Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharm Sci ; 195: 106661, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38052257

RESUMO

Temozolomide (TMZ) a DNA alkylating agent, is the standard-of-care for brain tumors, such as glioblastoma multiforme (GBM). Although the physicochemical and pharmacokinetic properties of TMZ, such as chemical stability and the ability to cross the blood-brain barrier (BBB), have been questioned in the past, the acquired chemoresistance has been the main limiting factor of long-term clinical use of TMZ. In the present study, an L-type amino acid transporter 1 (LAT1)-utilizing prodrug of TMZ (TMZ-AA, 6) was prepared and studied for its cellular accumulation and cytotoxic properties in human squamous cell carcinoma, UT-SCC-28 and UT-SCC-42B cells, and TMZ-sensitive human glioma, U-87MG cells that expressed functional LAT1. TMZ-AA 6 accumulated more effectively than TMZ itself into those cancer cells that expressed LAT1 (UT-SCC-42B). However, this did not correlate with decreased viability of treated cells. Indeed, TMZ-AA 6, similarly to TMZ itself, required adjuvant inhibitor(s) of DNA-repair systems, O6-methylguanine-DNA methyl transferase (MGMT) and base excision repair (BER), as well as active DNA mismatch repair (MMR), for maximal growth inhibition. The present study shows that improving the delivery of this widely-used methylating agent is not the main barrier to improved chemotherapy, although utilizing a specific transporter overexpressed at the BBB or glioma cells can have targeting advantages. To obtain a more effective anticancer prodrug, the compound design focus should shift to altering the major DNA alkylation site or inhibiting DNA repair systems.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Glioma , Pró-Fármacos , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Antineoplásicos Alquilantes/farmacologia , Dacarbazina/farmacologia , Resistencia a Medicamentos Antineoplásicos , Reparo do DNA , Glioblastoma/tratamento farmacológico , Glioma/tratamento farmacológico , Neoplasias Encefálicas/tratamento farmacológico , DNA , Pró-Fármacos/farmacologia , Pró-Fármacos/uso terapêutico , Linhagem Celular Tumoral
2.
RSC Chem Biol ; 4(10): 736-741, 2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37799580

RESUMO

It is 40 years since the publication of the patent that announced the imidazotetrazines temozolomide and mitozolomide to the world and 30 since the discovery that they function as prodrugs of alkyldiazonium reactive intermediates. Temozolomide combined with radiation is established as the first-line treatment for glioma but despite the attentions of the inventors and others, further examples of this intriguing ring system have yet to enter the clinic.

3.
Medchemcomm ; 9(3): 545-553, 2018 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-30108945

RESUMO

A series of 3-(benzyl-substituted)-imidazo[5,1-d]-1,2,3,5-tetrazines (13) and related derivatives with 3-heteromethyl groups has been synthesised and screened for growth-inhibitory activity in vitro against two pairs of glioma cell lines with temozolomide-sensitive and -resistant phenotypes dependent on the absence/presence of the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). In general the compounds had low inhibitory activity with GI50 values >50 µM against both sets of cell lines. Two silicon-containing derivatives, the TMS-methylimidazotetrazine (9) and the SEM-analogue (10), showed interesting differences: compound (9) had a profile very similar to that of temozolomide with the MGMT+ cell lines being 5 to 10-fold more resistant than MGMT- isogenic partners; the SEM-substituted compound (10) showed potency across all cell lines irrespective of their MGMT status.

5.
Angew Chem Int Ed Engl ; 57(31): 9799-9804, 2018 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-29863754

RESUMO

The ligands L1 and L2 both form separable dinuclear double-stranded helicate and mesocate complexes with RuII . In contrast to clinically approved platinates, the helicate isomer of [Ru2 (L1 )2 ]4+ was preferentially cytotoxic to isogenic cells (HCT116 p53-/- ), which lack the critical tumour suppressor gene. The mesocate isomer shows the reverse selectivity, with the achiral isomer being preferentially cytotoxic towards HCT116 p53+/+ . Other structurally similar RuII -containing dinuclear complexes showed very little cytotoxic activity. This study demonstrates that alterations in ligand or isomer can have profound effects on cytotoxicity towards cancer cells of different p53 status and suggests that selectivity can be "tuned" to either genotype. In the search for compounds that can target difficult-to-treat tumours that lack the p53 tumour suppressor gene, [Ru2 (L1 )2 ]4+ is a promising compound for further development.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Compostos Organometálicos/farmacologia , Rutênio/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Antineoplásicos/síntese química , Antineoplásicos/química , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Estrutura Molecular , Compostos Organometálicos/síntese química , Compostos Organometálicos/química , Rutênio/química , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/metabolismo
6.
Cancer Lett ; 403: 98-107, 2017 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-28624622

