Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Nat Prod ; 74(7): 1568-74, 2011 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-21718055

RESUMO

The natural products combretastatin A-4 (CA4) and combretastatin A-1 (CA1) are potent cancer vascular disrupting agents and inhibitors of tubulin assembly (IC50 = 1-2 µM). The phosphorylated prodrugs CA4P and CA1P are undergoing human clinical trials against cancer. CA1 is unique due to its incorporation of a vicinal phenol, which has afforded the opportunity to prepare both diphosphate and regioisomeric monophosphate derivatives. Here, we describe the first synthetic routes suitable for the regiospecific preparation of the CA1-monophosphates CA1MPA (8a/b) and CA1MPB (4a/b). The essential regiochemistry necessary to distinguish between the two vicinal phenolic groups was accomplished with a tosyl protecting group strategy. Each of the four monophosphate analogues (including Z and E isomers) demonstrated in vitro cytotoxicity against selected human cancer cell lines comparable to their corresponding diphosphate congeners. Furthermore, Z-CA1MPA (8a) and Z-CA1MPB (4a) were inactive as inhibitors of tubulin assembly (IC50 > 40 µM), as anticipated in this pure protein assay.


Assuntos
Antineoplásicos Fitogênicos/síntese química , Estilbenos/síntese química , Antineoplásicos Fitogênicos/farmacologia , Humanos , Concentração Inibidora 50 , Estrutura Molecular , Solubilidade , Estereoisomerismo , Estilbenos/farmacologia , Relação Estrutura-Atividade , Tubulina (Proteína)/metabolismo , Água
2.
Bioorg Med Chem Lett ; 20(22): 6610-5, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20933415

RESUMO

A series of thiosemicarbazone analogs based on the benzophenone, thiophene, pyridine, and fluorene molecular frameworks has been prepared by chemical synthesis and evaluated as small-molecule inhibitors of the cysteine proteases cathepsin L and cathepsin B. The two most potent inhibitors of cathepsin L in this series (IC(50)<135 nM) are brominated-benzophenone thiosemicarbazone analogs that are further functionalized with a phenolic moiety (2 and 6). In addition, a bromo-benzophenone thiosemicarbazone acetyl derivative (3) is also strongly inhibitory against cathepsin L (IC(50)=150.8 nM). Bromine substitution in the thiophene series results in compounds that demonstrate only moderate inhibition of cathepsin L. The two most active analogs in the benzophenone thiosemicarbazone series are highly selective for their inhibition of cathepsin L versus cathepsin B.


Assuntos
Benzofenonas/química , Catepsina L/antagonistas & inibidores , Inibidores de Cisteína Proteinase/farmacologia , Fluorenos/química , Piridinas/química , Tiofenos/química , Tiossemicarbazonas/farmacologia , Inibidores de Cisteína Proteinase/química , Tiossemicarbazonas/química
3.
Bioorg Med Chem Lett ; 20(4): 1415-9, 2010 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-20089402

RESUMO

A small library of 36 functionalized benzophenone thiosemicarbazone analogs has been prepared by chemical synthesis and evaluated for their ability to inhibit the cysteine proteases cathepsin L and cathepsin B. Inhibitors of cathepsins L and B have the potential to limit or arrest cancer metastasis. The six most active inhibitors of cathepsin L (IC50<85 nM) in this series incorporate a meta-bromo substituent in one aryl ring along with a variety of functional groups in the second aryl ring. These six analogs are selective for their inhibition of cathepsin L versus cathepsin B (IC50>10,000 nM). The most active analog in the series, 3-bromophenyl-2'-fluorophenyl thiosemicarbazone 1, also efficiently inhibits cell invasion of the DU-145 human prostate cancer cell line.


Assuntos
Catepsina B/antagonistas & inibidores , Catepsina L/antagonistas & inibidores , Inibidores de Cisteína Proteinase/síntese química , Inibidores de Cisteína Proteinase/farmacologia , Desenho de Fármacos , Tiossemicarbazonas/síntese química , Tiossemicarbazonas/farmacologia , Domínio Catalítico , Inibidores de Cisteína Proteinase/química , Humanos , Concentração Inibidora 50 , Modelos Moleculares , Estrutura Molecular , Tiossemicarbazonas/química
4.
J Nat Prod ; 73(6): 1093-101, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20496923

RESUMO

Synthetic routes have been established for the preparation of regio- and stereoisomerically pure samples of the mono-beta-d-glucuronic acid derivatives of combretastatin A-1, referred to as CA1G1 (5a) and CA1G2 (6a). Judicious choice of protecting groups for the catechol ring was required for the regiospecific introduction of the glucuronic acid moiety. The tosyl group proved advantageous in this regard. The two monoglucuronic acid analogues demonstrate low cytotoxicity (compared to CA1, 2) against selected human cancer cell lines, with CA1G1 being slightly more potent than CA1G2.


