhance specificity has been developed with the goal of preparing compounds that bind to two AT sites separated by G·C base pairs. Compounds of interest were probed using thermal melting, circular dichroism, mass spectrometry, biosensor-SPR, and molecular modeling methods. A new minor groove binder that can strongly and specifically recognize a single G·C base pair with flanking AT sequences has been prepared. This multi-site DNA recognition mode offers novel design principles to recognize entirely new DNA motifs.


Subject(s)
Base Pairing , Benzene Derivatives/chemistry , DNA/chemistry , Base Sequence , Molecular Docking Simulation , Molecular Structure
12.
Curr Top Med Chem ; 15(14): 1323-58, 2015.
Article in English | MEDLINE | ID: mdl-25866275

ABSTRACT

Transcription factors are recognized as the master regulators of gene expression. Interestingly, about 10% of the transcription factors described in mammals are up to date directly implicated in a very large number of human diseases. With the exception of ligand-inducible nuclear receptors, transcription factors have longtime been considered as "undruggable" targets for therapeutics. However, the significant breakthroughs in their protein biochemistry and interactions with DNA at the structural level, together with increasing needs for new targeted-approaches particularly in cancers, has changed this postulate and opened the way for targeting transcription factors. Along with a better knowledge of their specific DNA binding sequences by genome wide and high throughput sequencing assay, these informations make possible the potent targeting of the transcription factors by three approaches dependently of their mechanism of action. In this review, we discuss the different physicochemical interactions between the transcription factors and the DNA helix, and the protein/protein interactions within a transcription factor complex and their impacts on the DNA structure. In order to impair transcription factor activities, small molecules compounds can either act by direct interaction on the transcription factor, or by blocking the protein/protein interactions in a transcription complex, or by competing with the transcription factor itself and specifically targeting its cognate binding sequence. For this latter mode of transcription targeting, we pay special attention to the DNA intercalating, alkylating or groove binders for transcription factor/DNA binding modulation.


Subject(s)
DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Gene Expression Regulation/drug effects , Small Molecule Libraries/pharmacology , Transcription Factors/metabolism , Animals , Binding Sites/drug effects , DNA, Neoplasm/chemistry , Humans , Small Molecule Libraries/chemistry , Transcription Factors/chemistry
13.
J Biol Chem ; 290(10): 6293-302, 2015 Mar 06.
Article in English | MEDLINE | ID: mdl-25525263

ABSTRACT

Among the epigenetic marks, DNA methylation is one of the most studied. It is highly deregulated in numerous diseases, including cancer. Indeed, it has been shown that hypermethylation of tumor suppressor genes promoters is a common feature of cancer cells. Because DNA methylation is reversible, the DNA methyltransferases (DNMTs), responsible for this epigenetic mark, are considered promising therapeutic targets. Several molecules have been identified as DNMT inhibitors and, among the non-nucleoside inhibitors, 4-aminoquinoline-based inhibitors, such as SGI-1027 and its analogs, showed potent inhibitory activity. Here we characterized the in vitro mechanism of action of SGI-1027 and two analogs. Enzymatic competition studies with the DNA substrate and the methyl donor cofactor, S-adenosyl-l-methionine (AdoMet), displayed AdoMet non-competitive and DNA competitive behavior. In addition, deviations from the Michaelis-Menten model in DNA competition experiments suggested an interaction with DNA. Thus their ability to interact with DNA was established; although SGI-1027 was a weak DNA ligand, analog 5, the most potent inhibitor, strongly interacted with DNA. Finally, as 5 interacted with DNMT only when the DNA duplex was present, we hypothesize that this class of chemical compounds inhibit DNMTs by interacting with the DNA substrate.