RESUMO

Organometallic complexes offer the prospect of targeting multiple pathways that are important in cancer biology. Here, the preclinical activity and mechanism(s) of action of a silver-bis(N-heterocyclic carbine) complex (Ag8) were evaluated. Ag8 induced DNA damage via several mechanisms including topoisomerase I/II and thioredoxin reductase inhibition and induction of reactive oxygen species. DNA damage induction was consistent with cytotoxicity observed against proliferating cells and Ag8 induced cell death by apoptosis. Ag8 also inhibited DNA repair enzyme PARP1, showed preferential activity against cisplatin resistant A2780 cells and potentiated the activity of temozolomide. Ag8 was substantially less active against non-proliferating non-cancer cells and selectively inhibited glycolysis in cancer cells. Ag8 also induced significant anti-tumour effects against cells implanted intraperitoneally in hollow fibres but lacked activity against hollow fibres implanted subcutaneously. Thus, Ag8 targets multiple pathways of importance in cancer biology, is less active against non-cancer cells and shows activity in vivo in a loco-regional setting.


Assuntos
Antineoplásicos/farmacologia , Imidazóis/farmacologia , Neoplasias/tratamento farmacológico , Compostos Organometálicos/farmacologia , Antígenos de Neoplasias/metabolismo , Antineoplásicos/química , Antineoplásicos/toxicidade , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/farmacologia , Dano ao DNA , DNA Topoisomerases Tipo I/metabolismo , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos , Sinergismo Farmacológico , Glicólise/efeitos dos fármacos , Humanos , Imidazóis/química , Imidazóis/toxicidade , Concentração Inibidora 50 , Neoplasias/metabolismo , Neoplasias/patologia , Compostos Organometálicos/química , Compostos Organometálicos/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1/antagonistas & inibidores , Poli(ADP-Ribose) Polimerase-1/metabolismo , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida , Tiorredoxina Redutase 1/antagonistas & inibidores , Tiorredoxina Redutase 1/metabolismo , Inibidores da Topoisomerase I/farmacologia , Inibidores da Topoisomerase II/farmacologia
7.
Chemistry ; 21(19): 7278-84, 2015 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-25802084

RESUMO

Triplexes formed from oligonucleic acids are key to a number of biological processes. They have attracted attention as molecular biology tools and as a result of their relevance in novel therapeutic strategies. The recognition properties of single-stranded nucleic acids are also relevant in third-strand binding. Thus, there has been considerable activity in generating such moieties, referred to as triplex forming oligonucleotides (TFOs). Triplexes, composed of Watson-Crick (W-C) base-paired DNA duplexes and a Hoogsteen base-paired RNA strand, are reported to be more thermodynamically stable than those in which the third strand is DNA. Consequently, synthetic efforts have been focused on developing TFOs with RNA-like structural properties. Here, the structural and stability studies of such a TFO, composed of deoxynucleic acids, but with 3'-S-phosphorothiolate (3'-SP) linkages at two sites is described. The modification results in an increase in triplex melting temperature as determined by UV absorption measurements. (1) H NMR analysis and structure generation for the (hairpin) duplex component and the native and modified triplexes revealed that the double helix is not significantly altered by the major groove binding of either TFO. However, the triplex involving the 3'-SP modifications is more compact. The 3'-SP modification was previously shown to stabilise G-quadruplex and i-motif structures and therefore is now proposed as a generic solution to stabilising multi-stranded DNA structures.


