Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Cancer Chemother Pharmacol ; 48(2): 145-50, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11565561

RESUMO

PURPOSE: The purpose of this investigation was to evaluate the ability of oral PTAU, 5-(phenylthio)acyclouridine, to increase the concentration of endogenous plasma uridine. PTAU is a new potent and specific inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. This compound was designed as a lipophilic inhibitor in order to facilitate its access to the liver and intestine, the main organs involved in uridine catabolism. METHODS: PTAU was administered to mice orally and parenterally. The plasma levels of PTAU as well as those of uridine and its catabolite uracil were measured by HPLC, and pharmacokinetic analysis was performed. RESULTS: PTAU was fully adsorbed after oral administration (over 100% oral bioavailability) and no PTAU metabolites were detected. PTAU administered orally had no apparent toxicity at doses up to 120 mg/kg per day for 5 days. Parenteral administration of PTAU at 30, 45 and 60 mg/kg increased the concentration of endogenous plasma uridine (1.8 +/- 0.2 microM) by approximately six-, seven-, and nine-fold, respectively. Plasma uridine concentration remained higher than control values until 8 h after PTAU administration. Similar results were obtained following oral administration of PTAU. The baseline concentrations of endogenous plasma uridine were increased by approximately six-, seven- and ten-fold by oral administration of PTAU at 30, 45 and 60 mg/kg, respectively, and remained higher than the controls until 8 h after PTAU administration. PTAU did not alter the concentration of endogenous plasma uracil. CONCLUSION: The effectiveness of the PTAU in elevating and sustaining high plasma uridine concentrations may be useful in rescuing or protecting the host from toxicities of various chemotherapeutic pyrimidine analogues as well as in the management of medical disorders that respond to the administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Tiouracila/análogos & derivados , Tiouracila/farmacologia , Uridina/sangue , Animais , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/toxicidade , Feminino , Camundongos , Tiouracila/sangue , Tiouracila/farmacocinética , Tiouracila/toxicidade , Uridina Fosforilase/antagonistas & inibidores
2.
Antivir Chem Chemother ; 12(2): 99-108, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11527047

RESUMO

The beta-L-nucleoside analogue beta-L-2',3'-dideoxy adenosine (beta-L-ddA) has been shown to exhibit limited antiviral activities. This was attributed to its rapid catabolism through cleavage of the glycosidic bond and poor phosphorylation to the nucleotide beta-L-2',3'-dideoxyadenosine-5'-mono phosphate (beta-L-ddAMP) (Placidi et al., 2000). However, the nucleotide beta-L-2',3'-dideoxyadenosine-5'-triphosphate (beta-L-ddATP) inhibited the activity of both HIV-1 reverse transcriptase (RT) and viral DNA polymerase isolated from woodchuck hepatitis virus-infected serum (a model of hepatitis B) with an inhibitory concentration (IC50) of 2.0 microM without inhibiting human DNA polymerases alpha, beta, or gamma up to a concentration of 100 microM. These results suggested that prodrugs of beta-L-ddAMP may bypass the poor metabolic activation of beta-L-ddA and lead to more potent and selective antiviral activity. Therefore, the mononucleoside phosphotriester derivative of beta-L-ddAMP incorporating the S-pivaloyl-2-thioethyl (tButylSATE) groups, beta-L-ddAMP-bis(tButylSATE) was synthesized. Beta-L-ddAMP-bis(tButylSATE) inhibited HIV replication in human peripheral blood mononuclear cells (PBMCs) and HBV replication in 2.2.15 cells with effective concentrations (EC50s) of 2 and 80 nM, respectively. Intracellular metabolism of beta-L-ddAMP-bis(tButylSATE) demonstrated that beta-L-ddATP was the predominant intracellular metabolite in PBMC and liver cells. The intracellular half-life of beta-L-ddATP was 5.4 and 9.2 h in HepG2 and PBMCs, respectively. The intracellular concentrations of beta-L-ddATP were maintained above the EC50 for the inhibition of HIV RT and hepatitis B virus (HBV) for as long as 24 h after removal of the drug.


Assuntos
Antivirais/metabolismo , Antivirais/farmacologia , Didesoxiadenosina/farmacologia , HIV/efeitos dos fármacos , Vírus da Hepatite B/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Animais , Fármacos Anti-HIV/metabolismo , Fármacos Anti-HIV/farmacologia , Cromatografia Líquida de Alta Pressão , DNA Polimerase Dirigida por DNA/metabolismo , Didesoxiadenosina/análogos & derivados , Didesoxiadenosina/metabolismo , Didesoxinucleotídeos , HIV/enzimologia , HIV/fisiologia , Meia-Vida , Células-Tronco Hematopoéticas/efeitos dos fármacos , Vírus da Hepatite B/enzimologia , Vírus da Hepatite B/fisiologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Concentração Inibidora 50 , Lamivudina/farmacologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/virologia , Marmota/sangue , Marmota/virologia , Inibidores da Síntese de Ácido Nucleico , DNA Polimerase Dirigida por RNA/metabolismo , Inibidores da Transcriptase Reversa/farmacologia , Células Tumorais Cultivadas
3.
Nucleosides Nucleotides Nucleic Acids ; 20(12): 1915-25, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11794797

RESUMO

An asymmetric synthesis of carbocyclic orotidine 15 and its monophosphate 16 were accomplished via the key intermediate cyclopentanone 4, which was prepared from D-gamma-ribonolactone in steps. None of synthesized the compounds inhibited orotidine 5'-monophosphate decarboxylase (EC 4.1.1.23) or orotate phosphoribosyltransferase (EC 2.4.2.10).