Subject(s)
Aminoquinolines/chemistry , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methylation/genetics , Enzyme Inhibitors/chemistry , Pyrimidines/chemistry , Aminoquinolines/pharmacology , DNA/chemistry , DNA/genetics , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferases/genetics , Enzyme Inhibitors/therapeutic use , Epigenomics , Humans , Neoplasms/drug therapy , Neoplasms/genetics , Pyrimidines/pharmacology
14.
J Med Chem ; 57(24): 10329-42, 2014 Dec 26.
Article in English | MEDLINE | ID: mdl-25360689

ABSTRACT

A series of 6-methoxy-3,3,14-trimethyl-3,14-dihydro-7H-benzo[b]chromeno[6,5-g][1,8]naphthyridin-7-one (4), 13-aza derivatives of benzo[b]acronycine, the isomeric 5-methoxy-2,2,13-trimethyl-2,13-dihydro-6H-benzo[b]chromeno[7,6-g][1,8]naphthyridin-6-one (5), and related cis-diols mono- and diesters were designed and synthesized. Their in vitro and in vivo biological activities were evaluated. As previously observed in the acronycine series, esters were the most potent derivatives exhibiting submicromolar activities; among them monoesters are particularly active. Racemic diacetate 21 showed a strong activity against KB-3-1 cell lines and was selected for in vivo evaluation and proved to be active, inhibiting tumor growth by more than 80%. After separation of the two enantiomers, compounds 21a and 21b were also evaluated against C38 colon adenocarcinoma; their activities were found to be significantly different.


Subject(s)
Acronine/chemistry , Adenocarcinoma/drug therapy , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell/drug therapy , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/pharmacology , Naphthyridines/chemical synthesis , Naphthyridines/pharmacology , Adenocarcinoma/pathology , Animals , Carcinoma, Squamous Cell/pathology , Colonic Neoplasms/pathology , Drug Design , Drug Screening Assays, Antitumor , Electrophoretic Mobility Shift Assay , Humans , Inhibitory Concentration 50 , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Stereoisomerism , Structure-Activity Relationship , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
15.
Eur J Med Chem ; 87: 372-85, 2014 Nov 24.
Article in English | MEDLINE | ID: mdl-25282261

ABSTRACT

Novel phenyl-substituted (3a-3d, 4a, 5, 8a, 8b and 9a) and pyridyl-substituted (3e-3i, 4b, 8c-8e, 9b and 9c) isoindolines were prepared in the reaction of o-phthalaldehyde and corresponding substituted aromatic and heteroaromatic amines by modification of reaction conditions from low to high temperature and from neutral to acidic environment. The antiproliferative activity of chosen substituted isoindolines was assessed on a panel of tumour cell lines and normal human fibroblasts. The majority of tested compounds was active at the highest tested concentrations phenyl-substituted isoindolines 3a and 3b and pyridyl-substituted isoindoline 3g showed a selective effect at micromolar concentrations on HepG2 cell line in comparison with other tested tumour cell lines and normal human fibroblasts. The strongest yet non-selective effect was observed for the pyridyl-substituted isoindoline 8c. These isoindoline derivatives showed diverse mechanism of action on tumour cell death induction as compounds 3a and 8c probably induced mitotic catastrophe while compound 3b induced apoptosis. Indeed, DNA binding properties evidenced that compounds 8a, 8c and 8d bind to DNA as highly potent DNA intercalators. By contrast, compounds 3b, 3e, 3i, 4a and 5 did not target the DNA. At last, the phenyl-substituted compound 8b proved to be a strong DNA binding compound with sequence selective binding and without DNA intercalation profile.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , DNA/metabolism , Indoles/chemistry , Indoles/pharmacology , Pyridines/chemistry , Antineoplastic Agents/chemical synthesis , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Humans , Indoles/chemical synthesis
16.
Eur J Med Chem ; 80: 218-27, 2014 Jun 10.
Article in English | MEDLINE | ID: mdl-24780599

ABSTRACT

The synthesis of 5-amino substituted benzimidazo[1,2-a]quinolines prepared by microwave assisted amination from halogeno substituted precursor was described. The majority of compounds were active at micromolar concentrations against colon, lung and breast carcinoma cell lines in vitro. The N,N-dimethylaminopropyl 9 and piperazinyl substituted derivative 19 showed the most pronounced activity towards all of the three tested tumor cell lines, which could be correlated to the presence of another N heteroatom and its potential interactions with biological targets. The DNA binding studies, consisting of UV/Visible absorbency, melting temperature studies, and fluorescence and circular dichroism titrations, revealed that compounds 9, 19 and 20 bind to DNA as strong intercalators. The cellular distribution analysis, based on compounds' intrinsic fluorescence, showed that compound 20 does not enter the cell, while compounds 9 and 19 do, which is in agreement with their cytotoxic effects. Compound 9 efficiently targets the nucleus whereas 19, which also showed DNA intercalating properties in vitro, was mostly localised in the cytoplasm suggesting that the antitumor mechanism of action is DNA-independent.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Benzimidazoles/chemical synthesis , Benzimidazoles/pharmacology , Intercalating Agents/chemical synthesis , Intercalating Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Chemistry Techniques, Synthetic , Humans , Intercalating Agents/chemistry , Intercalating Agents/metabolism
17.
Biochemistry ; 53(7): 1218-27, 2014 Feb 25.
Article in English | MEDLINE | ID: mdl-24495039