Assuntos
DNA/química , Fosfatos/química , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Conformação de Ácido Nucleico , Desnaturação de Ácido Nucleico , Termodinâmica
8.
Mol Cancer Ther ; 14(1): 111-9, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25351918

RESUMO

The cellular responses to two new temozolomide (TMZ) analogues, DP68 and DP86, acting against glioblastoma multiforme (GBM) cell lines and primary culture models are reported. Dose-response analysis of cultured GBM cells revealed that DP68 is more potent than DP86 and TMZ and that DP68 was effective even in cell lines resistant to TMZ. On the basis of a serial neurosphere assay, DP68 inhibits repopulation of these cultures at low concentrations. The efficacy of these compounds was independent of MGMT and MMR functions. DP68-induced interstrand DNA cross-links were demonstrated with H2O2-treated cells. Furthermore, DP68 induced a distinct cell-cycle arrest with accumulation of cells in S phase that is not observed for TMZ. Consistent with this biologic response, DP68 induces a strong DNA damage response, including phosphorylation of ATM, Chk1 and Chk2 kinases, KAP1, and histone variant H2AX. Suppression of FANCD2 expression or ATR expression/kinase activity enhanced antiglioblastoma effects of DP68. Initial pharmacokinetic analysis revealed rapid elimination of these drugs from serum. Collectively, these data demonstrate that DP68 is a novel and potent antiglioblastoma compound that circumvents TMZ resistance, likely as a result of its independence from MGMT and mismatch repair and its capacity to cross-link strands of DNA.


Assuntos
Compostos de Anilina/administração & dosagem , Antineoplásicos Alquilantes/administração & dosagem , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Compostos Heterocíclicos com 2 Anéis/administração & dosagem , Recidiva Local de Neoplasia/tratamento farmacológico , Compostos de Anilina/síntese química , Compostos de Anilina/farmacocinética , Animais , Antineoplásicos Alquilantes/síntese química , Antineoplásicos Alquilantes/farmacocinética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Neoplasias Encefálicas/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/administração & dosagem , Dacarbazina/farmacocinética , Proteína do Grupo de Complementação D2 da Anemia de Fanconi/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/metabolismo , Compostos Heterocíclicos com 2 Anéis/síntese química , Compostos Heterocíclicos com 2 Anéis/farmacocinética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Recidiva Local de Neoplasia/metabolismo , Temozolomida , Proteínas Supressoras de Tumor/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Pharmaceuticals (Basel) ; 7(7): 797-838, 2014 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25014631

RESUMO

Temozolomide (TMZ) is the standard first line treatment for malignant glioma, reaching "blockbuster" status in 2010, yet it remains the only drug in its class. The main constraints on the clinical effectiveness of TMZ therapy are its requirement for active DNA mismatch repair (MMR) proteins for activity, and inherent resistance through O6-methyl guanine-DNA methyl transferase (MGMT) activity. Moreover, acquired resistance, due to MMR mutation, results in aggressive TMZ-resistant tumour regrowth following good initial responses. Much of the attraction in TMZ as a drug lies in its PK/PD properties: it is acid stable and has 100% oral bioavailability; it also has excellent distribution properties, crosses the blood-brain barrier, and there is direct evidence of tumour localisation. This review seeks to unravel some of the mysteries of the imidazotetrazine class of compounds to which TMZ belongs. In addition to an overview of different synthetic strategies, we explore the somewhat unusual chemical reactivity of the imidazotetrazines, probing their mechanisms of reaction, examining which attributes are required for an active drug molecule and reviewing the use of this combined knowledge towards the development of new and improved anti-cancer agents.

10.
Pharmaceuticals (Basel) ; 6(12): 1475-506, 2013 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-24287492

RESUMO

Glioblastoma multiforme (GBM) is a grade IV brain tumor characterized by a heterogeneous population of cells that are highly infiltrative, angiogenic and resistant to chemotherapy. The current standard of care, comprised of surgical resection followed by radiation and the chemotherapeutic agent temozolomide, only provides patients with a 12-14 month survival period post-diagnosis. Long-term survival for GBM patients remains uncommon as cells with intrinsic or acquired resistance to treatment repopulate the tumor. In this review we will describe the mechanisms of resistance, and how they may be overcome to improve the survival of GBM patients by implementing novel chemotherapy drugs, new drug combinations and new approaches relating to DNA damage, angiogenesis and autophagy.