Assuntos
Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Orotidina-5'-Fosfato Descarboxilase/antagonistas & inibidores , Uridina/análogos & derivados , Ácidos Carbocíclicos/química , Animais , Bioquímica/métodos , Inibidores Enzimáticos/metabolismo , Feminino , Humanos , Fígado/efeitos dos fármacos , Fígado/enzimologia , Camundongos , Camundongos Endogâmicos , Orotato Fosforribosiltransferase/antagonistas & inibidores , Relação Estrutura-Atividade , Uridina/química
4.
Cancer Chemother Pharmacol ; 48(5): 389-97, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11761457

RESUMO

PURPOSE: The purpose of this investigation was to evaluate the effectiveness of oral 5-(phenylthio)acyclouridine (PTAU) in improving the oral bioavailability of uridine. PTAU is a new potent and specific inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. This compound was designed as a lipophilic inhibitor in order to facilitate its access to the liver and intestine, the main organs involved in uridine catabolism. PTAU is not toxic to mice and is fully absorbed after oral administration (100% oral bioavailability). METHODS: PTAU was administered orally to mice alone or with uridine. The plasma levels of PTAU as well as those of uridine and its catabolite uracil were measured using HPLC, and pharmacokinetic analysis was performed. RESULTS: Co-administration of PTAU with uridine elevated the concentration of plasma uridine in a dose-dependent manner over that resulting from the administration of the same dose of uridine alone, and reduced the clearance of uridine as well as the peak plasma concentration (Cmax) and area under the curve (AUC) of plasma uracil. Coadministration of PTAU at 30, 45 and 60 mg/kg improved the low oral bioavailability (7.7%) of uridine administered at 1320 mg/kg by 4.3-, 5.9- and 9.9-fold, respectively, and reduced the AUC of plasma uracil (1227.8 micromol x h/l) by 5.7-, 6.8- and 8.2-fold, respectively. Similar results were observed when PTAU was coadministered with lower doses of uridine. Oral PTAU at 30, 45 and 60 mg/kg improved the oral bioavailability of 330 mg/kg uridine by 1.8-, 2.6- and 2.8-fold, and that of 660 mg/kg uridine by 2.2-, 2.6- and 3.2-fold, respectively. CONCLUSION: The effectiveness of PTAU in improving the oral bioavailability of uridine could be useful in the rescue or protection from host toxicities of various chemotherapeutic pyrimidine analogues as well as in the management of medical disorders that are remedied by administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Tiouracila/análogos & derivados , Tiouracila/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Uridina/farmacocinética , Administração Oral , Animais , Área Sob a Curva , Disponibilidade Biológica , Feminino , Camundongos
5.
Cancer Chemother Pharmacol ; 46(3): 235-40, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-11021741

RESUMO

PURPOSE: The purpose of this investigation was to study the effects of combining oral 5-(phenylselenenyl)acyclouridine (PSAU) with 2',3',5'-tri-O-acetyluridine (TAU) on the levels of plasma uridine in mice. PSAU is a new lipophilic and potent inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. PSAU has 100% oral bioavailability and is a powerful enhancer of the bioavailability of oral uridine. TAU is a prodrug of uridine and a far superior source of uridine than uridine itself. METHODS: Oral TAU was administered to mice alone or with PSAU. The plasma levels of uridine and its catabolites as well as PSAU were measured using HPLC and pharmacokinetic analysis was performed. RESULTS: Oral administration of 2000 mg/kg TAU increased plasma uridine by over 250-fold with an area under the curve (AUC) of 754 micromol x h/l. Coadministration of PSAU at 30 and 120 mg/kg with TAU further improved the bioavailability of plasma uridine resulting from the administration of TAU alone by 1.7- and 3.9-fold, respectively, and reduced the Cmax and AUC of plasma uracil. CONCLUSION: The exceptional effectiveness of PSAU plus TAU in elevating and sustaining a high plasma uridine concentration could be useful in the management of medical disorders that are remedied by administration of uridine, as well as the rescue or protection from host toxicities of various chemotherapeutic pyrimidine analogues.


Assuntos
Inibidores Enzimáticos/farmacologia , Compostos Organosselênicos/farmacologia , Pró-Fármacos/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia , Uridina/sangue , Uridina/farmacologia , Acetatos , Animais , Disponibilidade Biológica , Sinergismo Farmacológico , Inibidores Enzimáticos/sangue , Inibidores Enzimáticos/farmacocinética , Feminino , Camundongos , Compostos Organosselênicos/sangue , Compostos Organosselênicos/farmacocinética , Pró-Fármacos/farmacocinética , Uracila/sangue , Uracila/farmacocinética , Uridina/análogos & derivados , Uridina/farmacocinética , Uridina Fosforilase/antagonistas & inibidores
6.
Biochem Pharmacol ; 60(5): 687-92, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10927027

RESUMO

The effect of co-administration of 5-(phenylselenenyl)acyclouridine (PSAU), a new uridine phosphorylase (UrdPase, EC 2.4.2.3) inhibitor, on the efficacy of 5-fluoro-2'-deoxyuridine (FdUrd) was tested against murine colon C26-10 tumor xenografts. In contrast to our previous results with human tumors, co-administration of PSAU with FdUrd decreased instead of increasing the efficacy of FdUrd against tumor growth. However, co-administration of PSAU with FdUrd (300 mg/kg/day) protected the mice completely from the 83% mortality induced by the same dose of FdUrd alone. Enzyme studies indicated that UrdPase in colon C26-10 tumors is responsible for the catabolism of FdUrd to 5-fluorouracil (FUra), as colon C26-10 tumors do not have thymidine phosphorylase (dThdPase, EC 2.4.2.4). In contrast, colon C26-10 tumors had extraordinarily high UrdPase activity (300 micromol/min/mg protein), which was at least 200-fold higher than the highest UrdPase activity in any of the human xenografts we tested previously. Furthermore, the activities of UrdPase and orotate phosphoribosyltransferase (OPRTase, EC 2.4.2.10) were 192- and 2-fold higher, respectively, while that of dihydrouracil dehydrogenase (EC 1.3.1.2) was 1000-fold lower in the tumor than in the host liver. It is suggested that FdUrd exerts its anticancer effects against colon C26-10 tumors mainly through the catabolism of FdUrd to FUra by UrdPase, which then could be anabolized to 5-fluorouridine 5'-monophosphate (FUMP) by OPRTase and ultimately to other toxic 5-fluorouridine nucleotides, hence inducing the observed FdUrd toxic effects. Co-administration of PSAU with FdUrd inhibited UrdPase and the catabolism of FdUrd to FUra. This would result in the observed reduction of the antitumor efficacy of FdUrd. In addition, the increase in plasma uridine concentration induced by PSAU as well as the catabolism of FUra by the high dihydrouracil dehydrogenase activity in the liver also may have circumvented any residual FUra toxic effects against the host. These results clearly demonstrate that the anticancer efficacy of the combination of UrdPase inhibitors and FdUrd is not general and is dependent largely on the type of tumor under treatment and the mode of FdUrd metabolism in these tumors.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Floxuridina/farmacologia , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Uracila/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Interações Medicamentosas , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Transplante Heterólogo , Células Tumorais Cultivadas
7.
Biochem Pharmacol ; 60(6): 851-6, 2000 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-10930540