ABSTRACT

DB1255 is a symmetrical diamidinophenyl-dithiophene that exhibits cellular activity by binding to DNA and inhibiting binding of ERG, an ETS family transcription factor that is commonly overexpressed or translocated in leukemia and prostate cancer [Nhili, R., Peixoto, P., Depauw, S., Flajollet, S., Dezitter, X., Munde, M. M., Ismail, M. A., Kumar, A., Farahat, A. A., Stephens, C. E., Duterque-Coquillaud, M., Wilson, W. D., Boykin, D. W., and David-Cordonnier, M. H. (2013) Nucleic Acids Res. 41, 125-138]. Because transcription factor inhibition is complex but is an attractive area for anticancer and antiparasitic drug development, we have evaluated the DNA interactions of additional derivatives of DB1255 to gain an improved understanding of the biophysical chemistry of complex function and inhibition. DNase I footprinting, biosensor surface plasmon resonance, and circular dichroism experiments show that DB1255 has an unusual and strong monomer binding mode in minor groove sites that contain a single GC base pair flanked by AT base pairs, for example, 5'-ATGAT-3'. Closely related derivatives, such as compounds with the thiophene replaced with furan or selenophane, bind very weakly to GC-containing sequences and do not have biological activity. DB1255 is selective for the ATGAT site; however, a similar sequence, 5'-ATGAC-3', binds DB1255 more weakly and does not produce a footprint. Molecular docking studies show that the two thiophene sulfur atoms form strong, bifurcated hydrogen bond-type interactions with the G-N-H sequence that extends into the minor groove while the amidines form hydrogen bonds to the flanking AT base pairs. The central dithiophene unit of DB1255 thus forms an excellent, but unexpected, single-GC base pair recognition module in a monomer minor groove complex.


Subject(s)
Amidines/chemistry , DNA/chemistry , Thiophenes/chemistry , Amidines/chemical synthesis , Amidines/metabolism , Amino Acid Sequence , Biosensing Techniques , DNA/metabolism , DNA Footprinting , Deoxyribonuclease I/metabolism , Guanine/chemistry , Guanine/metabolism , Models, Molecular , Surface Plasmon Resonance , Thiophenes/chemical synthesis , Thiophenes/metabolism , Transition Temperature
18.
Eur J Med Chem ; 71: 267-81, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24334150

ABSTRACT

A series of new anilides (2a-c, 4-7, 17a-c, 18) and quinolones (3a-b, 8a-b, 9a-b, 10-15, 19) with nitrogen-bearing substituents from benzo[b]thiophene and thieno[2,3-c]thiophene series are prepared. Benzo[b]thieno[2,3-c]- and thieno[3',2':4,5]thieno[2,3-c]quinolones (3a-b, 8a-b) are synthesized by the reaction of photochemical dehydrohalogenation from corresponding anilides. Anilides and quinolones were tested for the antiproliferative activity. Fused quinolones bearing protonated aminium group, quaternary ammonium group, N-methylated and protonated aminium group, amino and protonated amino group (8a, 9b, 10-12) showed very prominent anticancer activity, whereby the hydrochloride salt of N',N'-dimethylaminopropyl-substituted quinolone (14) was the most active one, having the IC50 concentration at submicromolar range in accordance with previous QSAR predictions. On the other hand, flexible anilides were among the less active. Chemometric analysis of investigated compounds was performed. 3D-derived QSAR analysis identified solubility, metabolitic stability and the possibility of the compound to be ionized at pH 4-8 as molecular properties that are positively correlated with anticancer activity of investigated compounds, while molecular flexibility, polarizability and sum of hydrophobic surface areas were found to be negatively correlated. Anilides 2a-b, 4-7 and quinolones 3a-b, 8a-b, 9b and 10-14 were evaluated for DNA binding propensities and topoisomerases I/II inhibition as part of their mechanism of action. Among the anilides, only compound 7 presented some DNA binding propensity whereas the quinolones 8b, 9b and 10-14 intercalate in the DNA base pairs, compounds 8b, 9b and 14 being the most efficient ones. The strongest DNA intercalators, compounds 8b, 9b and 14, were clearly distinguished from the other compounds according to their molecular descriptors by the PCA and PLS analysis.