11.
J Med Chem ; 56(17): 7120-32, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23895620

RESUMO

The antitumor prodrug temozolomide is compromised by its dependence for activity on DNA mismatch repair (MMR) and the repair of the chemosensitive DNA lesion, O6-methylguanine (O6-MeG), by O6-methylguanine-DNA-methyltransferase (E.C. 2.1.1.63, MGMT). Tumor response is also dependent on wild-type p53. Novel 3-(2-anilinoethyl)-substituted imidazotetrazines are reported that have activity independent of MGMT, MMR, and p53. This is achieved through a switch of mechanism so that bioactivity derives from imidazotetrazine-generated arylaziridinium ions that principally modify guanine-N7 sites on DNA. Mono- and bifunctional analogues are reported, and a quantitative structure-activity relationship (QSAR) study identified the p-tolyl-substituted bifunctional congener as optimized for potency, MGMT-independence, and MMR-independence. NCI60 data show the tumor cell response is distinct from other imidazotetrazines and DNA-guanine-N7 active agents such as nitrogen mustards and cisplatin. The new imidazotetrazine compounds are promising agents for further development, and their improved in vitro activity validates the principles on which they were designed.


Assuntos
Pareamento Incorreto de Bases , Reparo do DNA , O(6)-Metilguanina-DNA Metiltransferase/química , Pró-Fármacos/química , Proteína Supressora de Tumor p53/metabolismo , Relação Quantitativa Estrutura-Atividade
12.
Biochem Pharmacol ; 83(11): 1514-22, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22387433

RESUMO

Alchemix is an exemplar of a class of anthraquinone with efficacy against multidrug resistant tumours. We have explored further the mechanism of action of alchemix and investigated the effect of extending its side arm bearing the alkylating functionality with regard to DNA binding and activity against multidrug resistant cancer cells. Increasing the distance between the intercalating chromophore and the alkylating functionality of ICT2901 (propyl), ICT2902 (butyl) and ICT2903 (pentyl), led to a higher number of DNA alkylation sites, more potent topoisomerase II inhibition and generated more apoptotic and necrotic cells when analysed in p53-proficient HCT116 cells. Intriguingly, alchemix, the compound with the shortest distance between its intercalative chromophore and alkylating functionality (ethyl), did not conform to this SAR. A different toxicity pattern against DNA repair defective CHO cell lines as well as arrest of cells in G1 supports a somewhat distinct mode of action by alchemix compared with its analogues. Importantly, both alchemix and ICT2901 demonstrated greater cytotoxic activity against anthraquinone-resistant MCF-7/adr cells than wild-type MCF-7 cells. Subtle synthetic modification in this anthraquinone series has led to significant changes to the stability of DNA-compound complexes and cellular activity. Given that the failure of chemotherapy in the clinic is often associated with MDR, the results of both alchemix and ICT2901 represent important advances towards improved therapies.


Assuntos
Antraquinonas/química , Antraquinonas/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Animais , Células CHO , Ciclo Celular , Morte Celular/efeitos dos fármacos , Cricetinae , Adutos de DNA , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HCT116 , Histonas/genética , Histonas/metabolismo , Humanos , Estrutura Molecular , Relação Estrutura-Atividade
13.
J Pharm Pharmacol ; 64(2): 237-43, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22221099

RESUMO

OBJECTIVES: To investigate the metabolism of cryptolepine and some cryptolepine analogues by aldehyde oxidase, and to assess the implications of the results on the potential of cryptolepine analogues as antimalarial agents. METHODS: The products resulting from the oxidation of cryptolepine and 2-fluorocryptolepine by a rabbit liver preparation of aldehyde oxidase were isolated and identified using chromatographic and spectroscopic techniques. The antiplasmodial activity of cryptolepine-11-one was assessed against Plasmodium falciparum using the parasite lactate dehydrogenase assay. KEY FINDINGS: Cryptolepine was oxidized by aldehyde oxidase give cryptolepine-11-one. Although 2-fluorocryptolepine was found to have less affinity for the enzyme than cryptolepine, it was a better substrate for aldehyde oxidase than the parent compound. In contrast, quindoline, the 11-chloro- , 2,7-dibromo- and 2-methoxy analogues of cryptolepine were not readily oxidized. Cryptolepine-11-one was found to be inactive against P. falciparum in vitro raising the possibility that the effectiveness of cryptolepine as an antimalarial, may be compromised by metabolism to an inactive metabolite by liver aldehyde oxidase. CONCLUSIONS: Cryptolepine and 2-fluorocryptolepine are substrates for aldehyde oxidase. This may have implications for the design and development of cryptolepine analogues as antimalarial agents.