Assuntos
Antineoplásicos Fitogênicos/síntese química , Ácido Glucurônico/química , Ácido Glucurônico/síntese química , Estilbenos/química , Estilbenos/síntese química , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Ácido Glucurônico/farmacologia , Humanos , Estrutura Molecular , Estereoisomerismo , Estilbenos/farmacologia
5.
Int J Radiat Oncol Biol Phys ; 69(2): 560-71, 2007 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-17869669

RESUMO

PURPOSE: To compare oxygen dependence and tissue transport properties of a new hypoxia-activated prodrug, PR-104A, with tirapazamine, and to evaluate the implications for antitumor activity when combined with radiotherapy. METHODS AND MATERIALS: Oxygen dependence of cytotoxicity was measured by clonogenic assay in SiHa cell suspensions. Tissue transport parameters were determined using SiHa multicellular layers. Spatially resolved pharmacokinetic (PK) and pharmacodynamic (PD) models were developed to predict cell killing in SiHa tumors and tested by clonogenic assay 18 h after treatment with the corresponding phosphate ester, PR-104. RESULTS: The K-value (oxygen concentration to halve cytotoxic potency) of PR-104A was 0.126 +/- 0.021 microM (10-fold lower than tirapazamine at 1.30 +/- 0.28 microM). The diffusion coefficient of PR-104A in multicellular layers (4.42 +/- 0.15 x 10(-7) cm2 s(-1)) was lower than that of tirapazamine (1.30 +/- 0.05 x 10(-6) cm2 s(-1)) but PK modeling predicted better penetration to hypoxic cells in tumors because of its slower metabolism. The tirapazamine PK/PD model successfully predicted the measured activity in combination with single-dose radiation against SiHa tumors, and the PR-104A model underpredicted the activity, which was greater for PR-104 than for tirapazamine (at equivalent host toxicity) both with radiation and as a single agent. CONCLUSION: PR-104/PR-104A has different PK/PD properties from tirapazamine and superior activity with single-dose radiotherapy against SiHa xenografts. We have inferred that PR-104A is better able to kill cells at intermediate partial pressure of oxygen in tumors than implied by the PK/PD model, most likely because of a bystander effect resulting from diffusion of its activated metabolites from severely hypoxic zones.


Assuntos
Antineoplásicos/farmacocinética , Hipóxia Celular/fisiologia , Compostos de Mostarda Nitrogenada/farmacocinética , Oxigênio/fisiologia , Pró-Fármacos/farmacocinética , Triazinas/farmacocinética , Animais , Transporte Biológico , Avaliação Pré-Clínica de Medicamentos , Humanos , Camundongos , Camundongos Nus , Modelos Biológicos , Pressão Parcial , Radiossensibilizantes/farmacocinética , Tirapazamina , Células Tumorais Cultivadas
6.
J Med Chem ; 50(25): 6392-404, 2007 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-18001018

RESUMO

Pharmacokinetic/pharmacodynamic (PK/PD) modeling has shown the antitumor activity of tirapazamine (TPZ), a bioreductive hypoxia-selective cytotoxin, to be limited by poor penetration through hypoxic tumor tissue. We have prepared a series of 1,2,4-benzotriazine 1,4-dioxide (BTO) analogues of TPZ to improve activity against hypoxic cells by increasing extravascular transport. The 6 substituents modified lipophilicity and rates of hypoxic metabolism. 3-Alkylamino substituents increased aqueous solubility and also influenced lipophilicity and hypoxic metabolism. PK/PD model-guided screening was used to select six BTOs for evaluation against hypoxic cells in HT29 human tumor xenografts. All six BTOs were active in vivo, and two provided greater hypoxic cell killing than TPZ because of improved transport and/or plasma PK. This PK/PD model considers two causes of therapeutic failure (limited tumor penetration and poor plasma pharmacokinetics) often not addressed early in drug development and provides a general strategy for selecting candidates for in vivo evaluation during lead optimization.