Subject(s)
Anilides/chemistry , Anilides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Quinolones/chemistry , Quinolones/pharmacology , Cell Line, Tumor , Cytostatic Agents/chemistry , Cytostatic Agents/pharmacology , DNA/metabolism , Drug Screening Assays, Antitumor , Humans , Neoplasms/drug therapy , Nitrogen/chemistry , Quantitative Structure-Activity Relationship , Thiophenes/chemistry , Topoisomerase II Inhibitors/chemistry , Topoisomerase II Inhibitors/pharmacology
19.
Chembiochem ; 15(1): 68-79, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24323836

ABSTRACT

Heterocyclic diamidines are strong DNA minor-groove binders and have excellent antiparasitic activity. To extend the biological activity of these compounds, a series of arylimidamides (AIAs) analogues, which have better uptake properties in Leishmania and Trypanosoma cruizi than diamidines, was prepared. The binding of the AIAs to DNA was investigated by Tm , fluorescence displacement titration, circular dichroism, DNase I footprinting, biosensor surface plasmon resonance, X-ray crystallography and molecular modeling. These compounds form 1:1 complexes with AT sequences in the DNA minor groove, and the binding strength varies with substituent size, charge and polarity. These substituent-dependent structure and properties provide a SAR that can be used to estimate K values for binding to DNA in this series. The structural results and molecular modeling studies provide an explanation for the differences in binding affinities for AIAs.


Subject(s)
Amides/metabolism , DNA/metabolism , Amides/chemistry , Base Sequence , Binding Sites , Circular Dichroism , Crystallography, X-Ray , DNA/chemistry , Deoxyribonuclease I/metabolism , Leishmania/metabolism , Molecular Docking Simulation , Nucleic Acid Conformation , Substrate Specificity , Surface Plasmon Resonance , Transition Temperature , Trypanosoma cruzi/metabolism
20.
Eur J Med Chem ; 63: 882-91, 2013 May.
Article in English | MEDLINE | ID: mdl-23603616

ABSTRACT

Novel amidino-derivatives of phenylene-bisbenzothiazoles were synthesized and tested for their antiproliferative activity against several human cancer cell lines, as well as DNA-binding properties. The synthetic approach used for preparation of isomeric amidino substituted-phenylene-bis-benzothyazoles 3a-3f was achieved by condensation reaction of isophthaloyl dichloride 1a and terephthaloyl dichloride 1b or with phthalic acid 1c with 5-amidinium-2-aminobenzothiolate 2a and 5-(imidazolinium-2-yl)-2-aminobenzothiolate 2b in good yields. The targeted compounds were converted in the desired water soluble dihydrochloride salts by reaction of appropriate free base with concd HCl in ethanol or acetic acid. All tested compounds (3a-3f) showed antiproliferative effects on tumour cells in a concentration-dependant manner. The strongest activity and cytotoxicity was observed for diimidazolinyl substituted phenylene-bisbenzothiazole compound 3b. These effects were shown to be related to DNA-binding properties, topoisomerase I and II poisoning effects and apoptosis induction. The highest tested selectivity towards tumour cells was observed for the imidazolyl substituted phenylene-benzothiazole 3d that showed no cytotoxic effects on normal fibroblasts making it an excellent candidate for further chemical optimization and preclinical evaluation.


Subject(s)
Antineoplastic Agents/pharmacology , Benzothiazoles/pharmacology , Cell Proliferation/drug effects , DNA/chemistry , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Benzothiazoles/chemical synthesis , Benzothiazoles/chemistry , Cell Cycle/drug effects , Cell Line , Cell Line, Tumor , Circular Dichroism , Dose-Response Relationship, Drug , HeLa Cells , Humans , MCF-7 Cells , Models, Chemical , Molecular Structure , Nucleic Acid Denaturation/drug effects , Spectrophotometry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...