Assuntos
Aldeído Oxidase/metabolismo , Antimaláricos/metabolismo , Alcaloides Indólicos/metabolismo , Fígado/enzimologia , Malária Falciparum/tratamento farmacológico , Quinolinas/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Cromatografia em Camada Fina , Cryptolepis , Feminino , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Oxirredução , Raízes de Plantas/química , Plantas Medicinais/química , Plasmodium falciparum , Coelhos
14.
ACS Med Chem Lett ; 3(12): 965-8, 2012 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-24900418

RESUMO

The imidazotetrazine ring is an acid-stable precursor and prodrug of highly reactive alkyl diazonium ions. We have shown that this reactivity can be managed productively in an aqueous system for the generation of aziridinium ions with 96% efficiency. The new compounds are potent DNA alkylators and have antitumor activity independent of the O6-methylguanine-DNA methyltransferase and DNA mismatch repair constraints that limit the use of Temozolomide.

16.
Methods Mol Biol ; 613: 55-70, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19997877

RESUMO

The DNA x RNA hybrid duplexes are functionally important structures in gene expression that are underutilized as potential drug targets. Several tools are described here for the discovery and characterization of small molecules capable of the selective recognition of DNA x RNA hybrid structures. Competition dialysis and thermal denaturation of mixtures of polynucleotide structures can be used to identify small molecules that bind selectively to DNA x RNA hybrids. An assay that measures small molecule inhibition of RNase H can be used to measure a functional response to these ligands.


Assuntos
DNA/química , DNA/metabolismo , Ácidos Nucleicos Heteroduplexes/metabolismo , Preparações Farmacêuticas/metabolismo , RNA/química , RNA/metabolismo , Sequência de Bases , Ligação Competitiva , DNA/genética , Determinação de Ponto Final , Ligantes , Desnaturação de Ácido Nucleico , Ácidos Nucleicos Heteroduplexes/química , Ácidos Nucleicos Heteroduplexes/genética , Hibridização de Ácido Nucleico , RNA/genética , Ribonuclease H/metabolismo , Especificidade por Substrato , Temperatura , Termodinâmica
17.
FEBS J ; 274(1): 287-99, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17222188

RESUMO

The cobalamin-dependent cytosolic enzyme, methionine synthase (EC.2.1.1.13), catalyzes the remethylation of homocysteine to methionine using 5-methyltetrahydrofolate as the methyl donor. The products of this remethylation--methionine and tetrahydrofolate--participate in the active methionine and folate pathways. Impaired methionine synthase activity has been implicated in the pathogenesis of anaemias, cancer and neurological disorders. Although the need for potent and specific inhibitors of methionine synthase has been recognized, there is a lack of such agents. In this study, we designed, synthesized and evaluated the inhibitory activity of a series of substituted benzimidazoles and small benzothiadiazoles. Kinetic analysis revealed that the benzimidazoles act as competitive inhibitors of the rat liver methionine synthase, whilst the most active benzothiadiazole (IC(50) = 80 microm) exhibited characteristics of uncompetitive inhibition. A model of the methyltetrahydrofolate-binding site of the rat liver methionine synthase was constructed; docking experiments were designed to elucidate, in greater detail, the binding mode and reveal structural requirements for the design of inhibitors of methionine synthase. Our results indicate that the potency of the tested compounds is related to a planar region of the inhibitor that can be positioned in the centre of the active site, the presence of a nitro functional group and two or three probable hydrogen-bonding interactions.


Assuntos
5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/antagonistas & inibidores , 5-Metiltetra-Hidrofolato-Homocisteína S-Metiltransferase/química , Benzimidazóis/farmacologia , Inibidores Enzimáticos/farmacologia , Tiadiazóis/farmacologia , Sequência de Aminoácidos , Animais , Benzimidazóis/síntese química , Sítios de Ligação , Inibidores Enzimáticos/química , Ligação de Hidrogênio , Cinética , Fígado/enzimologia , Modelos Moleculares , Dados de Sequência Molecular , Ratos , Homologia de Sequência , Relação Estrutura-Atividade , Tetra-Hidrofolatos/química , Tetra-Hidrofolatos/metabolismo , Tiadiazóis/química
18.
J Med Chem ; 49(17): 5187-98, 2006 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-16913707

RESUMO

Biarylpyrimidines are characterized as selective ligands for higher-order nucleic acid structures. A concise and efficient synthesis has been devised incorporating Suzuki biaryl cross-coupling of dihalopyrimidines. Two ligand series are described based on the parent thioether 4,6-bis[4-[[2-(dimethylamino)ethyl]mercapto]phenyl]pyrimidine (1a) and amide 4,6-bis(4[(2-(dimethylamino)ethyl)carboxamido]phenyl)pyrimidine (2a) compounds. In UV thermal denaturation studies with the poly(dA) x [poly(dT)]2 triplex structure, thioethers showed stabilization of the triplex form (Delta Tm < or = 20 degrees C). In contrast, amides showed duplex stabilization (Delta Tm < or = 15 degrees C) and either negligible stabilization or specific destabilization (Delta Tm = -5 degrees C) of the triplex structure. Full spectra of nucleic acid binding preferences were determined by competition dialysis. The strongest interacting thioether bound preferentially to the poly(dA) x [poly(dT)]2 triplex, K(app) = 1.6 x 10(5) M(-1) (40 x K(app) for CT DNA duplex). In contrast, the strongest binding amide selected the (T2G20T2)4 quadruplex structure, K(app) = 0.31 x 10(5) M(-1) (6.5 x K(app) for CT DNA duplex).


Assuntos
Ácidos Nucleicos/química , Pirimidinas , Telomerase/antagonistas & inibidores , Sítios de Ligação , DNA/química , Desenho de Fármacos , Humanos , Ligantes , Modelos Moleculares , Estrutura Molecular , Pirimidinas/química , Pirimidinas/classificação , Pirimidinas/farmacologia , Sensibilidade e Especificidade , Espectrofotometria Ultravioleta/métodos , Relação Estrutura-Atividade , Temperatura
19.
Eur J Med Chem ; 41(3): 330-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16458393

RESUMO

The protein O6-alkylguanine-DNA alkyltransferase (Atase) is responsible for the repair of DNA lesions generated by several clinically important anti-cancer drugs; this is manifest as active resistance in those cancer cell lines proficient in Atase expression. Novel O6-substituted guanine analogues have been synthesized, bearing acidic, basic and hydrogen bonding functional groups. In contrast to existing O6-modified purine analogues, such as methyl or benzyl, the new compounds were found to resist repair by Atase even when tested at concentrations much higher than O6-benzylguanine, a well-established Atase substrate active both in vitro and in vivo. The inactivity of the new purines as covalent substrates for Atase indicates that agents to deliver these groups to DNA would represent a new class of DNA-modifying drug that circumvents Atase-mediated resistance.


Assuntos
Enzimas Reparadoras do DNA/química , Desenho de Fármacos , Guanina/análogos & derivados , Guanina/química , O(6)-Metilguanina-DNA Metiltransferase/metabolismo , Purinas/química , Cristalografia por Raios X , DNA/química , DNA/efeitos dos fármacos , Guanina/farmacologia , Humanos , Modelos Biológicos , Estrutura Molecular , Mutação , O(6)-Metilguanina-DNA Metiltransferase/genética , Purinas/farmacologia , Especificidade por Substrato
20.
J Nat Prod ; 67(9): 1445-9, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15387639

RESUMO

Bioassay-guided fractionation was applied to the cytotoxic chloroform fraction of the red alga Polysiphonia lanosa. The major compounds of the most active fraction were identified using GLC-MS analysis as lanosol (1), methyl, ethyl, and n-propyl ethers of lanosol (1a, 1b, and 1c, respectively), and aldehyde of lanosol (2), although 1b appears to be an artifact arising during the fractionation procedure. These compounds and other known bromophenols were synthesized in addition to four novel isomers (3, 3a-c). The cytotoxic activities of all the synthetic compounds were determined against DLD-1 cells using the MTT assay. Compounds with IC(50) < 20 micromol were also tested against HCT-116 cells. Compound 3c (2,5-dibromo-3,4-dihydroxybenzyl n-propyl ether) was the most active compound against both cell lines (IC(50) = 1.72 and 0.80 micromol, respectively), and its effect on the cell cycle was studied using flow cytometry.


Assuntos
Antineoplásicos/isolamento & purificação , Hidrocarbonetos Bromados/isolamento & purificação , Fenóis/isolamento & purificação , Antineoplásicos/química , Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Inglaterra , Hidrocarbonetos Bromados/química , Hidrocarbonetos Bromados/farmacologia , Concentração Inibidora 50 , Isomerismo , Estrutura Molecular , Ressonância Magnética Nuclear Biomolecular , Fenóis/química , Fenóis/farmacologia , Rodófitas , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...