Assuntos
Modelos Biológicos , Neoplasias/metabolismo , Triazinas/farmacologia , Triazinas/farmacocinética , Algoritmos , Animais , Transporte Biológico , Hipóxia Celular , Linhagem Celular Tumoral , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias/irrigação sanguínea , Transplante Heterólogo , Triazinas/química
7.
J Med Chem ; 50(26): 6654-64, 2007 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-18052317

RESUMO

Tirapazamine (TPZ) and related 1,2,4-benzotriazine 1,4 dioxides (BTOs) are selectively toxic under hypoxia, but their ability to kill hypoxic cells in tumors is generally limited by their poor extravascular transport. Here we show that removing hydrogen bond donors by replacing the 3-NH2 group of TPZ with simple alkyl groups increased their tissue diffusion coefficients as measured in multicellular layer cultures. This advantage was largely retained using solubilizing 3-alkylaminoalkyl substituents provided these were sufficiently lipophilic at pH 7.4. The high reduction potentials of such compounds resulted in rates of metabolism too high for optimal penetration into hypoxic tissue, but electron-donating 6- and 7-substituents moderated metabolism. Pharmacokinetic/pharmacodynamic model-guided screening was used to select BTOs with optimal extravascular transport and hypoxic cytotoxicity properties for evaluation against HT29 human tumor xenografts in combination with radiation. This identified four novel 3-alkyl BTOs providing greater clonogenic killing of hypoxic cells than TPZ at equivalent host toxicity, with the 6-morpholinopropyloxy-BTO 22 being 3-fold more active.


Assuntos
Antineoplásicos/síntese química , Óxidos N-Cíclicos/síntese química , Triazinas/síntese química , Animais , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Transporte Biológico , Hipóxia Celular , Linhagem Celular Tumoral , Terapia Combinada , Óxidos N-Cíclicos/farmacocinética , Óxidos N-Cíclicos/farmacologia , Difusão , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Ligação de Hidrogênio , Camundongos , Modelos Biológicos , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/radioterapia , Relação Estrutura-Atividade , Transplante Heterólogo , Triazinas/farmacocinética , Triazinas/farmacologia , Ensaio Tumoral de Célula-Tronco
8.
Nat Rev Drug Discov ; 2(10): 803-11, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14526383

RESUMO

Sensing and responding to fluxes in oxygen tension is perhaps the single most important variable in physiology, and animal tissues have developed a number of essential mechanisms to cope with the stress of low physiological oxygen levels, or hypoxia. Among these coping mechanisms is the response mediated by the hypoxia-inducible transcription factor, or HIF-1. HIF-1 is an essential component in changing the transcriptional repertoire of tissues as oxygen levels drop, and could prove to be a very important target for drug development, as treatments evolve for diseases, such as cancer, heart disease and stroke, in which hypoxia is a central aspect.


Assuntos
Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas Nucleares/antagonistas & inibidores , Oxigênio/metabolismo , Fatores de Transcrição , Animais , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/terapia , Proteínas de Ligação a DNA/metabolismo , Desenho de Fármacos , Regulação da Expressão Gênica , Humanos , Fator 1 Induzível por Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Neoplasias/genética , Neoplasias/terapia , Proteínas Nucleares/metabolismo
9.
Cancer Res ; 64(2): 736-42, 2004 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-14744792

RESUMO

Tirapazamine (TPZ), a bioreductive drug with selective toxicity for hypoxic cells in tumors, is currently in Phase III clinical trials. It has been suggested to have a dual mechanism of action, both generating DNA radicals and oxidizing these radicals to form DNA breaks; whether the second (radical oxidation) step is rate-limiting in cells is not known. In this study we exploit the DNA radical oxidizing ability of the 1-N-oxide metabolite of TPZ, SR 4317, to address this question. SR 4317 at high, but nontoxic, concentrations potentiated the hypoxic (but not aerobic) cytotoxicity of TPZ in all four of the human tumor cell lines tested (HT29, SiHa, FaDu, and A549), thus providing a 2-3-fold increase in the hypoxic cytotoxicity ratio. In potentiating TPZ, SR 4317 was 20-fold more potent than the hypoxic cell radiosensitizers misonidazole and metronidazole but was less potent than misonidazole as a radiosensitizer, suggesting that the initial DNA radicals from TPZ and radiation are different. SR 4317 had favorable pharmacokinetic properties in CD-1 nude mice; coadministration with TPZ provided a large increase in the SR 4317 plasma concentrations relative to that for endogenous SR 4317 from TPZ. It also showed excellent extravascular transport properties in oxic and anoxic HT29 multicellular layers (diffusion coefficient 3 x 10(-6) cm(2)s(-1), with no metabolic consumption). Coadministration of SR 4317 (1 mmol/kg) with TPZ at a subtherapeutic dose (0.133 mmol/kg) significantly enhanced hypoxic cell killing in HT29 tumor xenografts without causing oxic cell killing, and the combination at its maximum tolerated dose was less toxic to hypoxic cells in the retina than was TPZ alone at its maximum tolerated dose. This study demonstrates that benzotriazine mono-N-oxides have potential use for improving the therapeutic utility of TPZ as a hypoxic cytotoxin in cancer treatment.


Assuntos
Antineoplásicos/toxicidade , Hipóxia Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Triazinas/toxicidade , Adenocarcinoma , Animais , Linhagem Celular Tumoral , Neoplasias do Colo , Sinergismo Farmacológico , Feminino , Humanos , Neoplasias Pulmonares , Camundongos , Neoplasias Faríngeas , Tirapazamina , Triazinas/síntese química , Triazinas/farmacocinética , Neoplasias do Colo do Útero
10.
J Med Chem ; 46(11): 2132-51, 2003 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-12747786

RESUMO

A set of 10 compounds, each combining the seco-1,2,9,9a-tetrahydrocyclopropa[c]benz[e]indol-4-one (seco-CBI) and pyrrolo[2,1-c][1,4]benzodiazepine (PBD) pharmacophores, was designed and prepared. These compounds were anticipated to cross-link between N3 of adenine and N2 of guanine in the minor groove of DNA. The compounds, which differ in the chain length separating the two alkylation subunits, and the configuration of the CBI portion, showed great variation in cellular toxicity (over 4 orders of magnitude in a cell line panel) with the most potent example exhibiting IC50s in the pM range. Cytotoxicity correlated with the ability of the compounds to cross-link naked DNA. Cross-linking was also observed in living cells, at much lower concentrations than for a related symmetrical PBD dimer. A thermal cleavage assay was used to assess sequence selectivity, demonstrating that the CBI portion controlled the alkylation sites, while the PBD substituent increased the overall efficiency of alkylation. Several compounds were tested for in vivo activity using a tumor growth delay assay against WiDr human colon carcinoma xenografts, with one compound (the most cytotoxic and most efficient cross-linker) showing a statistically significant increase in survival time following a single iv dose.


Assuntos
Antineoplásicos Alquilantes/síntese química , Benzodiazepinas/química , Benzodiazepinonas/síntese química , Reagentes de Ligações Cruzadas/síntese química , Ciclopropanos/química , DNA de Neoplasias/química , Indóis/química , Pirróis/química , Animais , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacologia , Benzodiazepinonas/química , Benzodiazepinonas/farmacologia , Ensaio Cometa , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Camundongos , Camundongos Nus , Transplante de Neoplasias , Relação Estrutura-Atividade , Células Tumorais Cultivadas
11.
Radiat Res ; 161(6): 656-66, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15161354

RESUMO

The hypoxic cytotoxin tirapazamine (TPZ) is currently in phase III clinical trial and appears to have clinical activity. One hypothesis as to why TPZ has been used more successfully in the clinic than most other bioreductive drugs is that its unusual O(2) dependence allows killing of radioresistant cells at "intermediate" O(2) concentrations. We have determined the O(2) dependence of the metabolism of TPZ to its reduction product SR 4317, and its cytotoxicity, in stirred suspensions of HT29 colon carcinoma cells while monitoring O(2) in solution with an Oxylite trade mark probe. The O(2) dependence of the cytotoxicity of TPZ is entirely accounted for by its inhibition of the metabolism of TPZ, with a K(O(2)) value (O(2) concentration for 50% inhibition) of 1.21 +/- 0.09 (SEM) microM. We used this experimental O(2) dependence to extend a recent (Hicks et al., Cancer Res. 63, 5970-5977, 2003) pharmacokinetic/pharmacodynamic model for the cytotoxicity of TPZ in anoxic HT29 multicellular layers to model cell killing in tumors. The model indicates that the O(2) dependence of killing by TPZ complements that of radiation well during fractionated radiotherapy. It predicts that lowering K(O(2)) would decrease killing in radioresistant cells at intermediate O(2) concentrations, while higher K(O(2)) values would exacerbate metabolic consumption of TPZ and thus further impede its penetration into hypoxic regions. Raising K(O(2)) would also increase metabolic activation at physiological O(2) concentrations, thereby compromising hypoxic selectivity. We conclude that the K(O(2)) value of TPZ is indeed close to the optimum for a bioreductive drug of this class (i.e. one that kills only cells in which it is reduced).


Assuntos
Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Neoplasias do Colo/metabolismo , Modelos Biológicos , Oxigênio/metabolismo , Triazinas/farmacologia , Triazinas/farmacocinética , Antineoplásicos/farmacocinética , Antineoplásicos/toxicidade , Carcinoma/metabolismo , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/metabolismo , Simulação por Computador , Relação Dose-Resposta a Droga , Humanos , Tolerância a Radiação/efeitos dos fármacos , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/toxicidade , Tirapazamina
12.
Cancer Chemother Pharmacol ; 51(1): 43-52, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12497205

RESUMO

PURPOSE: To determine whether there is a therapeutic interaction between the antivascular agent 5,6-dimethylxanthenone-4-acetic acid (DMXAA) and nine chemotherapy drugs against an early-passage mouse mammary tumour (MDAH-MCa-4), and to investigate the mechanism of any such interaction. METHODS AND RESULTS: Female C3H/HeN mice bearing intramuscular MDAH-MCa-4 tumours were injected intraperitoneally with DMXAA (80 micro mol/kg) or chemotherapy drug (at a range up to the maximum tolerated dose) alone, or coadministered. A small reduction in the dose of the chemotherapy drug was required in most cases, but the increase in antitumour effect was much greater than the increase in host toxicity (body weight loss). The therapeutic gain increased in the order 5-fluorouracil (no gain)<(etoposide, carboplatin, cyclophosphamide, doxorubicin, cisplatin)<(docetaxel, vincristine)

Assuntos
Antineoplásicos/administração & dosagem , Estilbenos , Xantenos/administração & dosagem , Xantonas , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bibenzilas/administração & dosagem , Carboplatina/administração & dosagem , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Neoplasias Mamárias Experimentais/irrigação sanguínea , Neoplasias Mamárias Experimentais/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C3H , Paclitaxel/administração & dosagem , Fatores de Tempo , Xantenos/farmacocinética , Xantenos/farmacologia
13.
Clin Cancer Res ; 16(20): 4946-57, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20732963

RESUMO

PURPOSE: Tirapazamine (TPZ) has attractive features for targeting hypoxic cells in tumors but has limited clinical activity, in part because of poor extravascular penetration. Here, we identify improved TPZ analogues by using a spatially resolved pharmacokinetic/pharmacodynamic (SR-PKPD) model that considers tissue penetration explicitly during lead optimization. EXPERIMENTAL DESIGN: The SR-PKPD model was used to guide the progression of 281 TPZ analogues through a hierarchical screen. For compounds exceeding hypoxic selectivity thresholds in single-cell cultures, SR-PKPD model parameters (kinetics of bioreductive metabolism, clonogenic cell killing potency, diffusion coefficients in multicellular layers, and plasma pharmacokinetics at well tolerated doses in mice) were measured to prioritize testing in xenograft models in combination with radiation. RESULTS: SR-PKPD-guided lead optimization identified SN29751 and SN30000 as the most promising hypoxic cytotoxins from two different structural subseries. Both were reduced to the corresponding 1-oxide selectively under hypoxia by HT29 cells, with an oxygen dependence quantitatively similar to that of TPZ. SN30000, in particular, showed higher hypoxic potency and selectivity than TPZ in tumor cell cultures and faster diffusion through HT29 and SiHa multicellular layers. Both compounds also provided superior plasma PK in mice and rats at equivalent toxicity. In agreement with SR-PKPD predictions, both were more active than TPZ with single dose or fractionated radiation against multiple human tumor xenografts. CONCLUSIONS: SN30000 and SN29751 are improved TPZ analogues with potential for targeting tumor hypoxia in humans. Novel SR-PKPD modeling approaches can be used for lead optimization during anticancer drug development.


Assuntos
Antineoplásicos/farmacocinética , Neoplasias/tratamento farmacológico , Triazinas/farmacocinética , Algoritmos , Animais , Antineoplásicos/sangue , Antineoplásicos/farmacologia , Antineoplásicos/toxicidade , Hipóxia Celular/efeitos dos fármacos , Células HT29 , Humanos , Masculino , Camundongos , Camundongos Nus , Modelos Biológicos , Neoplasias/metabolismo , Neoplasias/patologia , Neoplasias/radioterapia , Oxigênio/metabolismo , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/farmacologia , Radiossensibilizantes/toxicidade , Ratos , Ratos Sprague-Dawley , Tirapazamina , Triazinas/sangue , Triazinas/farmacologia , Triazinas/toxicidade , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA