Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
JCI Insight ; 7(1)2022 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-34807849

RESUMO

Sangivamycin is a nucleoside analog that is well tolerated by humans and broadly active against phylogenetically distinct viruses, including arenaviruses, filoviruses, and orthopoxviruses. Here, we show that sangivamycin is a potent antiviral against multiple variants of replicative severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with half-maximal inhibitory concentration in the nanomolar range in several cell types. Sangivamycin suppressed SARS-CoV-2 replication with greater efficacy than remdesivir (another broad-spectrum nucleoside analog). When we investigated sangivamycin's potential for clinical administration, pharmacokinetic; absorption, distribution, metabolism, and excretion (ADME); and toxicity properties were found to be favorable. When tested in combination with remdesivir, efficacy was additive rather than competitive against SARS-CoV-2. The proven safety in humans, long half-life, potent antiviral activity (compared to remdesivir), and combinatorial potential suggest that sangivamycin is likely to be efficacious alone or in combination therapy to suppress viremia in patients. Sangivamycin may also have the ability to help combat drug-resistant or vaccine-escaping SARS-CoV-2 variants since it is antivirally active against several tested variants. Our results support the pursuit of sangivamycin for further preclinical and clinical development as a potential coronavirus disease 2019 therapeutic.


Assuntos
Antivirais , Nucleosídeos de Pirimidina , SARS-CoV-2/efeitos dos fármacos , Animais , Antivirais/farmacocinética , Antivirais/farmacologia , Antivirais/toxicidade , COVID-19/virologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Feminino , Humanos , Masculino , Camundongos , Nucleosídeos de Pirimidina/farmacocinética , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Células Vero
2.
Acc Chem Res ; 54(13): 2798-2811, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34152729

RESUMO

Without question, natural products have provided the lion share of leads, if not drugs themselves, for the treatment of bacterial infections. The bacterial arms race, fueled by selection and survival pressures has delivered a natural arsenal of small molecules targeting the most essential of life processes. Antibiotics that target these critical intracellular processes face the formidable defense of both penetrating a bacterial cell membrane and avoiding efflux to exert their effect. These challenges are especially effective in Gram-negative (Gram-(-)) bacteria, which have a double membrane structure and efficient efflux systems from the combination of outer-membrane porins and inner membrane proton pumps. In this landscape of offense and defense, our clinically used antibiotics have only successfully targeted three intracellular processes for therapeutic intervention in Gram-(-) bacteria: dihydrofolate biosynthesis, transcription, and translation. Not surprisingly, such critical survival machinery is a popular target for bacterial warfare, and eight of our 14 classes of commonly used antibiotics target translation with the bacterial ribosome remaining one the most vetted targets for antimicrobial therapy. On the plus side, its anionic character attracts cationic inhibitors, which are generally more capable of penetrating the bacterial cell wall, and clinical resistance rates are usually manageable as mutation of such a highly evolved machine is difficult. On the down side, this highly evolved machine renders it difficult to inhibit selectively, and the inhibition of prokaryotic translation versus both eukaryotic cellular and mitochondrial translation is critical for clinical development and minimization of undesired toxicities.A class of natural products known as the "nucleoside antibiotics" have historically been recognized as universal inhibitors of the ribosome and can inhibit translation in prokaryotes, eukaryotes, and archaea. While they have served an essential role in dissecting the biochemical underpinnings of the enzymatic functions of the ribosome, they have not proven therapeutically useful as they target the highly conserved rRNA in the P-site and are toxic to mammalian cells. In this Account, we describe our studies on the natural product amicetin, a nucleoside antibiotic that we have demonstrated to break the rule of being a universal translation inhibitor. While the cytosine of amicetin mimics C75 of the 3'-CCA tail of the P-site tRNA akin to other nucleoside antibiotics, we advance a hypothesis that amicetin's unique interaction with the ribosomal protein uL16 exploits an untapped mechanism for selectively targeting the bacterial ribosome. A complex molecule comprised of a nucleoside, carbohydrates and amino acids, amicetin is also chemically unstable. Our initial attempts to stabilize and simplify this scaffold are presented with the ultimate goal of rebuilding the compound with improved penetrance to bacterial cells. If successful, this scaffold would demonstrate a path forward for a new class of antibiotics capable of selectively targeting the ribosomal P-site.


Assuntos
Antibacterianos/farmacologia , Bactérias/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Ribossomos/metabolismo , Animais , Antibacterianos/síntese química , Antibacterianos/metabolismo , Antibacterianos/toxicidade , Bactérias/química , Sítios de Ligação , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Testes de Sensibilidade Microbiana , Ligação Proteica , Nucleosídeos de Pirimidina/síntese química , Nucleosídeos de Pirimidina/metabolismo , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Ribossomos/química
3.
Molecules ; 25(16)2020 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-32823658

RESUMO

Inhibition of DNA repair enzymes tyrosyl-DNA phosphodiesterase 1 and poly(ADP-ribose)polymerases 1 and 2 in the presence of pyrimidine nucleoside derivatives was studied here. New effective Tdp1 inhibitors were found in a series of nucleoside derivatives possessing 2',3',5'-tri-O-benzoyl-d-ribofuranose and 5-substituted uracil moieties and have half-maximal inhibitory concentrations (IC50) in the lower micromolar and submicromolar range. 2',3',5'-Tri-O-benzoyl-5-iodouridine manifested the strongest inhibitory effect on Tdp1 (IC50 = 0.6 µM). A decrease in the number of benzoic acid residues led to a marked decline in the inhibitory activity, and pyrimidine nucleosides lacking lipophilic groups (uridine, 5-fluorouridine, 5-chlorouridine, 5-bromouridine, 5-iodouridine, and ribothymidine) did not cause noticeable inhibition of Tdp1 (IC50 > 50 µM). No PARP1/2 inhibitors were found among the studied compounds (residual activity in the presence of 1 mM substances was 50-100%). Several O-benzoylated uridine and cytidine derivatives strengthened the action of topotecan on HeLa cervical cancer cells.


Assuntos
Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Interações Hidrofóbicas e Hidrofílicas , Diester Fosfórico Hidrolases/metabolismo , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/toxicidade , Células HeLa , Humanos , Nucleosídeos de Pirimidina/toxicidade
4.
Clin Cancer Res ; 25(20): 6035-6043, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31337643

RESUMO

PURPOSE: Iododeoxyuridine (IUdR) is a potent radiosensitizer; however, its clinical utility is limited by dose-limiting systemic toxicities and the need for prolonged continuous infusion. 5-Iodo-2-pyrimidinone-2'-deoxyribose (IPdR) is an oral prodrug of IUdR that, compared with IUdR, is easier to administer and less toxic, with a more favorable therapeutic index in preclinical studies. Here, we report the clinical and pharmacologic results of a first-in-human phase I dose escalation study of IPdR + concurrent radiation therapy (RT) in patients with advanced metastatic gastrointestinal (GI) cancers. PATIENTS AND METHODS: Adult patients with metastatic GI cancers referred for palliative RT to the chest, abdomen, or pelvis were eligible for study. Patients received IPdR orally once every day × 28 days beginning 7 days before the initiation of RT (37.5 Gy in 2.5 Gy × 15 fractions). A 2-part dose escalation scheme was used, pharmacokinetic studies were performed at multiple time points, and all patients were assessed for toxicity and response to Day 56. RESULTS: Nineteen patients were entered on study. Dose-limiting toxicity was encountered at 1,800 mg every day, and the recommended phase II dose is 1,200 mg every day. Pharmacokinetic analyses demonstrated achievable and sustainable levels of plasma IUdR ≥1 µmol/L (levels previously shown to mediate radiosensitization). Two complete, 3 partial, and 9 stable responses were achieved in target lesions. CONCLUSIONS: Administration of IPdR orally every day × 28 days with RT is feasible and tolerable at doses that produce plasma IUdR levels ≥1 µmol/L. These results support the investigation of IPdR + RT in phase II studies.


Assuntos
Quimiorradioterapia/métodos , Neoplasias Gastrointestinais/terapia , Idoxuridina/farmacocinética , Nucleosídeos de Pirimidina/administração & dosagem , Radiossensibilizantes/administração & dosagem , Administração Oral , Adulto , Idoso , Idoso de 80 Anos ou mais , Fracionamento da Dose de Radiação , Estudos de Viabilidade , Feminino , Neoplasias Gastrointestinais/patologia , Humanos , Idoxuridina/administração & dosagem , Idoxuridina/toxicidade , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Pró-Fármacos/administração & dosagem , Pró-Fármacos/farmacocinética , Pró-Fármacos/toxicidade , Nucleosídeos de Pirimidina/farmacocinética , Nucleosídeos de Pirimidina/toxicidade , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/toxicidade , Resultado do Tratamento
5.
Chem Biol Interact ; 240: 102-9, 2015 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-26296760

RESUMO

Fluorinated nucleoside analogues are a major class of cancer chemotherapy agents, and include the drugs 5-fluorouracil (5FU) and 5-fluoro-2'-deoxyuridine (FdUrd). The aim of this study was to examine the cellular toxicity of two novel fluorinated pyrimidine L-nucleosides that are enantiomers of D-nucleosides and may be able to increase selectivity for cancer cells as a result of their unnatural L-configuration. Two fluorinated pyrimidine L-nucleosides were examined in this study, L110 ([ß-L, ß-D]-5-fluoro-2'-deoxyuridine) and L117 (ß-L-deoxyuridine:ß-D-5'-fluoro-2'-deoxyuridine). The cytotoxicity of these L-nucleoside was determined in primary mouse fibroblasts and was compared with 5FU and FdUrd. In addition, the influence of p53 status on cytotoxicity was investigated. These cytotoxicity assays were performed on a matched set of primary mouse fibroblasts that were either wild type or null for the p53 tumour suppressor gene. It was found that cells lacking functional p53 were over 7500 times more sensitive to the drugs L110, L117 and FdUrd than cells containing wild type p53.


Assuntos
Antineoplásicos/toxicidade , Nucleosídeos de Pirimidina/toxicidade , Proteína Supressora de Tumor p53/metabolismo , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacologia , Desoxiuridina/uso terapêutico , Fluoruracila/farmacologia , Fluoruracila/uso terapêutico , Fluoruracila/toxicidade , Concentração Inibidora 50 , Camundongos , Estrutura Molecular , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/uso terapêutico
6.
Bioorg Med Chem ; 21(17): 4874-84, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23891229

RESUMO

Two sets of pyrimidine nucleoside derivatives bearing extended alkyloxymethyl or alkyltriazolidomethyl substituents at position 5 of the nucleobase were synthesized and evaluated as potential antituberculosis agents. The impact of modifications at 3'- and 5'-positions of the carbohydrate moiety on the antimycobacterial activity and cytotoxicity was studied. The highest effect was shown for 5-dodecyloxymethyl-2'-deoxyuridine, 5-decyltriazolidomethyl-2'-deoxyuridine, and 5-dodecyltriazolidomethyl-2'-deoxycytidine. They effectively inhibited the growth of two Mycobacterium tuberculosis strains in vitro, laboratory H37Rv (MIC99=20, 10, and 20µg/mL, respectively) and clinical MDR MS-115 resistant to five top antituberculosis drugs (МIC99=50, 10, and 10µg/mL, respectively).


Assuntos
Antituberculosos/síntese química , Nucleosídeos de Pirimidina/química , Animais , Antituberculosos/farmacologia , Antituberculosos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Chlorocebus aethiops , Humanos , Células Jurkat , Testes de Sensibilidade Microbiana , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/isolamento & purificação , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Relação Estrutura-Atividade , Células Vero
7.
Bioorg Med Chem ; 20(13): 4088-97, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22664188

RESUMO

Discovery of novel antimycobacterial compounds that work on distinctive targets and by diverse mechanisms of action is urgently required for the treatment of mycobacterial infections due to the emerging global health threat of tuberculosis. We have identified a new class of 5-ethyl or hydroxy (or methoxy) methyl-substituted pyrimidine nucleosides as potent inhibitors of Mycobacterium bovis, Mycobacterium tuberculosis (H37Ra, H37Rv) and Mycobacterium avium. A series of 2'-'up' fluoro (or hydroxy) nucleosides (1, 2, 4-6, 9, 10, 13, 16, 18, 21, 24) was synthesized and evaluated for antimycobacterial activity. Among 2'-fluorinated compounds, 1-(3-bromo-2,3-dideoxy-2-fluoro-ß-d-arabinofuranosyl)-5-ethyluracil (13) exhibited promising activity against M. bovis and Mtb alone, and showed synergism when combined with isoniazid. The most active compound emerging from these studies, 1-(ß-d-arabinofuranosyl)-4-thio-5-hydroxymethyluracil (21) inhibited Mtb (H37Ra) (MIC(50)=0.5 µg/mL) and M. bovis (MIC(50)=0.5 µg/mL) at low concentrations, and was ten times more potent against Mtb (H37Ra) than cycloserine (MIC(50)=5.0 µg/mL), a second line drug. It also showed an additive effect when combined with isoniazid. Compound 21 retained sensitivity against a rifampicin-resistant (H37Rv) strain of Mtb (MIC(50)=1 µg/mL) at concentrations similar to that for a rifampicin-sensitive (H37Rv) strain, suggesting that it has no cross-resistance to a first-line anti-TB drug. In addition, the replication of M. avium was also inhibited by 21 (MIC(50)=10 µg/mL). No cellular toxicity of 13 or 21 was observed up to the highest concentration tested (CC(50)>100 µg/mL). These observations offer promise for a new drug treatment regimen to augment and complement the current chemotherapy of TB.


Assuntos
Antituberculosos/química , Mycobacterium avium/efeitos dos fármacos , Mycobacterium bovis/efeitos dos fármacos , Mycobacterium tuberculosis/efeitos dos fármacos , Nucleosídeos de Pirimidina/química , Antituberculosos/farmacologia , Antituberculosos/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Farmacorresistência Bacteriana/efeitos dos fármacos , Humanos , Pentoxil (Uracila)/análogos & derivados , Pentoxil (Uracila)/química , Pentoxil (Uracila)/farmacologia , Pentoxil (Uracila)/toxicidade , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Uracila/análogos & derivados , Uracila/química , Uracila/farmacologia , Uracila/toxicidade
8.
Cancer Chemother Pharmacol ; 61(2): 323-34, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17562042

RESUMO

PURPOSE: A toxicology and pharmacokinetic study of orally administered (po) IPdR (5-3iodo-2-pyrimidinone-2'deoxyribose, NSC-726188) was performed in Fischer-344 rats using a once daily (qd) x 28 days dosing schedule as proposed for an initial phase I clinical trial of IPdR as a radiosensitizer. METHODS: For the toxicology assessment, 80 male and female rats (10/sex/dosage group) were randomly assigned to groups receiving either 0, 0.2, 1.0 or 2.0 g kg(-1)day(-1) of po IPdR x 28 days and one-half were observed to day 57 (recovery group). Animals were monitored for clinical signs during and following treatment with full necropsy of one-half of each dosage group at day 29 and 57. For the plasma pharmacokinetic assessment, 40 rats (10/sex/dosage group) were randomly assigned to groups receiving either 0.2 or 1.0 g kg(-1)day(-1) of po IPdR x 28 days with multiple blood samplings on days 1 and 28 and single blood sampling on days 8 and 15. RESULTS: No drug-related deaths occurred. Higher IPdR doses resulted in transient weight loss and transient decreased hemoglobins but had no effect on white cells or platelets. Complete serum chemistry evaluation showed transient mild decreases in total protein, alkaline phosphatase, and serum globulin. Necropsy evaluation at day 29 showed minimal to mild histopathologic changes in bone marrow, lymph nodes and liver; all reversed by day 59. There were no sex-dependent differences in plasma pharmacokinetics of IPdR noted and the absorption and elimination kinetics of IPdR were found to be linear over the dose range studied. CONCLUSIONS: A once-daily dosing schedule of po IPdR for 28 days with doses up to 2.0 g kg(-1)day(-1) appeared to be well tolerated in Fischer-344 rats. Drug-related weight loss and microscopic changes in bone marrow, lymph nodes and liver were observed. These changes were all reversed by day 57. IPdR disposition was linear over the dose range used. However, based on day 28 kinetics it appears that IPdR elimination is enhanced following repeated administration. These toxicology and pharmacokinetic data were used when considering the design of our initial phase I trial of po IPdR as a clinical radiosensitizer.


Assuntos
Nucleosídeos de Pirimidina/farmacocinética , Nucleosídeos de Pirimidina/toxicidade , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/toxicidade , Animais , Antimetabólitos/farmacocinética , Antimetabólitos/toxicidade , Área Sob a Curva , Disponibilidade Biológica , Contagem de Células Sanguíneas , Ensaios Clínicos Fase I como Assunto , Feminino , Meia-Vida , Idoxuridina/farmacocinética , Idoxuridina/toxicidade , Absorção Intestinal , Masculino , Ratos , Ratos Endogâmicos F344
9.
Antimicrob Agents Chemother ; 51(6): 2268-73, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17420214

RESUMO

Murine polyomavirus and simian virus 40 were used to evaluate the potencies of the compounds of three classes of acyclic nucleoside phosphonates: (i) the original HPMP (3-hydroxy-2-phosphonomethoxypropyl) and PME (2-phosphonomethoxyethyl) derivatives, (ii) the 6-[2-(phosphonomethoxy)alkoxy]-2,4-diaminopyrimidine (DAPy) derivatives, and (iii) a new class of HPMP derivatives containing a 5-azacytosine moiety. The last class showed the highest activities and selectivities against both polyomaviruses.


Assuntos
Antivirais/farmacologia , Nucleosídeos/farmacologia , Organofosfonatos/farmacologia , Polyomavirus/efeitos dos fármacos , Vírus 40 dos Símios/efeitos dos fármacos , Animais , Antivirais/química , Antivirais/classificação , Antivirais/toxicidade , Linhagem Celular , Cidofovir , Citosina/análogos & derivados , Citosina/farmacologia , Citosina/toxicidade , Camundongos , Testes de Sensibilidade Microbiana , Nucleosídeos/química , Nucleosídeos/classificação , Nucleosídeos/toxicidade , Organofosfonatos/química , Organofosfonatos/classificação , Organofosfonatos/toxicidade , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Pirimidinas/química , Pirimidinas/farmacologia , Pirimidinas/toxicidade
10.
Antimicrob Agents Chemother ; 51(6): 2240-3, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17371827

RESUMO

The susceptibilities of drug-resistant hepatitis B virus (HBV) mutants to lamivudine, adefovir, tenofovir, entecavir, and 2,4-diamino-6-[2-(phosphonomethoxy)ethoxy]-pyrimidine (PMEO-DAPym), a novel acyclic pyrimidine analogue, were assessed in vitro. Most drug-resistant mutants, including multidrug-resistant strains, remained sensitive to tenofovir and PMEO-DAPym. Therefore, the latter molecule deserves further evaluation for the treatment of HBV infection.


Assuntos
Antivirais/farmacologia , Farmacorresistência Viral Múltipla , Vírus da Hepatite B/efeitos dos fármacos , Mutação , Nucleosídeos de Pirimidina/farmacologia , Adenina/análogos & derivados , Adenina/química , Adenina/farmacologia , Adenina/toxicidade , Antivirais/química , Antivirais/toxicidade , Linhagem Celular Tumoral , Vírus da Hepatite B/enzimologia , Vírus da Hepatite B/genética , Hepatite B Crônica/virologia , Humanos , Testes de Sensibilidade Microbiana , Organofosfonatos/química , Organofosfonatos/farmacologia , Organofosfonatos/toxicidade , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/toxicidade , Inibidores da Transcriptase Reversa/química , Inibidores da Transcriptase Reversa/farmacologia , Inibidores da Transcriptase Reversa/toxicidade , Tenofovir , Replicação Viral/efeitos dos fármacos
11.
Biochemistry ; 45(4): 1087-98, 2006 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-16430205

RESUMO

To better understand nucleoside transport processes and intracellular fates of nucleosides, we have developed a pair of fluorescent nucleoside analogues, FuPmR and dFuPmR, that differ only in the sugar moiety (ribofuranosyl versus 2'-deoxy, respectively), for real-time analysis of nucleoside transport into living cells by confocal microscopy. The binding and transportability of the two compounds were assessed for five recombinant human nucleoside transporters (hENT1/2, hCNT1/2/3) produced in Saccharomyces cerevisiae and/or oocytes of Xenopus laevis. The ribosyl derivative (FuPmR) was used to demonstrate proof of principle in live cell imaging studies in 11 cultured human cancer cell lines with different hENT1 activities. The autofluorescence emitted from FuPmR enabled direct visualization of its movement from the extracellular medium into the intracellular compartment of live cells, and this process was blocked by inhibitors of hENT1 (nitrobenzylmercaptopurine ribonucleoside, dipyridamole, and dilazep). Quantitative analysis of fluorescence signals revealed two stages of FuPmR uptake: a fast first stage that represented the initial uptake rate (i.e., transport rate) followed by a slow long-lasting second stage. The accumulation of FuPmR and/or its metabolites in nuclei and mitochondria was also visualized by live cell imaging. Measurements of fluorescence intensity increases in nuclei and mitochondria revealed rate-limited processes of permeant translocation across intracellular membranes, demonstrating for the first time the intracellular distribution of nucleosides and/or nucleoside metabolites in living cells. The use of autofluorescent nucleosides in time-lapse confocal microscopy is a novel strategy to quantitatively study membrane transport of nucleosides and their metabolites that will provide new knowledge of nucleoside biology.


Assuntos
Corantes Fluorescentes/química , Proteínas de Transporte de Nucleosídeos/metabolismo , Nucleosídeos de Pirimidina/química , Animais , Sítios de Ligação/efeitos dos fármacos , Transportador Equilibrativo 1 de Nucleosídeo/metabolismo , Transportador Equilibrativo 2 de Nucleosídeo/metabolismo , Feminino , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/metabolismo , Células HeLa , Humanos , Proteínas de Membrana Transportadoras/metabolismo , Microscopia Confocal , Proteínas de Transporte de Nucleosídeos/química , Oócitos/metabolismo , Fotodegradação , Nucleosídeos de Pirimidina/síntese química , Nucleosídeos de Pirimidina/metabolismo , Nucleosídeos de Pirimidina/toxicidade , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Tioinosina/análogos & derivados , Tioinosina/metabolismo , Tioinosina/farmacologia , Fatores de Tempo , Células Tumorais Cultivadas , Xenopus laevis/genética , Xenopus laevis/metabolismo
12.
Antivir Chem Chemother ; 13(2): 67-82, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12238531

RESUMO

Twenty 5-alkyl-2-thiopyrimidine nucleosides were newly synthesized and examined for antiviral activities against herpes simplex virus (HSV), varicella-zoster virus (VZV) and human cytomegalovirus (HCMV). In this study, 2'-deoxy-5-alkyl-2-thiocytidine analogues had lower 50% effective concentration (EC50) values against HSV-1, and 2'-deoxy-5-alkyl-2-thiouridine analogues showed lower EC50 against VZV than their congeners of arabinoside form. Among the compounds examined, 2'-deoxy-5-ethyl and 5-propyl-2-thiocytidine (TN-53 and TN-54) were most potent and selective anti-HSV compounds. Their EC50s were 0.04 and 0.15 microM, and selectivity indexes were more than 7,215 and 1,849, respectively. On the other hand, 2'-deoxy-5-propyl-2-thiouridine (TN-51), 5-bromovinyl-2-thiouracil arabinoside (TN-65) and 5-styryl-2-thiouracil arabinoside (TN-67) were most potent and selective anti-VZV compounds. Their EC50s were 3.1, 3.8 and 2.6 pM for CaQu strain of VZV, respectively, and 2.1 to 3.0 times lower than that of acyclovir. All 2-thiopyrimidine nucleoside analogues did not show antiviral activities against thymidine kinase (TK) negative strains of HSV-1 and VZV. Only three 2-thiocytosine arabinoside compounds showed marginal anti-CMV activities (EC50s were 57-159 pM). All of the five alkyl-2-thio-pyrimidine nucleoside analogues examined were not cytotoxic to human lymphoblastoid cells (RPM18226) and human embryonic fibroblast cells (MRC-5) at 240 microM (100 microg/ml) or more. Regarding the structure-activity relationship of 5-alkyl-2-thiopyrimidine nucleoside analogues, the following remarks will be noted. Elongation of 5-alkyl chain (methyl to ethyl) of 2-thiocytosine in both deoxyribosyl and arabinosyl nucleosides increased anti-HSV-1 activity but not anti-VZV activity. Furthermore, elongation of the same chain (ethyl to propyl) of 2-thiodeoxyuridine increased anti-VZV activity whereas it did not in the case of 2-thiouracil arabinosides.


Assuntos
Antivirais/síntese química , Herpesviridae/efeitos dos fármacos , Nucleosídeos de Pirimidina/farmacologia , Tionucleosídeos/farmacologia , Antivirais/farmacologia , Antivirais/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Citomegalovirus/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Herpesvirus Humano 3/efeitos dos fármacos , Humanos , Nucleosídeos de Pirimidina/síntese química , Nucleosídeos de Pirimidina/toxicidade , Relação Estrutura-Atividade , Tionucleosídeos/síntese química , Tionucleosídeos/toxicidade , Células Tumorais Cultivadas
13.
J Med Chem ; 45(10): 2032-40, 2002 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-11985471

RESUMO

A novel class of 5-substituted acyclic pyrimidine nucleosides, 1-[(2-hydroxyethoxy)methyl]-5-(1-azidovinyl)uracil (9a), 1-[(2-hydroxy-1-(hydroxymethyl)ethoxy)methyl]-5-(1-azidovinyl)uracil (9b), and 1-[4-hydroxy-3-(hydroxymethyl)-1-butyl]-5-(1-azidovinyl)uracil (9c), were synthesized by regiospecific addition of bromine azide to the 5-vinyl substituent of the respective 5-vinyluracils (2a-c) followed by treatment of the obtained 5-(1-azido-2-bromoethyl) compounds (3a-c) with t-BuOK, to affect the base-catalyzed elimination of HBr. Thermal decomposition of 9b and 9c at 110 degrees C in dioxane yielded corresponding 5-[2-(1-azirinyl)]uracil analogues (10b,c). The 5-(1-azidovinyl)uracil derivatives 9a-c were found to exhibit potent and selective in vitro anti-HBV activity against duck hepatitis B virus (DHBV) infected primary duck hepatocytes at low concentrations (EC(50) = 0.01-0.1 microg/mL range). The most active anti-DHBV agent (9c), possessing a [4-hydroxy-3-(hydroxymethyl)-1-butyl] substituent at N-1, exhibited an activity (EC(50) of 0.01-0.05 microg/mL) comparable to that of reference compound (-)-beta-L-2',3'-dideoxy-3'-thiacytidine (3-TC) (EC(50) = 0.01-0.05 microg/mL). In contrast, related 5-[2-(1-azirinyl)]uracil analogues (10b,c) were devoid of anti-DHBV activity, indicating that an acyclic side chain at C-5 position of the pyrimidine ring is essential for anti-HBV activity. The pyrimidine nucleosides (9a-c, 10b,c) exhibited no cytotoxic activity against a panel of 60 human cancer cell lines. All of the compounds investigated did not show any detectable toxicity to several stationary and proliferating host cell lines or to mitogen stimulated proliferating human T lymphocytes, up to the highest concentration tested.


Assuntos
Antivirais/síntese química , Vírus da Hepatite B/efeitos dos fármacos , Nucleosídeos de Pirimidina/síntese química , Animais , Antineoplásicos/farmacologia , Antivirais/química , Antivirais/farmacologia , Antivirais/toxicidade , Divisão Celular , Células Cultivadas , DNA/biossíntese , Ensaios de Seleção de Medicamentos Antitumorais , Patos , Vírus da Hepatite B do Pato/efeitos dos fármacos , Hepatócitos/efeitos dos fármacos , Hepatócitos/virologia , Humanos , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Relação Estrutura-Atividade , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo
14.
Clin Cancer Res ; 6(9): 3670-9, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10999760