RESUMO

5-Phenylthioacyclouridine (PTAU or 1-[(2-hydroxyethoxy)methyl]-5-phenylthiouracil) was synthesized as a highly specific and potent inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2.3). PTAU has inhibition constant (K(is)) values of 248 and 353 nM towards UrdPase from mouse and human livers, respectively. PTAU was neither an inhibitor nor a substrate for thymidine phosphorylase (EC 2.4.2.4), uridine-cytidine kinase (EC 2. 7.1.48), thymidine kinase (EC 2.7.1.21), dihydrouracil dehydrogenase (EC 1.3.1.2), orotate phosphoribosyltransferase (EC 2.4.2.10), or orotidine 5'-monophosphate decarboxylase (EC 4.1.2.23), the enzymes that could utilize the substrate (uridine or thymidine) or products (uracil or thymine) of UrdPase. Different isomers of 5-tolylthiouracil also were synthesized and tested as inhibitors of UrdPase. The meta-substituted isomer was 3- to 4-fold more potent as an inhibitor of UrdPase than the para- or ortho-substituted isomers. These data indicate that the hydrophobic pocket in the active site of UrdPase adjacent to the 5-position of the pyrimidine ring can accommodate the meta-substituted 5-phenyluracils better than the other isomers, leading to improved inhibition. Therefore, it is anticipated that the potency of PTAU can be increased further by the addition of certain hydrophobic groups at the meta position of the phenyl ring. PTAU has potential usefulness in the therapy of cancer and AIDS as well as other pathological and physiological disorders that can be remedied by the administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Tiouracila/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Ligação Competitiva , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Feminino , Humanos , Cinética , Fígado/enzimologia , Camundongos , Tiouracila/análogos & derivados , Tiouracila/síntese química , Tiouracila/química , Tiouracila/farmacocinética
8.
Biochem Pharmacol ; 60(3): 427-31, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10856438

RESUMO

Administration of 200 mg/kg of 5-fluorouracil (FUra) to mice bearing human colon carcinoma DLD-1 xenografts resulted in 100% mortality. Oral administration of 2000 mg/kg of 2',3',5'-tri-O-acetyluridine (TAU), a prodrug of uridine, in combination with 120 mg/kg of 5-(benzyloxybenzyl)barbituric acid acyclonucleoside (BBBA), the most potent known inhibitor of uridine phosphorylase (UrdPase, EC 2.4.2. 3), 2 hr after the administration of the same dose of FUra completely protected the mice (100% survival) from the toxicity of FUra. This combination also reduced tumor weight by 67% compared with 46% achieved by the maximum tolerated dose (50 mg/kg) of FUra alone. Similarly, administration of BBBA plus TAU 1 hr before or 4 hr after the administration of FUra reduced the tumor weight by 53 and 37%, respectively. However, these schedules were less effective in protecting the host from the toxicity of FUra than when the treatment was carried out at 2 hr after FUra administration. TAU alone did not protect from FUra host toxicity. The efficiency of the BBBA plus TAU combination in rescuing from FUra host toxicities is attributed to the exceptional effectiveness of this combination in raising and maintaining higher plasma uridine concentrations than those achieved by TAU alone or by equimolar doses of uridine (Ashour et al., Biochem Pharmacol 51: 1601-1612, 1996). The present results suggest that the BBBA plus TAU combination can provide a better substitute for the massive doses of uridine required to achieve the high levels of uridine necessary to rescue or protect from FUra host toxicities without the toxic side-effects associated with such doses of uridine. The combination of TAU plus BBBA may also allow the escalation of FUra doses for better chemotherapeutic efficacy. Alternatively, the combination may be used as a rescue regimen in the occasional cases where cancer patients receive a lethal overdose of FUra.


Assuntos
Antimetabólitos Antineoplásicos/toxicidade , Barbitúricos/farmacologia , Inibidores Enzimáticos/farmacologia , Fluoruracila/toxicidade , Uridina Fosforilase/antagonistas & inibidores , Uridina/análogos & derivados , Acetatos , Animais , Antimetabólitos Antineoplásicos/uso terapêutico , Barbitúricos/uso terapêutico , Interações Medicamentosas , Feminino , Fluoruracila/uso terapêutico , Humanos , Camundongos , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Pró-Fármacos/farmacologia , Células Tumorais Cultivadas , Uridina/farmacologia
9.
Cancer Chemother Pharmacol ; 45(5): 351-61, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10803917

RESUMO

PURPOSE: The purpose of this investigation was to evaluate the efficacy of oral 5-(phenylselenenyl)-acyclouridine (PSAU) in increasing endogenous plasma uridine concentration as well as its ability to improve the bioavailability of oral uridine. PSAU is a new potent and specific inhibitor of uridine phosphorylase (Urd-Pase, EC 2.4.2.3), the enzyme responsible for uridine catabolism. This compound was designed as a lipophilic inhibitor in order to facilitate its access to the liver and intestine, the main organs involved in uridine catabolism. METHODS: Oral PSAU was administered orally to mice alone or with uridine. The plasma levels of PSAU as well as uridine and its catabolites were measured using high-performance liquid chromatography and pharmacokinetic analysis was performed. RESULTS: PSAU has an oral bioavailability of 100% and no PSAU metabolites were detected. PSAU has no apparent toxicity at high doses. Oral administration of PSAU at 30 and 120 mg/kg increased baseline concentration of endogenous plasma uridine (2.6 +/- 0.7 microM) by 3.2- and 8.7-fold, respectively, and remained three- and six-fold higher, respectively, than the controls for over 8 h. PSAU, however, did not alter the concentration of endogenous plasma uracil. Co-administration of PSAU with uridine elevated the concentration of plasma uridine over that resulting from the administration of either alone, and reduced the peak plasma concentration (C(max)) and area under the curve (AUC) of plasma uracil. Co-administration of PSAU at 30 mg/kg and 120 mg/kg improved the low bioavailability of oral uridine (7.7%) administered at 1,320 mg/kg by 4.8- and 4.2-fold, respectively, and reduced the AUC of plasma uracil from 1,421 to 787 micromol/h x l and 273 micromol/h x l, respectively. Similar results were observed when PSAU was co-administered with lower doses of uridine. Oral PSAU at 30 mg/kg and 120 mg/kg improved the bioavailability of oral 330 mg/kg uridine by 5.2- and 8.9-fold, and that of oral 660 mg/kg uridine by 6.4- and 9.0-fold, respectively. However, the reduction in the AUC values of plasma uracil was less dramatic than that seen when the high dose of 1,320 mg/kg uridine was used. CONCLUSION: The effectiveness of the PSAU plus uridine combination in elevating and sustaining high plasma uridine concentration may be useful to rescue or protect from host toxicity of various chemotherapeutic pyrimidine analogs as well as in the management of medical disorders that are remedied by administration of uridine.


Assuntos
Inibidores Enzimáticos/farmacologia , Compostos Organosselênicos/farmacologia , Uracila/análogos & derivados , Uridina Fosforilase/antagonistas & inibidores , Uridina/sangue , Animais , Disponibilidade Biológica , Feminino , Camundongos , Compostos Organosselênicos/farmacocinética , Uracila/farmacocinética , Uracila/farmacologia
10.
Antimicrob Agents Chemother ; 44(4): 853-8, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10722481

RESUMO

The intracellular metabolism of the beta-L- enantiomer of 2', 3'-dideoxyadenosine (beta-L-ddA) was investigated in HepG2 cells, human peripheral blood mononuclear cells (PBMC), and primary cultured human hepatocytes in an effort to understand the metabolic basis of its limited activity on the replication of human immunodeficiency virus and hepatitis B virus. Incubation of cells with 10 microM [2',3',8-(3)H]-beta-L-ddA resulted in an increased intracellular concentration of beta-L-ddA with time, demonstrating that these cells were able to transport beta-L-ddA. However, it did not result in the phosphorylation of beta-L-ddA to its pharmacologically active 5'-triphosphate (beta-L-ddATP). Five other intracellular metabolites were detected and identified as beta-L-2', 3'-dideoxyribonolactone, hypoxanthine, inosine, ADP, and ATP, with the last being the predominant metabolite, reaching levels as high as 5.14 +/- 0.95, 8.15 +/- 2.64, and 15.60 +/- 1.74 pmol/10(6) cells at 8, 4, and 2 h in HepG2 cells, PBMC, and hepatocytes, respectively. In addition, a beta-glucuronic derivative of beta-L-ddA was detected in cultured hepatocytes, accounting for 12.5% of the total metabolite pool. Coincubation of hepatocytes in primary culture with beta-L-ddA in the presence of increasing concentrations of 5'-methylthioadenosine resulted in decreased phosphorolysis of beta-L-ddA and formation of associated metabolites. These results indicate that the limited antiviral activity of beta-L-ddA is the result of its inadequate phosphorylation to the nucleotide level due to phosphorolysis and catabolism of beta-L-ddA by methylthioadenosine phosphorylase (EC 2.4.2.28).


Assuntos
Antivirais/metabolismo , Didesoxiadenosina/metabolismo , Adenosina/análogos & derivados , Adenosina/farmacologia , Antivirais/farmacologia , Linhagem Celular , Células Cultivadas , Cromatografia Líquida de Alta Pressão , Criopreservação , Didesoxiadenosina/farmacologia , HIV/efeitos dos fármacos , Vírus da Hepatite B/efeitos dos fármacos , Humanos , Fígado/citologia , Fígado/metabolismo , Monócitos/metabolismo , Tionucleosídeos/farmacologia
11.
J Biol Chem ; 274(49): 35255-61, 1999 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-10575012

RESUMO

Purine transport into the protozoan parasite Toxoplasma gondii plays an indispensable nutritional function for this pathogen. To facilitate genetic and biochemical characterization of the adenosine transporter of the parasite, T. gondii tachyzoites were transfected with an insertional mutagenesis vector, and clonal mutants were selected for resistance to the cytotoxic adenosine analog adenine arabinoside (Ara-A). Whereas some Ara-A-resistant clones exhibited disruption of the adenosine kinase (AK) locus, others displayed normal AK activity, suggesting that a second locus had been tagged by the insertional mutagenesis plasmid. These Ara-A(r) AK+ mutants displayed reduced adenosine uptake capability, implying a defect in adenosine transport. Sequences flanking the transgene integration point in one mutant were rescued from a genomic library of Ara-A(r) AK+ DNA, and Southern blot analysis revealed that all Ara-A(r) AK+ mutants were disrupted at the same locus. Probes derived from this locus, designated TgAT, were employed to isolate genomic and cDNA clones from wild-type libraries. Conceptual translation of the TgAT cDNA open reading frame predicts a 462 amino acid protein containing 11 transmembrane domains, a primary structure and membrane topology similar to members of the mammalian equilibrative nucleoside transporter family. Expression of TgAT cRNA in Xenopus laevis oocytes increased adenosine uptake capacity in a saturable manner, with an apparent K(m) value of 114 microM. Uptake was inhibited by various nucleosides, nucleoside analogs, hypoxanthine, guanine, and dipyridamole. The combination of genetic and biochemical studies demonstrates that TgAT is the sole functional adenosine transporter in T. gondii and a rational target for therapeutic intervention.


Assuntos
Adenosina/genética , Adenosina/farmacocinética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Toxoplasma/genética , Toxoplasma/metabolismo , Adenosina Quinase/genética , Sequência de Aminoácidos , Animais , Membrana Celular/metabolismo , Clonagem Molecular , DNA Complementar/metabolismo , Dados de Sequência Molecular , Mutagênese Insercional , Hibridização de Ácido Nucleico , Proteínas Recombinantes/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Fatores de Tempo , Transcrição Gênica , Xenopus
12.
Antimicrob Agents Chemother ; 43(10): 2437-43, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508021

RESUMO

The purine nucleoside analogue NBMPR (nitrobenzylthioinosine or 6-[(4-nitrobenzyl)thio]-9-beta-D-ribofuranosylpurine) was selectively phosphorylated to its nucleoside 5'-monophosphate by Toxoplasma gondii but not mammalian adenosine kinase (EC 2.7.1.20). NBMPR was also cleaved in toxoplasma to its nucleobase, nitrobenzylmercaptopurine. However, nitrobenzylmercaptopurine was not a substrate for either adenosine kinase or hypoxanthine-guanine-xanthine phosphoribosyltransferase (EC 2.4.2.8). Because of this unique and previously unknown metabolism of NBMPR by the parasite, the effect of NBMPR as an antitoxoplasmic agent was tested. NBMPR killed T. gondii grown in human fibroblasts in a dose-dependent manner, with a 50% inhibitory concentration of approximately 10 microM and without apparent toxicity to host cells. Doses of up to 100 microM had no significant toxic effect on uninfected host cells. The promising antitoxoplasmic effect of NBMPR led to the testing of other 6-substituted 9-beta-D-ribofuranosylpurines, which were shown to be good ligands of the parasite adenosine kinase (M. H. Iltzsch, S. S. Uber, K. O. Tankersley, and M. H. el Kouni, Biochem. Pharmacol. 49:1501-1512, 1995), as antitoxoplasmic agents. Among the analogues tested, 6-benzylthioinosine, p-nitrobenzyl-6-selenopurine riboside, N(6)-(p-azidobenzyl)adenosine, and N(6)-(p-nitrobenzyl)adenosine, like NBMPR, were selectively toxic to parasite-infected cells. Thus, it appears that the unique characteristics of purine metabolism in T. gondii render certain 6-substituted 9-beta-D-ribofuranosylpurines promising antitoxoplasmic drugs.


Assuntos
Tioinosina/análogos & derivados , Toxoplasma/metabolismo , Adenosina Quinase/metabolismo , Animais , Antiprotozoários/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Humanos , Nucleotídeos/metabolismo , Fosforilação , Purinas/metabolismo , Tioinosina/metabolismo , Tioinosina/farmacologia , Toxoplasma/citologia , Toxoplasma/efeitos dos fármacos , Toxoplasma/enzimologia
13.
Biochem Pharmacol ; 58(9): 1457-65, 1999 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-10513989

RESUMO

Sixty-seven pyrimidine nucleobase analogues were evaluated as ligands of Toxoplasma gondii orotate phosphoribosyltransferase (OPRTase, EC 2.4.2.10) by measuring their ability to inhibit this enzyme in vitro. Apparent Ki values were determined for compounds that inhibited T. gondii OPRTase by greater than 20% at a concentration of 400 microM. 1-Deazaorotic acid (0.47 microM) and 5-azaorotic acid (2.1 microM) were found to bind better (8.3- and 1.9-fold, respectively) to T. gondii OPRTase than orotic acid, the natural substrate of the enzyme. Based on these results, a structure-activity relationship of ligand binding to OPRTase was formulated using uracil, barbituric acid, and orotic acid as reference compounds. It was concluded that the following structural features of pyrimidine nucleobase analogues were required or strongly preferred for binding: (i) an endocyclic pyridine-type nitrogen or methine at the 1-position; (ii) exocyclic oxo groups at the 2- and 4-positions; (iii) a protonated endocyclic pyridine-type nitrogen at the 3-position; (iv) an endocyclic pyridine-type nitrogen or methine at the 5-position; (v) an exocyclic hydrogen or fluorine at the 5-position; (vi) an endocyclic pyridine-type nitrogen or methine at the 6-position; and (vii) an exocyclic negatively charged or electron-withdrawing group at the 6-position. A comparison of the results from the present study with those from a previous study on mammalian OPRTase [Niedzwicki et al., Biochem Pharmacol 33: 2383-2395, 1984] identified four compounds (6-chlorouracil, 5-azaorotic acid, 1-deazaorotic acid, and 6-iodouracil) that may bind selectively to T. gondii OPRTase.


Assuntos
Orotato Fosforribosiltransferase/metabolismo , Pirimidinas/metabolismo , Toxoplasma/enzimologia , Animais , Sítios de Ligação , Desenho de Fármacos , Estudos de Avaliação como Assunto , Ligantes , Mamíferos , Orotato Fosforribosiltransferase/antagonistas & inibidores , Ácido Orótico/química , Pirimidinas/química , Pirimidinas/farmacologia , Relação Estrutura-Atividade
14.
Mol Biochem Parasitol ; 103(1): 1-14, 1999 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-10514076

RESUMO

A genetic approach has been exploited to investigate adenylate salvage pathways in the protozoan parasite Toxoplasma gondii, a purine auxotroph. Using a new insertional mutagenesis vector designed to facilitate the rescue of tagged loci even when multiple plasmids integrate as a tandem array, 15 independent clonal lines resistant to the toxic nucleoside analog adenine arabinoside (AraA) were generated. Approximately two-thirds of these clones lack adenosine kinase (AK) activity. Parallel studies identified an expressed sequence tag (EST) exhibiting a small region of weak similarity to human AK, and this locus was tagged in several AK-deficient insertional mutants. Library screening yielded full-length cDNA and genomic clones. The T. gondii AK gene contains five exons spanning a approximately 3 kb locus, and the predicted coding sequence was employed to identify additional AK genes and cDNAs in the GenBank and dbEST databases. A genomic construct lacking essential coding sequence was used to create defined genetic knock-outs at the T. gondii AK locus, and AK activity was restored using a cDNA-derived minigene. Hybridization analysis of DNA from 13 AraA-resistant insertional mutants reveals three distinct classes: (i) AK-mutants tagged at the AK locus; (ii) AK- mutants not tagged at the AK locus, suggesting the possibility that another locus may be involved in regulating AK expression; and (iii) mutants with normal AK activity (potential transport mutants).


Assuntos
Adenosina Quinase/genética , Toxoplasma/enzimologia , Toxoplasma/genética , Vidarabina/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Clonagem Molecular , Primers do DNA/genética , DNA Complementar/genética , DNA de Protozoário/genética , Resistência a Medicamentos/genética , Genes de Protozoários , Humanos , Modelos Biológicos , Dados de Sequência Molecular , Mutagênese Insercional , Purinas/metabolismo , Homologia de Sequência de Aminoácidos , Sitios de Sequências Rotuladas , Toxoplasma/efeitos dos fármacos
15.
Bioorg Med Chem Lett ; 9(11): 1477-80, 1999 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-10386920

RESUMO

5-(o-Benzyloxy)benzylbarbituric acid (6) and 5-(p-benzyloxy)benzylbarbituric acid (7) were prepared and their inhibitory activities compared to 5-(m-benzyloxy)-benzylbarbituric acid (BBB) a known, potent inhibitor of uridine phosphorylase (UrdPase). Compounds 6 and 7 were 18-fold and 51-fold less active, respectively, than BBB in inhibiting UrdPase. These data provide solid evidence that the 5-benzylbarbituric acids possessing meta substituents are the most active inhibitors. In addition, 2-thioBBB (11) was synthesized and it was shown to be as active an inhibitor as BBB.


Assuntos
Uracila/análogos & derivados , Uridina Fosforilase/antagonistas & inibidores , Humanos , Cinética , Fígado/enzimologia , Espectroscopia de Ressonância Magnética , Uracila/síntese química , Uracila/química
16.
Biochem Pharmacol ; 51(12): 1601-11, 1996 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-8687475

RESUMO

5-(m-Benzyloxybenzyl)barbituric acid acyclonucleoside (BBBA), the most potent inhibitor known of uridine phosphorylase (UrdPase, EC 2.4.2.3), the enzyme responsible for uridine catabolism, and 2',3',5'-tri-O-acetyluridine (TAU), a prodrug of uridine, were used to investigate the possibility of improving the bioavailability of oral uridine in mice. Oral BBBA administered at 30, 60, 120, and 240 mg/kg increased the concentration of plasma uridine (2.6 +/- 0.7 microM) by 3.2-, 4.6-, 5.4-, and 7.2-fold, respectively. After administration of 120 and 240 mg/kg BBBA, plasma uridine concentration remained 3- and 6-fold, respectively, higher than the plasma concentration at zero time (C0) for over 8 hr. On the other hand, BBBA did not change the concentration of plasma uracil. TAU was far more superior than uridine in improving the bioavailability of plasma uridine. The relative bioavailability of plasma uridine released from oral TAU (53%) was 7-fold higher than that (7.7%) obtained by oral uridine. Oral TAU at 460, 1000, and 2000 mg/kg achieved area under the curve (AUC) values of plasma uridine of 82, 288, and 754 mumol.hr/L, respectively. Coadministration of BBBA with uridine or TAU further improved the bioavailability of plasma uridine resulting from the administration of either alone and reduced the Cmax and AUC of plasma uracil. Coadministration of BBBA at 30, 60, and 120 mg/kg improved the relative bioavailability of uridine released from 2000 mg/kg TAU (53%) by 1.7-, 2.7-, and 3.9-fold, respectively, while coadministration of the same doses of BBBA with an equimolar dose of uridine (1320 mg/kg) increased the relative bioavailability of oral uridine (7.7%) by 4.1-, 5.3-, and 7.8-fold, respectively. Moreover, the AUC and Cmax of plasma uridine after BBBA (120 mg/kg) coadministration with TAU were 3.5- and 11.5-fold, respectively, higher than those obtained from coadministration of BBBA with an equimolar dose of uridine. The exceptional effectiveness of the BBBA plus TAU combination in elevating and sustaining high plasma uridine concentration can be useful in the management of medical disorders that are remedied by administration of uridine as well as to rescue or protect from host-toxicities of various chemotherapeutic pyrimidine analogues.


Assuntos
Barbitúricos/metabolismo , Barbitúricos/farmacologia , Pró-Fármacos/farmacologia , Uridina/urina , Animais , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Feminino , Camundongos , Camundongos Endogâmicos , Fatores de Tempo , Uridina/farmacocinética
17.
Biochem Pharmacol ; 51(12): 1687-700, 1996 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-8687484

RESUMO

One hundred and fifty analogues of uridine, with various modifications to the uracil and pentose moieties, have been tested and compared with uridine with respect to their potency to bind to uridine phosphorylase (UrdPase, EC 2.4.2.3) from Toxoplasma gondii. The effects of the alpha- and beta-anomers, the L- and D-enantiomers, as well as restricted syn and anti rotamers, on binding were examined. Pseudo-, lyxo-, 2,3'-anhydro-2'-deoxy-, 6,5'-cyclo-, 6,3'-methano-, O5',6-methano- and carbocyclic uridines did not bind to the enzyme. Ribosides bound better than the corresponding xylosides, which were better than the deoxyribosides. The binding of deoxyribosides was in the following manner: 2',3'-dideoxynucleosides > 2',5'-dideoxynucleosides > 2'-deoxyribosides > 3'- and 5'-deoxyribosides. alpha-2'-Deoxyribosides bound to the enzyme, albeit less tightly than the corresponding beta-anomers. The acyclo- and 2,2'-anhydrouridines bound strongly, with the 2,2'-anhydro-derivatives being the better ligands. 2,5'-Anhydrouridine bound to UrdPase less effectively than 2,2'-anhydrouridine and acyclouridine. Arabinosyluracil was at best a very poor ligand, but bound better if a benzyl group was present at the 5-position of the pyrimidine ring. This binding was enhanced further by adding a 5-benzyloxybenzyl group. A similar enhancement of the binding by increased hydrophobicity at the 5-position of the pyrimidine ring was observed with ribosides, alpha- and beta-anomers of the 2'-deoxyribosides, acyclonucleosides, and 2,2'-anhydronucleosides. Among all the compounds tested, 5-(benzyloxybenzyl)-2,2'-anhydrouridine was identified as the best ligand of T. gondii UrdPase with an apparent Ki value of 60 +/- 3 nM. It is concluded that the presence of an N-glycosyl bond is a prerequisite for a nucleoside ligand to bind to T. gondii UrdPase. On the other hand, the presence of a 2'-, 3'-, or 5'-hydroxyl group, or an N-glycosyl bond in the beta-configuration, enhanced but was not essential for binding. Furthermore, the potency of the binding of 2,2'-anhydrouridines (fixed high syn isomers) in contrast to the weaker binding of the 6,1'-anhydro- or 2,5'-anhydrouridines (fixed syn isomers), and the complete lack of binding of the 6,5'-cyclo, O5',6-methano- and 6,3'-methanouridines (fixed anti isomers) to T. gondii UrdPase indicate that the binding of ligands to this enzyme is in the syn/high syn conformation around the N-glycosyl bond. The results also indicate that the parasite but not the mammalian host UrdPase can participate in hydrogen bonding with N3 of the pyrimidine ring of nucleoside ligands. T. gondii UrdPase also has a larger hydrophobic pocket adjacent to the C5 of the pyrimidine moiety than the host enzyme, and can accommodate modifications in the pentose moiety which cannot be tolerated by the host enzyme. Most prominent among these modifications is the absence and/or lack of the ribo orientation of the 3'-hydroxyl group, which is a requirement for a ligand to bind to mammalian UrdPase. These differences between the parasite and host, enzymes can be useful in designing specific inhibitors or "subversive" substrates for T. gondii UrdPase.


Assuntos
Relação Estrutura-Atividade , Uridina Fosforilase/efeitos dos fármacos , Uridina Fosforilase/metabolismo , Uridina/análogos & derivados , Uridina/química , Animais , Feminino , Ligantes , Camundongos , Camundongos Endogâmicos , Toxoplasma
18.
Biochem Pharmacol ; 50(10): 1685-93, 1995 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-7503772

RESUMO

Preliminary characterization of Toxoplasma gondii phosphoribosyltransferase activity towards purine nucleobases indicates that there are at least two enzymes present in these parasites. One enzyme uses hypoxanthine, guanine, and xanthine as substrates, while a second enzyme uses only adenine. Furthermore, competition experiments using the four possible substrates suggest that there may be a third enzyme that uses xanthine. Therefore, sixty-eight purine analogues and thirteen related derivatives were evaluated as ligands of T. gondii phosphoribosyltransferase, using xanthine or guanine as substrates, by examining their ability to inhibit these reactions in vitro. Inhibition was quantified by determining apparent Ki values for compounds that inhibited these activities by greater than 10% at a concentration of 0.9 mM. On the basis of these data, a structure-activity relationship for the binding of ligands to these enzymes was formulated using hypoxanthine (6-oxopurine) as a reference compound. It was concluded that the following structural features of purine analogues are required or strongly preferred for binding to both enzymes: (1) a pyrrole-type nitrogen (lactam form) at the 1-position; (2) a methine (= CH-), a pyridine type nitrogen (= N-), or an exocyclic amino or oxo group at the 2-position; (3) no exocyclic substituents at the 3-position; (4) an exocyclic oxo or thio group in the one or thione tautomeric form at the 6-position; (5) a pyridine-type nitrogen (= N-) or a methine group at the 7-position; (6) a methine group at the 8-position; (7) a pyrrole-type nitrogen or a carbon at the 9-position; and (8) no exocyclic substituents at the 9-position. These findings provide the basis for the rational design of additional ligands of hypoxanthine, guanine, and xanthine phosphoribosyltransferase activities in T. gondii.


Assuntos
Compostos Heterocíclicos/metabolismo , Hipoxantina Fosforribosiltransferase/metabolismo , Nitrilas/metabolismo , Pentosiltransferases/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/enzimologia , Animais , Guanina/metabolismo , Compostos Heterocíclicos/farmacologia , Cinética , Ligantes , Nitrilas/farmacologia , Pentosiltransferases/antagonistas & inibidores , Nucleosídeos de Purina/metabolismo , Nucleosídeos de Purina/farmacologia , Relação Estrutura-Atividade , Especificidade por Substrato , Xantina , Xantinas/metabolismo
19.
Biochem Pharmacol ; 49(10): 1501-12, 1995 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-7763293

RESUMO

One hundred and twenty-eight purine nucleoside analogs were evaluated as ligands of Toxoplasma gondii adenosine kinase (EC 2.7.1.20) by examining their ability to inhibit this enzyme in vitro. Inhibition was quantified by determining apparent Ki (appKi) values for those compounds that inhibited this enzyme by greater than 10% at a concentration of 1 mM. Two compounds, N6-(p-methoxybenzoyl)adenosine and 7-iodo-7-deazaadenosine (iodotubercidin), were found to bind to the enzyme (appKi = 3.9 and 1.6 microM, respectively) better than adenosine. On the basis of these data, a structure-activity relationship for the binding of ligands to T. gondii adenosine kinase was formulated using adenosine as a reference compound. It was concluded that the following structural features of purine nucleoside analogs are required or strongly preferred for binding: (1) "pyridine-type" endocyclic nitrogens at the 1- and 3-positions; (2) an exocyclic hydrogen at the 2-position; (3) 6-position exocyclic substituents in the lactim tautomeric form; (4) a "pyridine-type" endocyclic nitrogen at the 7-position or hydrophobic exocyclic substituents attached to an endocyclic carbon at the 7-position; (5) an endocyclic methine or "pyridine-type" nitrogen at the 8-position; (6) an endocyclic nitrogen at the 9-position; (7) a pentose or "pentose-like" (e.g. hydroxylated cyclopentene) moiety attached to the 9-position nitrogen; (8) hydroxyl groups at the 2'- and 3'-positions in a ribose configuration; (9) a hydroxymethyl or methyl (i.e. 5'-deoxy) group at the 5'-position; (10) a beta-D-nucleoside configuration; and (11) an anti conformation around the N-glycosidic bond. In addition, there appears to be a "pocket" in the catalytic site of T. gondii adenosine kinase, adjacent to the 6-position of adenosine, that can accommodate large (preferably unsaturated or aromatic) substituents (e.g. phenyl). These findings provide the basis for the rational design of additional ligands of T. gondii adenosine kinase.


Assuntos
Adenosina Quinase/antagonistas & inibidores , Antiprotozoários/química , Nucleosídeos de Purina/química , Nucleosídeos de Purina/metabolismo , Toxoplasma/enzimologia , Adenosina/análogos & derivados , Adenosina/química , Adenosina Quinase/metabolismo , Animais , Desenho de Fármacos , Ligantes , Nucleosídeos de Purina/farmacologia , Relação Estrutura-Atividade , Toxoplasma/efeitos dos fármacos , Tubercidina/análogos & derivados , Tubercidina/química
20.
Cancer Res ; 55(5): 1092-8, 1995 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-7866994

RESUMO

5-(Benzyloxybenzyl)barbituric acid acyclonucleoside (BBBA) was recently synthesized as a potent and specific inhibitor of uridine phosphorylase (EC 2.4.2.3), the enzyme responsible for the catabolism of 5-fluoro-2'-deoxyuridine (FdUrd) in many types of tumors that are deficient or have little thymidine phosphorylase (EC 2.4.2.4) activity. The effect of BBBA on modulating the antitumor efficacy of FdUrd was evaluated in vitro, against the human colon carcinomas DLD-1 and HCT-15 grown in culture, and in vivo, against DLD-1 grown as xenografts in anti-thymocyte serum immunosuppressed mice. The concentrations of FdUrd that produced 50% growth inhibition after a 3-h exposure were 88 and 340 nM for HCT-15 and DLD-1, respectively. BBBA alone, at all concentrations tested, had no significant effect on the growth of DLD-1 and HCT-15 in culture. However, BBBA at 5, 10, 20, and 40 nM potentiated (P < 0.05) the cytotoxicity of FdUrd (340 nM; 3 h) against DLD-1 in culture by 20, 33, 55, and 63%, respectively. Similarly, BBBA at 10 and 20 nM potentiated the cytotoxicity of FdUrd (88 nM; 3 h) against HCT-15 in culture by 37 and 45%, respectively. In soft agar, BBBA (10 nM) also enhanced the cytocidal effect of FdUrd (10 and 32 nM) against DLD-1 by 41 and 55%, respectively, and against HCT-15 by 6 and 31%, respectively. Increasing BBBA dose to 20 nM enhanced further the FdUrd (10 and 32 nM) cytotoxicity against DLD-1 by 76 and 77%, respectively, and HCT-15 by 31 and 48%, respectively. BBBA also potentiated the chemotherapeutic efficacy of FdUrd in anti-thymocyte serum immunosuppressed mice bearing DLD-1 xenografts with no apparent host toxicity. At a low tumor burden (2.5 x 10(6) cells/mouse), 2 days treatment with FdUrd alone (50 mg/kg/day x 2) did not result in significant reduction in tumor volume. Coadministration of BBBA at 5 and 10 mg/kg/day x 2 did not potentiate the efficacy of FdUrd over that achieved by FdUrd alone, but it significantly reduced the tumor volume by 27 and 32%, respectively, when compared with untreated controls. FdUrd alone at 150 mg/kg/day x 2 reduced the tumor volume by 29%. This reduction in tumor volume was enhanced 1.8-fold by coadministration of BBBA (10 mg/kg/day x 2). At a higher tumor burden (5 x 10(6) cells/mouse) and 4 days treatment, BBBA at 10 and 30 mg/kg/day x 4 reduced further the tumor volume produced by FdUrd alone (200 mg/kg/day x 4) by 1.2- and 1.4-fold, respectively.(ABSTRACT TRUNCATED AT 400 WORDS)


Assuntos
Barbitúricos/farmacologia , Floxuridina/farmacologia , Uridina Fosforilase/antagonistas & inibidores , Animais , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/enzimologia , Sinergismo Farmacológico , Feminino , Floxuridina/metabolismo , Humanos , Terapia de Imunossupressão/métodos , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Transplante Heterólogo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...