RESUMO

We have demonstrated previously an improved therapeutic index for oral 5-iodo-2-deoxypyrimidinone-2'-deoxyribose (IPdR) compared with oral and continuous infusion of 5-iodo-2'-deoxyuridine (IUdR) as a radiosensitizing agent using three different human tumor xenografts in athymic mice. IPdR is a prodrug that is efficiently converted to IUdR by a hepatic aldehyde oxidase, resulting in high IPdR and IUdR plasma levels in mice for > or =1 h after p.o. IPdR. Athymic mice tolerated oral IPdR at up to 1500 mg/kg/day given four times per day for 6-14 days without significant systemic toxicities. In anticipation of an investigational new drug application for the first clinical Phase I and pharmacology study of oral IPdR in humans, we studied the drug pharmacokinetics and host toxicities in two non-rodent, animal species. For the IPdR systemic toxicity and toxicology study, twenty-four male or female ferrets were randomly assigned to four IPdR dosage groups receiving 0, 15, 150, and 1500 mg/kg/day by oral gavage x 14 days prior to sacrifice on study day 15. All ferrets survived the 14-day treatment. Ferrets receiving 1500 mg/kg/day showed observable systemic toxicities with diarrhea, emesis, weight loss, and decreased motor activity beginning at days 5-8 of the 14-day schedule. Overall, both male and female ferrets receiving IPdR at 1500 mg/kg/day experienced significant weight loss (9 and 19%, respectively) compared with controls after the 14-day treatment. No weight loss or other systemic toxicities were observed in other IPdR dosage groups. Grossly, no anatomical lesions were noted at complete necropsy, although liver weights were increased in both male and female ferrets in the two higher IPdR dosage groups. Histologically, IPdR-treated animals showed dose-dependent microscopic changes in liver consisting of minimal to moderate cytoplasmic vacuolation of hepatocytes, which either occurred in the periportal area (high dosage group) or diffusely throughout the liver (lower dosage groups). Female ferrets in the highest IPdR dose group also showed decreased kidney and uterus weights at autopsy without any associated histological changes. No histological changes were found in central nervous system tissues. No significant abnormalities in blood cell counts, liver function tests, kidney function tests, or urinalysis were noted. Hepatic aldehyde oxidase activity was decreased to approximately 50 and 30% of control ferrets in the two higher IPdR dosage groups, respectively, after the 14-day treatment period. The % IUdR-DNA incorporation in ferret bone marrow at the completion of IPdR treatment was < or =0.05% in the two lower dosage groups and approximately 2% in the 1500 mg/kg/day dosage group. The % IUdR-DNA in normal liver was < or =0.05% in all IPdR dosage groups. In a pharmacokinetic study in four Rhesus monkeys, we determined the plasma concentrations of IPdR after a single i.v. bolus of 50 mg/kg over 20 min. Using a two-compartment model to fit the plasma pharmacokinetic data, we found that IPdR was cleared in these non-human primates in a biexponential manner with an initial rapid distributive phase (mean T1/2alpha = 6.5 min), followed by an elimination phase with a mean T1/2 of 63 min. The mean maximum plasma concentration of IPdR was 124+/-43 microM with a mean total body clearance of 1.75+/-0.95 l/h/kg. IPdR was below detection (<0.5 microM) in the cerebrospinal fluid. We conclude that there are dose-limiting systemic toxicities to a 14-day schedule of p.o. IPdR at 1500 mg/kg/day in ferrets that were not found previously in athymic mice. However, no significant hematological, biochemical, or histopathological changes were found. Hepatic aldehyde oxidase activity was reduced in a dose-dependent in ferret liver, suggesting partial enzyme saturation by this IPdR schedule. The plasma pharmacokinetic profile in Rhesus monkeys showing biexponential clearance is similar to our published data in athymic mice. These data are being applied


Assuntos
Nucleosídeos de Pirimidina/farmacocinética , Nucleosídeos de Pirimidina/toxicidade , Radiossensibilizantes/farmacocinética , Radiossensibilizantes/toxicidade , Aldeído Oxirredutases/metabolismo , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , DNA/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Furões , Testes Hematológicos , Idoxuridina/metabolismo , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Macaca mulatta , Masculino , Pró-Fármacos/farmacocinética , Pró-Fármacos/toxicidade , Estômago/efeitos dos fármacos , Estômago/patologia , Urina/química
15.
Clin Cancer Res ; 6(4): 1468-75, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10778979

RESUMO

In anticipation of an initial clinical Phase I trial in patients with high-grade gliomas of p.o. administered 5-iodo2-pyrimidinone-2'-deoxyribose (IPdR) given daily for 14 days as a prodrug for 5-iodo-2'-deoxyuridine (IUdR)-mediated tumor radiosensitization, we determined the systemic toxicities and the percentage IUdR-DNA incorporation in normal athymic mouse tissues and a human glioblastoma xenograft (U251) after this dosing schedule of IPdR. Using a tumor regrowth assay of s.c. U251 xenografts, we also compared radiosensitization with this IPdR-dosing schedule to radiation therapy (XRT) alone (2 Gy/day for 4 days) or to XRT after continuous infusion IUdR for 14 days at the maximum tolerated dose in mice (100 mg/kg/day). Athymic mice with and without U251 s.c. xenografts tolerated 750 or 1500 mg/kg/day of p.o. IPdR (using gastric lavage) for 14 days without weight loss or activity level changes during treatment and for a 28-day posttreatment observation period. The percentage IUdR-DNA incorporation in U251 tumor cells was significantly higher after p.o. IPdR (750 and 1500 mg/kg/day) for 14 days (3.1 +/- 0.2% and 3.7 +/- 0.3%, respectively) than continuous infusion IUdR for 14 days (1.4 +/- 0.1%). Compared to XRT alone, a significant sensitizer enhancement ratio (SER) was found with the combination of p.o. IPdR (1500 mg/kg/d) + XRT (SER = 1.31; P = 0.05) but not for the combination of continuous infusion IUdR + XRT (SER = 1.07; P = 0.57) in the U251 xenografts. The percentage IUdR-DNA incorporation after IPdR at 1500 mg/kg/day for 14 days in normal bone marrow, normal small intestine, and normal liver were 1.2 +/-0.2%, 3.3 +/- 0.3%, and 0.2 +/- 0.1%, respectively. We conclude that a 14-day p.o. schedule of IPdR at up to 1500 mg/kg/day results in no significant systemic toxicity in athymic mice and is associated with significant radiosensitization using this human glioblastoma multiforme xenograft model. Based on these data and our previously published data using shorter IPdR dosing schedules, which also demonstrate an improved therapeutic index for IPdR compared to IUdR, an initial clinical Phase I and pharmacokinetic study of p.o. IPdR daily for 14 days is being designed.


Assuntos
Idoxuridina/uso terapêutico , Neoplasias Experimentais/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Nucleosídeos de Pirimidina/uso terapêutico , Radiossensibilizantes/uso terapêutico , Administração Oral , Aldeído Oxidase , Aldeído Oxirredutases/efeitos dos fármacos , Aldeído Oxirredutases/metabolismo , Animais , Peso Corporal/efeitos dos fármacos , DNA/efeitos dos fármacos , DNA/genética , DNA/metabolismo , Esquema de Medicação , Feminino , Humanos , Idoxuridina/metabolismo , Extratos Hepáticos/metabolismo , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Neoplasias Experimentais/radioterapia , Nucleosídeos de Pirimidina/metabolismo , Nucleosídeos de Pirimidina/toxicidade , Transplante Heterólogo , Células Tumorais Cultivadas
16.
J Med Chem ; 42(7): 1320-8, 1999 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-10197975

RESUMO

The synthesis of L-nucleoside analogues containing 2'-vinylic fluoride was accomplished by direct condensation method, and their anti-HIV and anti-HBV activities were evaluated in vitro. The key intermediate 8, the sugar moiety of our target compounds, was prepared from 1,2-O-isopropylidene-L-glyceraldehyde via (R)-2-fluorobutenolide intermediate 5 in five steps. Coupling of the acetate 8 with the appropriate heterocycles (silylated uracil, thymine, N4-benzoylcytosine, N4-benzoyl-5-fluorocytosine, 6-chloropurine, and 6-chloro-2-fluoropurine) in the presence of Lewis acid afforded a series of 2'-fluorinated L-nucleoside analogues (15-18, 23-26, 36-45). The newly synthesized compounds were evaluated for their antiviral activities against HIV-1 in human peripheral blood mononuclear (PBM) cells and HBV in 2.2.15 cells. Cytosine 23, 5-fluorocytosine 25, and adenine 36 derivatives exhibited moderate to potent anti-HIV (EC50 0.51, 0.17, and 1.5 microM, respectively) and anti-HBV (EC50 0.18, 0.225, and 1.7 microM, respectively) activities without significant cytotoxicity up to 100 microM in human PBM, Vero, CEM, and HepG2 cells.


Assuntos
Adenosina/análogos & derivados , Fármacos Anti-HIV/síntese química , Citosina/análogos & derivados , Vírus da Hepatite B/efeitos dos fármacos , Nucleosídeos de Purina/síntese química , Nucleosídeos de Pirimidina/síntese química , Adenosina/síntese química , Adenosina/química , Adenosina/farmacologia , Adenosina/toxicidade , Fármacos Anti-HIV/química , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/toxicidade , Linhagem Celular , Citosina/síntese química , Citosina/química , Citosina/farmacologia , Citosina/toxicidade , HIV-1/efeitos dos fármacos , Humanos , Técnicas In Vitro , Concentração Inibidora 50 , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/virologia , Nucleosídeos de Purina/química , Nucleosídeos de Purina/farmacologia , Nucleosídeos de Purina/toxicidade , Nucleosídeos de Pirimidina/química , Nucleosídeos de Pirimidina/farmacologia , Nucleosídeos de Pirimidina/toxicidade , Relação Estrutura-Atividade , Replicação Viral/efeitos dos fármacos
18.
Clin Cancer Res ; 4(1): 99-109, 1998 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9516958

RESUMO

We reported previously that p.o. administered 5-iodo-2-pyrimidinone-2'-deoxyribose (IPdR) was efficiently converted to 5-iodo-2'-deoxyuridine (IUdR) in athymic mice (T. J. Kinsella et al., Cancer Res., 54: 2695-2700, 1994). Here, we further evaluate IPdR metabolism, systemic toxicity, and percentage DNA incorporation in athymic mouse normal tissues and a human colon cancer xenograft (HT29) using higher p.o. doses of IPdR. These data are compared to results using a continuous infusion of IUdR at the maximum tolerable dose. We also evaluate IPdR metabolism in cytosolic extracts from normal human liver, normal human intestine, and human colorectal cancer specimens. Athymic mice tolerated a daily p.o. bolus of up to 2 g/kg IPdR for 6 days with minimal host toxicity (< or = 10% body weight loss). There was rapid conversion of IPdR to IUdR, with peak plasma levels of IUdR of 40-75 microM at 10 min following a p.o. IPdR bolus of 250-1500 mg/kg. The percentage IUdR-DNA in the HT29 s.c. human tumor xenografts increased 1.5 times (2.3-3.6%) with IPdR doses above 1 g/kg/day for 6 days, whereas the percentage IUdR-DNA incorporation in two proliferating normal tissues (4-4.5% in intestine; 1.6-2.2% in bone marrow) and a quiescent normal tissue (< or = 1% in liver) showed < 1.5-fold increases with the IPdR dose escalation between 1-2 g/kg/day for 6 days. In contrast, using a continuous infusion of IUdR at 100 mg/kg/day, significant systemic toxicity (> 20% body weight loss) was found by day 6 of the infusion. Steady-state plasma IUdR levels were 1.0-1.2 microM during the 6-day infusion, and percentage IUdR-DNA incorporations of 2.3, 8, 6, and 1% were measured in s.c. tumors, normal intestine, normal bone marrow, and normal liver, respectively, following the 6-day infusion. Thus, the p.o. IPdR schedule has an improved therapeutic index, based on percentage IUdR-DNA incorporation in normal and tumor tissues, compared to continuous infusion IUdR at the maximum tolerable dose in athymic mice with this human tumor xenograft. Additionally, a tumor regrowth assay to assess the radiation response of HT29 s.c. xenografts showed a 1.5-fold enhancement (time to regrow to 300% initial tumor volume) with IPdR (1000 mg/kg/day for 6 days) plus fractionated irradiation (XRT; 2 Gy/day for 4 days), compared to XRT (2 Gy/day for 4 days) alone. No enhancement in the radiation response of HT29 s.c. xenografts was found with continuous infusion IUdR (100 mg/kg/day for 6 days) plus XRT (2 Gy/day for 4 days), compared to XRT alone. Using cytosolic extracts from normal human liver specimens, we found a rapid (15-min) conversion of IPdR to IUdR. Coincubation of liver cytosol with IPdR and allopurinol, an inhibitor of xanthine oxidase, had no inhibitory effect on IPdR metabolism, whereas coincubation with IPdR and isovanillin or menadione, analogue substrates for aldehyde oxidase, effectively reduced the amount of IPdR oxidized to IUdR. Significantly less metabolism of IPdR to IUdR was seen in cytosolic extracts from normal human intestine specimens, and no metabolism of IPdR was found in cytosolic extracts from colorectal liver metastases in two patients and from the HT29 human colon cancer xenografts in athymic mice. These additional data indicate that IPdR has the potential for clinical use as a p.o. prodrug for IUdR-mediated radiosensitization of resistant human cancers.


Assuntos
Idoxuridina/uso terapêutico , Pró-Fármacos/uso terapêutico , Nucleosídeos de Pirimidina/uso terapêutico , Radiossensibilizantes/uso terapêutico , Animais , DNA/metabolismo , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais/radioterapia , Nucleosídeos de Pirimidina/metabolismo , Nucleosídeos de Pirimidina/toxicidade , Células Tumorais Cultivadas
19.
Biochem Pharmacol ; 48(11): 2117-26, 1994 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-7528507

RESUMO

We examined the cytotoxicity, biochemical effects and metabolism of 4-methoxy-8-(beta-D-ribofuranosylamino)pyrimido[5,4-d]pyrimidine (MRPP), a synthetic nucleoside inhibitor of phosphoribosylpyrophosphate synthetase, in HCT 116 human colorectal cancer cells. A 4-hr exposure to 1 and 10 microM MRPP inhibited cell growth over a 72-hr period by 76 and 89%, and inhibited clonogenic capacity by 36 and 65%, respectively. MRPP was avidly metabolized to the 5'-monophosphate derivative (MRPP-MP), and MRPP-MP formation increased with increasing MRPP exposure (microM.hr). MRPP-MP was stable, and the intracellular half-life was in excess of 48 hr. A 4-hr exposure to 10 microM MRPP resulted in significant decreases in ATP, UTP, GTP, CTP, dATP, dTTP, and PRPP pools. Near maximal ribonucleotide triphosphate depletion was achieved with > or = 24 microM.hr MRPP, and growth inhibition as a function of MRPP microM.hr closely reflected the biochemical effects. Ribonucleotide triphosphate pools remained depleted for up to 48 hr after drug removal, apparently as a consequence of the prolonged retention of MRPP-MP. MRPP (10 microM) inhibited the salvage of [3H]guanine, [3H]adenine and [3H]guanosine, and concurrent exposure to MRPP and either 100 microM adenine, hypoxanthine, or guanine did not reverse ATP or GTP depletion. Concurrent exposure to 10 microM MRPP and either 10 microM adenosine, uridine or thymidine was accompanied by repletion of ATP, UTP, and dTTP pools, respectively, but depletion of other nucleotide pools was not corrected. In contrast, 10 microM guanosine did not correct GTP depletion in the presence of MRPP. The combination of 10 microM each of thymidine, uridine, adenosine and guanosine during and following a 24-hr exposure to MRPP provided partial protection against 0.1 or 1 microM MRPP, but did not affect the cytotoxicity associated with 10 microM MRPP. MRPP is a novel antimetabolite that inhibits both de novo and salvage pathways for purine synthesis and de novo pyrimidine synthesis.


Assuntos
Antimetabólitos/toxicidade , Nucleosídeos de Pirimidina/toxicidade , Trifosfato de Adenosina/metabolismo , Antimetabólitos/metabolismo , Linhagem Celular , Citidina Trifosfato/metabolismo , DNA/biossíntese , DNA/isolamento & purificação , Humanos , Concentração de Íons de Hidrogênio , Nucleosídeos/farmacologia , Fosforilação , Nucleosídeos de Pirimidina/metabolismo , RNA/biossíntese , Uridina Trifosfato/metabolismo
20.
Cancer Res ; 54(10): 2695-700, 1994 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8168099

RESUMO

5-iodo-2-pyrimidinone-2'-deoxyribose (IPdR) was recently reported to be converted to 5-iodo-2'-deoxyuridine (IUdR) by an aldehyde oxidase, most concentrated in liver tissue. We questioned whether IPdR could be used as a p.o. hepatotropic prodrug to increase the percentage of IUdR-DNA incorporation into liver tumors compared to normal liver with acceptable systemic toxicity. Athymic nude mice with human colon cancer (HCT-116) xenograft tumors as liver metastases and s.c. flank tumors received daily p.o. boluses (via gastric tubes) of IUdR or IPdR for 6 days. The maximum tolerated dose of IUdR was 250 mg/kg/day and was associated with a > 10% weight loss and a high percentage of IUdR-DNA incorporation (> 5%) into normal bone marrow and intestine. In contrast, animals tolerated escalating doses of IPdR to 1 gm/kg/day without weight loss and with less (1.5-4%) IUdR-DNA incorporation in normal tissues. Pharmacokinetic analysis of p.o. IPdR showed peak plasma levels of IPdR and IUdR within 15-45 min, suggesting efficient conversion of IPdR to IUdR. Aldehyde oxidase activity was found in normal liver tissue but not in other normal or tumor tissues. Additionally, we found a 2-3 times greater percentage of IUdR-DNA incorporation in tumor with IPdR than IUdR at the highest doses used. However, no differential effect in the percentage of IUdR-DNA incorporation was noted between liver metastases and s.c. tumors with either IPdR or IUdR. We conclude that p.o. IPdR offers a greater therapeutic index for tumor incorporation (and presumably radiosensitization) than a similar schedule of IUdR.


Assuntos
DNA/metabolismo , Idoxuridina/farmacocinética , Neoplasias Hepáticas/metabolismo , Fígado/metabolismo , Nucleosídeos de Pirimidina/farmacocinética , Administração Oral , Animais , Neoplasias do Colo/metabolismo , Feminino , Humanos , Idoxuridina/administração & dosagem , Idoxuridina/toxicidade , Neoplasias Hepáticas/secundário , Camundongos , Transplante de Neoplasias , Nucleosídeos de Pirimidina/administração & dosagem , Nucleosídeos de Pirimidina/toxicidade , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA