Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 160(8): 1916-28, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20649590

RESUMO

BACKGROUND AND PURPOSE: Carboxylesterases (CEs) metabolize a wide range of xenobiotic substrates including heroin, cocaine, meperidine and the anticancer agent CPT-11. In this study, we have purified to homogeneity human liver and intestinal CEs and compared their ability with hydrolyse heroin, cocaine and CPT-11. EXPERIMENTAL APPROACH: The hydrolysis of heroin and cocaine by recombinant human CEs was evaluated and the kinetic parameters determined. In addition, microsomal samples prepared from these tissues were subjected to chromatographic separation, and substrate hydrolysis and amounts of different CEs were determined. KEY RESULTS: In contrast to previous reports, cocaine was not hydrolysed by the human liver CE, hCE1 (CES1), either as highly active recombinant protein or as CEs isolated from human liver or intestinal extracts. These results correlated well with computer-assisted molecular modelling studies that suggested that hydrolysis of cocaine by hCE1 (CES1), would be unlikely to occur. However, cocaine, heroin and CPT-11 were all substrates for the intestinal CE, hiCE (CES2), as determined using both the recombinant protein and the tissue fractions. Again, these data were in agreement with the modelling results. CONCLUSIONS AND IMPLICATIONS: These results indicate that the human liver CE is unlikely to play a role in the metabolism of cocaine and that hydrolysis of this substrate by this class of enzymes is via the human intestinal protein hiCE (CES2). In addition, because no enzyme inhibition is observed at high cocaine concentrations, potentially this route of hydrolysis is important in individuals who overdose on this agent.


Assuntos
Carboxilesterase/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Cocaína/metabolismo , Heroína/metabolismo , Intestinos/enzimologia , Fígado/enzimologia , Camptotecina/análogos & derivados , Camptotecina/química , Camptotecina/metabolismo , Carboxilesterase/química , Carboxilesterase/genética , Carboxilesterase/isolamento & purificação , Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/isolamento & purificação , Cromatografia , Cocaína/química , Heroína/química , Humanos , Hidrólise , Irinotecano , Cinética , Modelos Moleculares , Estrutura Molecular , Conformação Proteica , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
2.
Cancer Gene Ther ; 15(3): 183-92, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18188187

RESUMO

CPT-11 is a potent antitumor agent that is activated by carboxylesterases (CE) and intracellular expression of CEs that can activate the drug results in increased cytotoxicity to the drug. As activation of CPT-11 (irinotecan-7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) by human CEs is relatively inefficient, we have developed enzyme/prodrug therapy approaches based on the CE/CPT-11 combination using a rabbit liver CE (rCE). However, the in vivo application of this technology may be hampered by the development of an immune response to rCE. Therefore, we have developed a mutant human CE (hCE1m6), based on the human liver CE hCE1, that can activate CPT-11 approximately 70-fold more efficiently than the wild-type protein and can be expressed at high levels in mammalian cells. Indeed, adenoviral-mediated delivery of hCE1m6 with human tumor cells resulted in up to a 670-fold reduction in the IC(50) value for CPT-11, as compared to cells transduced with vector control virus. Furthermore, xenograft studies with human tumors expressing hCE1m6 confirm the ability of this enzyme to activate CPT-11 in vivo and induce antitumor activity. We propose that this enzyme should likely be less immunogenic than rCE and would be suitable for the in vivo application of CE/CPT-11 enzyme/prodrug therapy.


Assuntos
Camptotecina/análogos & derivados , Carboxilesterase/genética , Pró-Fármacos/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto , Adenoviridae/genética , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/metabolismo , Antineoplásicos Fitogênicos/uso terapêutico , Western Blotting , Células COS , Camptotecina/metabolismo , Camptotecina/uso terapêutico , Carboxilesterase/química , Carboxilesterase/metabolismo , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Terapia Combinada , Cristalografia por Raios X , Terapia Genética/métodos , Humanos , Irinotecano , Camundongos , Camundongos SCID , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Pró-Fármacos/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Transfecção
3.
Biochem Soc Trans ; 31(Pt 3): 620-4, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12773168

RESUMO

Human carboxylesterase 1 (hCE1) is a serine esterase involved in both drug metabolism and activation, as well as other biological processes. hCE1 catalyses the hydrolysis of heroin and cocaine, and the transesterification of cocaine in the presence of ethanol to the toxic metabolite cocaethylene. We have determined the crystal structures of hCE1 in complex with either the cocaine analogue homatropine or the heroin analogue naloxone. These are the first structures of a human carboxylesterase, and they provide details about narcotic metabolism in humans. hCE1's active site contains rigid and flexible pockets, explaining the enzyme's ability to act both specifically and promiscuously. hCE1 has also been reported to contain cholesteryl ester hydrolase, fatty acyl-CoA hydrolase and acyl-CoA:cholesterol acyltransferase activities, and thus appears to be involved in cholesterol metabolism. Since the enzyme may be useful as a treatment for cocaine overdose, and may afford protection against chemical weapons like Sarin, Soman and VX gas, hCE1 could serve as both a drug and a drug target. Selective hCE1 inhibitors targeted to several sites on the enzyme may also pave the way for novel clinical tools to manage cholesterol homoeostasis in humans.


Assuntos
Hidrolases de Éster Carboxílico/química , Hidrolases de Éster Carboxílico/metabolismo , Desenho de Fármacos , Preparações Farmacêuticas/metabolismo , Cocaína/farmacocinética , Cristalografia por Raios X , Heroína/farmacocinética , Humanos , Modelos Moleculares , Conformação Proteica , Especificidade por Substrato
4.
Mol Pharmacol ; 60(2): 355-62, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11455023

RESUMO

7-Ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin [CPT-11 (irinotecan)] is a water-soluble camptothecin-derived prodrug that is activated by esterases to yield the potent topoisomerase I poison SN-38. We identified a rabbit liver carboxylesterase (CE) that was very efficient at CPT-11 metabolism; however, a human homolog that was more than 81% identical to this protein activated the drug poorly. Recently, two other human CEs have been isolated that are efficient in the conversion of CPT-11 to SN-38, yet both demonstrate little homology to the rabbit protein. To understand this phenomenon, we have characterized a series of esterases from human and rabbit, including several chimeric proteins, for their ability to metabolize CPT-11. Computer predictive modeling indicated that the ability of each enzyme to activate CPT-11 was dependent on the size of the entrance to the active site. Kinetic studies with a series of nitrophenyl and naphthyl esters confirmed these predictions, indicating that activation of CPT-11 by a CE is constrained by size-limited access of the drug to the active site catalytic amino acid residues.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Camptotecina/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Pró-Fármacos/metabolismo , Sequência de Aminoácidos , Animais , Células COS , Camptotecina/análogos & derivados , Camptotecina/química , Hidrolases de Éster Carboxílico/química , Catálise , Esterases/metabolismo , Ésteres/química , Ésteres/metabolismo , Humanos , Irinotecano , Cinética , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Coelhos , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos
5.
Cancer Res ; 61(13): 5078-82, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431344

RESUMO

Irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11) is activated by carboxylesterases (CE) to yield the potent topoisomerase I inhibitor, SN-38. We have demonstrated previously that a rabbit liver CE is approximately 100-1000-fold more efficient at drug activation than a highly homologous human CE. In an attempt to use rabbit CE expression in combination with CPT-11 for gene therapy approaches for the treatment of cancer, we have developed an adenoviral vector expressing this intracellular CE. After transduction, this virus produces very high levels of CE activity in a panel of human tumor cell lines and results in marked sensitization to CPT-11 of all of the transduced cells. Reductions in IC(50) values for this drug ranged from 11-127-fold. Additionally, comparison with an adenovirus expressing a secreted form of the rabbit CE indicated that a collateral effect could be achieved with reductions in the IC(50) values ranging from 4-19-fold. These data suggest that the described reagents may be suitable for use in vivo in a viral-directed enzyme prodrug therapy approach using CPT-11.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/farmacologia , Hidrolases de Éster Carboxílico/genética , Terapia Genética/métodos , Fígado/enzimologia , Adenoviridae/genética , Animais , Antineoplásicos Fitogênicos/farmacocinética , Biotransformação , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Carboxilesterase , Hidrolases de Éster Carboxílico/metabolismo , DNA Complementar/genética , Ensaios de Seleção de Medicamentos Antitumorais , Vetores Genéticos/genética , Humanos , Irinotecano , Coelhos , Rabdomiossarcoma/enzimologia , Rabdomiossarcoma/genética , Rabdomiossarcoma/terapia , Transdução Genética , Células Tumorais Cultivadas
6.
Cancer Res ; 61(13): 5083-9, 2001 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-11431345

RESUMO

Tumor cells that contaminate hematopoietic cell preparations contribute to the relapse of neuroblastoma patients who receive autologous stem cell rescue as a component of therapy. Therefore, effective purging methods are needed. This study details in vitro experiments to develop a viral-directed enzyme prodrug purging method that specifically targets neuroblastoma cells. The approach uses an adenovirus to deliver the cDNA encoding a rabbit liver carboxylesterase that efficiently activates the prodrug irinotecan,7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11). The data show that an adenoviral multiplicity of infection of 50 transduces 100% of cultured neuroblastoma cells and primary tumor cells, irrespective of the level of tumor cell line contamination. Exposure of neuroblastoma cell lines or of mixtures of these cell lines with CD34(+) cells at a ratio of 10:90 to replication-deficient AdRSVrCE for 24 h and subsequent exposure of cells to 1-5 microM CPT-11 for 4 h increased the toxicity of CPT-11 to three neuroblastoma cell lines (SJNB-1, NB-1691, and SK-N-SH) from approximately 20-50-fold and eradicated their clonogenic potential. Also, after "purging," RNA for neuroblastoma cell markers (tyrosine hydroxylase, synaptophysin, and N-MYC) was undetectable by reverse transcription-PCR. In contrast, the purging protocol did not affect the number or type of colonies formed by CD34(+) cells in an in vitro progenitor cell assay. No bystander effect on CD34(+) cells was observed. The method described is being investigated for its potential clinical utility, particularly its efficacy for use with patients having relatively high tumor burdens, because no published methods have been shown to be efficacious when the tumor burden exceeds 1%.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Purging da Medula Óssea/métodos , Camptotecina/farmacologia , Hidrolases de Éster Carboxílico/genética , Terapia Genética , Neuroblastoma/terapia , Pró-Fármacos/farmacologia , Adenoviridae/genética , Adenoviridae/fisiologia , Antígenos CD34/biossíntese , Antineoplásicos Fitogênicos/farmacocinética , Biomarcadores Tumorais/genética , Biotransformação , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Carboxilesterase , Hidrolases de Éster Carboxílico/metabolismo , DNA Complementar/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/virologia , Humanos , Irinotecano , Leucócitos Mononucleares/virologia , Neuroblastoma/genética , Neuroblastoma/patologia , Pró-Fármacos/farmacocinética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução Genética , Células Tumorais Cultivadas
7.
Cancer Res ; 61(7): 3045-52, 2001 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-11306486

RESUMO

One of the advantages of viral-directed enzyme prodrug therapy (VDEPT) is its potential for tumor-specific cytotoxicity. However, the viruses used to deliver cDNAs encoding prodrug-activating enzymes transduce normal cells as well as tumor cells, and several approaches to achieve tumor-specific expression of the delivered cDNAs are being investigated. One such approach is to regulate transcription of the prodrug-activating enzyme with a promoter that is preferentially activated by tumor cells. Published data suggest that the most promising transcription factor/promoter/enhancer combinations are those activated by a tumor-specific transcription factor to retain tumor cell specificity but that are equal in strength to nonspecific viral promoters in their ability to up-regulate target cDNAs. This report identifies MYC-responsive, modified ornithine decarboxylase (ODC) promoter/enhancer sequences that up-regulate target protein expression in tumor cells overexpressing either N-MYC or c-MYC protein. The most efficient of the four constructs assessed contained six additional CACGTG MYC binding sites 5' to the endogenous ODC promoter (R6ODC). Reporter assays with this chimeric promoter/enhancer regulating expression of chloramphenicol acetyltransferase demonstrated 50-250-fold more activity in MYC-expressing cells compared with similar assays with promoterless plasmids. The R6ODC regulatory sequence was approximately equivalent to the CMV promoter in inducing expression of the neomycin resistance gene in c-MYC-expressing SW480 and HT-29 colon carcinoma cells and in N-MYC-expressing NB-1691 neuroblastoma cells. The modified ODC promoter may, therefore, be useful in achieving tissue-specific expression of target proteins in tumor cells that overexpress c- or N-MYC.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Camptotecina/análogos & derivados , Camptotecina/farmacologia , Regulação Neoplásica da Expressão Gênica , Genes myc/genética , Ornitina Descarboxilase/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/biossíntese , Animais , Antineoplásicos Fitogênicos/farmacocinética , Biotransformação , Camptotecina/farmacocinética , Carboxilesterase , Hidrolases de Éster Carboxílico/biossíntese , Hidrolases de Éster Carboxílico/genética , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Genes Reporter , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Immunoblotting , Irinotecano , Proteína MyoD/biossíntese , Proteína MyoD/genética , Neuroblastoma/genética , Neuroblastoma/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Coelhos , Rabdomiossarcoma/genética , Rabdomiossarcoma/metabolismo , Transfecção , Transgenes , Células Tumorais Cultivadas
8.
Cancer Res ; 60(17): 4725-8, 2000 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-10987276

RESUMO

Irinotecan [7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin (CPT-11)] is metabolized by esterases to yield the potent topoisomerase I poison 7-ethyl-10-hydroxycamptothecin. One of the major side effects observed with CPT-11 is gastrointestinal toxicity, and we supposed that this might be due to local activation of CPT-11 within the gut. Carboxylesterase (CE) activity was detected in human gut biopsies, and extracts of these tissues converted CPT-11 to 7-ethyl-10-hydroxycamptothecin in vitro. Expression of a human intestinal CE cDNA in COS-7 cells produced extracts that demonstrated proficient CPT-11 activation and conferred sensitivity of cells to CPT-11. These results suggest that gut toxicity from CPT-11 may be due in part to direct drug conversion by CEs present within the small intestine.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Camptotecina/análogos & derivados , Camptotecina/farmacocinética , Hidrolases de Éster Carboxílico/metabolismo , Intestinos/enzimologia , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/metabolismo , Biotransformação , Células COS/citologia , Células COS/efeitos dos fármacos , Células COS/enzimologia , Camptotecina/metabolismo , Hidrolases de Éster Carboxílico/biossíntese , Hidrolases de Éster Carboxílico/genética , Divisão Celular/efeitos dos fármacos , DNA Complementar/genética , DNA Complementar/isolamento & purificação , DNA Complementar/metabolismo , Humanos , Irinotecano , Fígado/enzimologia , Dados de Sequência Molecular , Coelhos , Homologia de Sequência de Aminoácidos , Transfecção
9.
Mol Ther ; 1(5 Pt 1): 457-63, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10933967

RESUMO

Overexpression of specific transcription factors by tumor cells can be exploited to regulate expression of proteins that induce apoptosis or activate prodrugs, thereby producing tumor-selective toxicity. A majority of advanced-stage neuroblastomas overexpress the transcription factor N-MYC, and this overexpression is associated with poor prognosis. This study describes regulation of expression by N-MYC, via the ornithine decarboxylase (ODC) promoter, of a rabbit liver carboxylesterase (CE) that activates the prodrug CPT-11. Chloramphenicol acetyltransferase reporter assays and CE activity assays in transiently transfected neuroblastoma cell lines (SJNB-1, SJNB-4, NB-1691) and rhabdomyosarcoma cell lines (JR1neo20, JR1Nmyc6, JR1Nmyc9) support this approach as a potential method for sensitizing tumor cells to CPT-11. Clonogenic assays with IMR32 human neuroblastoma cells which express N-MYC and that had been stably transfected with a plasmid containing an ODC promoter/CE cassette corroborated results of enzyme activity assays. Specifically, IMR32.ODC.CE cells expressed approximately eightfold more CE activity than IMR32.CMV.neo cells; and 5 microM CPT-11 reduced the clonogenic potential of IMR32.ODC.CE cells to zero, while 50 microM CPT-11 was required to produce the same effect with IMR32.CMV.neo cells. Current experiments focus on adenoviral delivery of an ODC promoter/CE cDNA cassette for potential virus-directed enzyme prodrug therapy applications.


Assuntos
Camptotecina/análogos & derivados , Camptotecina/farmacologia , Hidrolases de Éster Carboxílico/biossíntese , Neuroblastoma/tratamento farmacológico , Ornitina Descarboxilase/genética , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Northern Blotting , Southern Blotting , Western Blotting , Carboxilesterase , Cloranfenicol O-Acetiltransferase/biossíntese , Relação Dose-Resposta a Droga , Vetores Genéticos , Humanos , Irinotecano , Neuroblastoma/enzimologia , Coelhos , Rabdomiossarcoma/tratamento farmacológico , Rabdomiossarcoma/enzimologia , Transfecção , Células Tumorais Cultivadas
10.
Cancer Res ; 60(15): 4206-10, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10945631

RESUMO

The camptothecin prodrug CPT-11 (irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) is converted by esterases to yield the potent topoisomerase I poison SN-38 (7-ethyl-10-hydroxycamptothecin). Recently, a mouse strain (Es1(e)) has been identified that demonstrates reduced plasma esterase activity, and we have monitored the ability of plasma from these mice to metabolize CPT-11. Total plasma esterase activity was reduced 3-fold in Esl(e)mice in comparison to control mice, and this resulted in a 200-fold reduction in SN-38 production after incubation with CPT-11 in vitro. In addition, pharmacokinetic studies of CPT-11 and SN-38 in these animals demonstrated approximately 5-fold less conversion to SN-38. However, extracts derived from tissues from Es1(e) animals revealed total esterase activities similar to those of control mice, and these extracts metabolized CPT-11 with equal efficiency. Northern analysis of RNA isolated from organs indicated that the liver was the primary source of Es-1 gene expression and that very low levels of Es-1 RNA were present in Es1(e) mice. These results suggest that the reduced levels of Es-1 esterase present in Es1(e) mice are due to down-regulation of gene transcription, and that this plasma esterase is responsible for the majority of CPT-11 metabolism in mice.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Camptotecina/análogos & derivados , Inibidores Enzimáticos/farmacocinética , Esterases/sangue , Pró-Fármacos/farmacocinética , Animais , Antineoplásicos Fitogênicos/sangue , Biotransformação , Camptotecina/sangue , Camptotecina/farmacocinética , Cruzamentos Genéticos , Inibidores Enzimáticos/sangue , Esterases/genética , Expressão Gênica , Irinotecano , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Extratos de Tecidos/metabolismo , Inibidores da Topoisomerase I
11.
Mol Biotechnol ; 15(2): 105-14, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10949823

RESUMO

A series of plasmid vectors have been generated to allow the rapid construction of adenoviral vectors designed to express small RNA sequences. A truncated human U6 gene containing convenient restriction sites has been shown to be expressed at high levels following electroporation into a series of human cell lines. This gene was ligated into a promoterless adenoviral plasmid, and we have generated high titer virus by homologous recombination with adenoviral Addl327 DNA in 293 cells. Recombinant adenovirus containing a hammerhead ribozyme sequence targeted toward the Bcl-2 mRNA has been used to transduce a panel of human tumor cell lines. We have demonstrated high level expression of the recombinant U6 gene containing the ribozyme and reduction of Bcl-2 protein in transduced cells. These plasmids are suitable for the development of adenoviral vectors designed to express both ribozymes and antisense RNA in human cells.


Assuntos
Adenoviridae/genética , DNA Viral/genética , Terapia Genética , Vetores Genéticos , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Catalítico/genética , Western Blotting , Primers do DNA/química , Expressão Gênica , Marcação de Genes , Humanos , Plasmídeos , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Antissenso/farmacologia , RNA Catalítico/metabolismo , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleoproteína Nuclear Pequena U4-U6/genética , Transfecção , Células Tumorais Cultivadas/metabolismo
12.
Biochem Pharmacol ; 59(7): 773-81, 2000 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10718335

RESUMO

Butyrylcholinesterases (BuChEs; acylcholine acylhydrolase; EC 3.1.1.8) have been demonstrated to convert the anticancer agent CPT-11 (irinotecan, 7-ethyl-10-[4-(1-piperidino)-1-piperidino]carbonyloxycamptothecin) into its active metabolite SN-38 (7-ethyl-10-hydroxycamptothecin). In addition, significant differences in the extent of drug metabolism have been observed with BuChEs derived from different species. In an attempt to understand these differences, we have isolated the cDNA encoding a horse BuChE. Based upon the NH2-terminal amino acid sequence of a purified horse BuChE, we designed degenerate primers to amplify the coding sequence from horse liver cDNA. Following polymerase chain reaction and rapid amplification of the cDNA ends, we generated an 1850-bp DNA fragment, containing an 1806-bp open reading frame. The cDNA encodes a protein of 602 amino acid residues, including a 28-amino-acid NH2-terminal signal peptide. Furthermore, the DNA sequence and the deduced amino acid sequence revealed extensive homology to butyrylcholinesterase genes from several other species. In vitro transcription-translation of the cDNA produced a 66-kDa protein, identical to the size of native horse serum BuChE following removal of carbohydrate residues with endoglycosidase F. Additionally, transient expression of the cDNA in Cos-7 cells yielded extracts that exhibited cholinesterase activity and demonstrated a Km value for butyrylthiocholine of 106+/-9 nM. This extract converted the anticancer drug CPT-11 into SN-38, demonstrating that this drug can be activated by enzymes other than carboxylesterases.


Assuntos
Butirilcolinesterase/genética , Fígado/enzimologia , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/metabolismo , Sequência de Bases , Northern Blotting , Butirilcolinesterase/biossíntese , Butirilcolinesterase/metabolismo , Células COS , Camptotecina/análogos & derivados , Camptotecina/metabolismo , DNA Complementar/isolamento & purificação , Cavalos , Humanos , Irinotecano , Dados de Sequência Molecular , Pró-Fármacos/metabolismo , Biossíntese de Proteínas , Homologia de Sequência de Aminoácidos , Transfecção
13.
Mol Biotechnol ; 16(3): 193-202, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11252804

RESUMO

Expression of a rabbit liver carboxylesterase has been achieved in several different model systems including Escherichia coli, Pichia pastoris, Saccharomyces cerevisiae, Spodoptera frugiperda, and COS7 cells. Although, recombinant protein was observed in E. coli sonicates, little or no enzymatic activity was detected. Similarly, no activity was observed following expression in S. cerevisiae. In contrast, active protein was produced in P. pastoris, from S. frugiperda following baculoviral infection and in COS7 cells following transient transfection of plasmid DNA. For the preparation of small amounts of protein for kinetic and biochemical studies, enzyme expressed in P. pastoris has proved sufficient. However, to produce large amounts of carboxylesterase for structural studies, baculoviral-mediated expression of a secreted form of the protein in S. frugiperda was the most efficient. Using this system, we have generated and purified milligram quantities of essentially pure protein. These results demonstrate that the choice of in vitro system for the generation of large amounts of active carboxylesterase, and probably most endoplasmic reticulum processed proteins, is crucial for high level expression and subsequent purification.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Clonagem Molecular/métodos , Pichia/genética , Saccharomyces cerevisiae/genética , Spodoptera/genética , Animais , Baculoviridae/genética , Biotecnologia , Células COS , Hidrolases de Éster Carboxílico/biossíntese , Hidrolases de Éster Carboxílico/isolamento & purificação , Evolução Molecular Direcionada , Expressão Gênica , Vetores Genéticos , Focalização Isoelétrica/métodos , Plasmídeos/genética , Coelhos , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação
14.
Cancer Res ; 59(14): 3424-8, 1999 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-10416605

RESUMO

Water-soluble 20(S)-glycinate esters of two highly potent 10,11-methylenedioxy analogues of camptothecin (CPT) have been synthesized and evaluated for their ability to eradicate human breast cancer tumor xenografts. The glycinate ester moiety increases the water solubility of the 10,11-methylenedioxy analogues 4-16-fold. However, in contrast to CPT-11, a water-soluble CPT analogue that was recently approved for second line treatment of colorectal cancer, the 20(S)-glycinate esters do not require carboxylesterase for conversion to their active forms. The glycinate esters are hydrolyzed to their parent, free 20(S)-hydroxyl active analogues in phosphate buffer (pH 7.5) and in mouse and human plasma. The glycinate esters are also 20-40-fold less potent than CPT-11 in inhibiting human acetylcholinesterase. In vivo, we examined 20(S)-glycinate-10,11-methylenedioxycamptothecin, 20(S)-glycinate-7-chloromethyl-10,11-methylenedioxycamptothecin, and CPT-11. We found that the two 10,11-methylenedioxy analogues had antitumor activity against breast cancer xenografts that was comparable to that of CPT-11. Our results indicate that water-soluble 20(S)-glycinate esters of highly potent CPT analogues provide compounds that maintain biological activity, do not require interactions with carboxylesterases, and do not inhibit human acetylcholinesterase.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Camptotecina/análogos & derivados , Glicina/análogos & derivados , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacocinética , Neoplasias da Mama/patologia , Camptotecina/química , Camptotecina/farmacocinética , Camptotecina/uso terapêutico , Inibidores da Colinesterase/química , Inibidores da Colinesterase/farmacocinética , Inibidores da Colinesterase/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Ésteres , Feminino , Glicina/química , Glicina/farmacocinética , Glicina/uso terapêutico , Humanos , Recém-Nascido , Irinotecano , Camundongos , Camundongos Nus , Estrutura Molecular , Transplante de Neoplasias , Solubilidade , Relação Estrutura-Atividade , Células Tumorais Cultivadas/efeitos dos fármacos
15.
Cancer Res ; 59(7): 1458-63, 1999 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-10197614

RESUMO

Patients treated with high doses of CPT-11 rapidly develop a cholinergic syndrome that can be alleviated by atropine. Although CPT-11 was not a substrate for acetylcholinesterase (AcChE), in vitro assays confirmed that CPT-11 inhibited both human and electric eel AcChE with apparent K(i)s of 415 and 194 nM, respectively. In contrast, human or equine butyryl-cholinesterase (BuChE) converted CPT-11 to SN-38 with K(m)s of 42.4 and 44.2 microM for the human and horse BuChE, respectively. Modeling of CPT-11 within the predicted active site of AcChE and BuChE corroborated experimental results indicating that, although the drug was oriented correctly for activation, the constraints dictated by the active site gorge were such that CPT-11 would be unlikely to be activated by AcChE.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Butirilcolinesterase/metabolismo , Camptotecina/análogos & derivados , Inibidores da Colinesterase/farmacologia , Pró-Fármacos/farmacologia , Animais , Sítios de Ligação , Células COS , Camptotecina/metabolismo , Camptotecina/farmacologia , Enguias , Humanos , Irinotecano , Camundongos , Modelos Moleculares
16.
Clin Cancer Res ; 5(4): 917-24, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10213229

RESUMO

Several recent studies have examined the possibility of producing tumor-specific cytotoxicity with various enzyme/ prodrug combinations. The enzymes are targeted to tumor cells either with antibodies (ADEPT, antibody directed enzyme prodrug therapy) or with viruses (VDEPT). The goal of the present study was to identify an appropriate enzyme for use in activating the prodrug 7-ethyl-10-[4-(1-piper-idino)-1-piperidino]carbonyloxycamptothe cin (CPT-11). In this study, we compared the efficiency of CPT-11 metabolism by rabbit and human carboxylesterases in in vitro and in situ assays. Although the rabbit and human enzymes are very similar (81% identical; 86% homologous) and the active site amino acids are 100% identical, the rabbit enzyme was 100-1000-fold more efficient at converting CPT-11 to SN-38 in vitro and was 12-55-fold more efficient in sensitizing transfected cells to CPT-11. In vivo, Rh30 rhabdomyosarcoma cells expressing the rabbit carboxylesterase and grown as xenografts in immune-deprived mice were also more sensitive to CPT-11 than were control xenografts or xenografts expressing the human enzyme. Each of the three types of xenografts regressed when the mice were treated with CPT-11 given i.v. at 2.5 mg of CPT-11/kg/daily for 5 days/week for 2 weeks [(dx5)2] (one cycle of therapy), repeated every 21 days for a total of three cycles. However, following cessation of treatment, recurrent tumors were detected in seven of seven mice bearing control Rh30 xenografts and in two of seven mice bearing Rh30 xenografts that expressed the human enzyme. No tumors recurred in mice bearing xenografts that expressed the rabbit carboxylesterase. We conclude that rabbit carboxylesterase/CPT-11 may be a useful enzyme/prodrug combination.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Camptotecina/análogos & derivados , Hidrolases de Éster Carboxílico/metabolismo , Neoplasias Experimentais/tratamento farmacológico , Pró-Fármacos/metabolismo , Sequência de Aminoácidos , Animais , Antineoplásicos Fitogênicos/uso terapêutico , Biotransformação , Camptotecina/metabolismo , Camptotecina/uso terapêutico , Carboxilesterase , Hidrolases de Éster Carboxílico/genética , Hidrolases de Éster Carboxílico/uso terapêutico , Catálise , Divisão Celular/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Irinotecano , Camundongos , Camundongos Endogâmicos CBA , Dados de Sequência Molecular , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Nitrobenzenos/metabolismo , Fenilacetatos/metabolismo , Pró-Fármacos/uso terapêutico , Coelhos , Células Tumorais Cultivadas
17.
Clin Cancer Res ; 4(12): 3089-94, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9865925

RESUMO

The anticancer drug CPT-11 (7-ethyl-[4(1-piperidino)-1-piperidino]carbonyloxycamptothecin) is a water-soluble derivative of camptothecin. We report here the conversion of APC (7-ethyl-[4-N-(5-aminopentanoic acid)-1-piperidino] carbonyloxycamptothecin), an inactive metabolite of CPT-11, to SN-38 (7-ethyl-10-hydroxycamptothecin), the active metabolite of CPT-11, by a rabbit liver carboxylesterase. This reaction is not catalyzed by any known human enzyme. The formation of SN-38 from APC was characterized by an apparent Km of 37.9 +/- 7.1 microM and a Vmax of 16.9 +/- 0.9 pmol/units/min. SN-38 was confirmed as a reaction product by high-performance liquid chromatography and mass spectrometry. A 24-h incubation of 10 microM APC with 500 units/ml of rabbit carboxylesterase produced 4 microM SN-38. The product of this reaction inhibited the growth of U373 MG human glioblastoma cells in vitro. The IC50 for a 24-h exposure of U373 MG cells to APC in the presence of 50 units/ml of rabbit carboxylesterase was 0.27 +/- 0.08 microM, whereas APC alone demonstrated no inhibition of growth at concentrations up to 1 microM. The IC50 of U373 MG cells transfected with the cDNA encoding the rabbit carboxylesterase (U373pIRESrabbit) and exposed to APC for 24 h was 0.8 +/- 0.1 microM APC, whereas the growth of cells transfected with vector control (U373pIRES) was unaffected by up to 1 microM APC. Because APC is nontoxic to human cells, we are investigating the possibility of using APC/rabbit carboxylesterase in a prodrug/enzyme therapeutic approach.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Camptotecina/análogos & derivados , Hidrolases de Éster Carboxílico/metabolismo , Fígado/enzimologia , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Camptotecina/química , Camptotecina/metabolismo , Camptotecina/farmacologia , Carboxilesterase , Divisão Celular/efeitos dos fármacos , Meios de Cultura/metabolismo , Humanos , Irinotecano , Espectrometria de Massas , Coelhos , Especificidade por Substrato , Células Tumorais Cultivadas
18.
J Pharmacol Exp Ther ; 286(2): 1066-73, 1998 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9694970

RESUMO

We examined a panel of cell lines for the expression of the myogenic proteins myoD and myogenin. High level expression of both proteins was seen in rhabdomyosarcoma (RMS). To determine whether promoter elements from these genes could direct RMS cell-specific expression, we generated reporter constructs containing one or two copies of the myoD enhancer coupled to the SV40 promoter. Transient transfection reporter assays confirmed the selective expression of beta-galactosidase (beta-gal) in 8 RMS cell lines. In contrast, very low expression from the myoD enhancer/SV40 promoter was detected in four non-RMS cell lines. To determine whether the hybrid promoter could elicit RMS-specific cytotoxicity, a mammalian expression vector containing the herpes simplex virus thymidine kinase (HSVtk) under control of the hybrid myoD enhancer/SV40 promoter was constructed. After electroporation into several cell lines, selective RMS cell kill was observed after treatment with ganciclovir. These data suggest that in vivo tumor-specific expression of HSVtk from the myoD enhancer/SV40 promoter may provide an alternative to current chemotherapy.


Assuntos
Antivirais/farmacologia , Ganciclovir/farmacologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Neoplasias Musculares/enzimologia , Rabdomiossarcoma/enzimologia , Simplexvirus/efeitos dos fármacos , Simplexvirus/enzimologia , Timidina Quinase/biossíntese , Timidina Quinase/genética , Western Blotting , Células Clonais , Galactosidases/genética , Galactosidases/metabolismo , Humanos , Proteína MyoD/genética , Plasmídeos/genética , Reação em Cadeia da Polimerase , Simplexvirus/genética , Transfecção , Células Tumorais Cultivadas
19.
Cancer Res ; 58(16): 3627-32, 1998 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-9721871

RESUMO

Enzyme activation of prodrugs to improve the therapeutic index of specific anticancer agents is an attractive alternative to current chemotherapy regimens. This study addresses the potential for activating irinotecan (CPT-11) with recombinant carboxylesterases (CEs). CEs are a ubiquitous class of enzymes thought to be involved in the detoxification of xenobiotics. Their primary amino acid sequence indicates that these proteins should be localized to the endoplasmic reticulum. By PCR-mediated mutagenesis of a rabbit liver and a human alveolar macrophage CE cDNA, expression in Cos7 cells, and subsequent immunohistochemical localization, we have determined that an 18-amino acid NH2-terminal hydrophobic signal peptide is responsible for the localization of these proteins to the endoplasmic reticulum. By similar approaches, we have demonstrated that the COOH-terminal amino acids HIEL prevent secretion of the proteins from the cell. Enzymatic activity was lost by removing the NH2-terminal domain; however, active enzyme could be detected in the culture media of cells expressing the COOH-terminally truncated proteins. Secretion of CEs lacking the six COOH-terminal amino acids could be prevented with brefeldin A, confirming that these truncated enzymes were processed and released from cells by endoplasmic reticulum-mediated exocytosis. Double-truncation mutant enzymes lacking both NH2- and COOH-terminal sequences demonstrated immunostaining patterns similar to those of the NH2-terminally truncated proteins and also lacked CE activity. In all cases, metabolism of the classic esterase substrate o-nitrophenyl acetate predicted the sensitivity of cells expressing the rabbit CE to the anticancer agent CPT-11. In addition, the secreted enzyme sensitized Cos7 cells to this drug, indicating that protein association with a lipid bilayer is not required for substrate metabolism.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Camptotecina/análogos & derivados , Hidrolases de Éster Carboxílico/análise , Retículo Endoplasmático/enzimologia , Animais , Células COS , Camptotecina/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Meios de Cultura Livres de Soro , Vetores Genéticos , Humanos , Irinotecano , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase/métodos , Coelhos , Especificidade da Espécie , Transfecção
20.
Cytometry ; 32(3): 223-32, 1998 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-9667512

RESUMO

Carboxylesterases are a ubiquitous class of enzymes thought to be involved in xenobiotic metabolism and detoxification. Primary amino acid sequence data suggest that these proteins localize to the endoplasmic reticulum. However, since this family of proteins is highly homologous, the generation of specific reagents to monitor expression and subcellular localization has been unsuccessful. To accomplish in situ detection of a human alveolar macrophage carboxylesterase and a rabbit liver carboxylesterase, we constructed plasmids that expressed recombinant proteins containing an 11 amino acid influenza hemagglutinin tag near the C-terminus. These proteins retained carboxylesterase activity as determined by the conversion of o-nitrophenol acetate to o-nitrophenol. Following transfection of plasmids encoding these proteins into mammalian cells, cells were analyzed by both fluorescence and electron microscopy. The tagged enzymes were localized to the endoplasmic reticulum of both Cos7 monkey kidney cells and Rh30 human rhabdomyosarcoma cells. No tagged protein was detectable in the culture media. Hence, epitope tagging allowed the analysis of expression and localization of specific carboxylesterases. The methods described in this paper are, therefore, applicable to any protein, including those that are highly homologous to other candidate molecules.


Assuntos
Hidrolases de Éster Carboxílico/metabolismo , Epitopos/análise , Proteínas Recombinantes/biossíntese , Sequência de Aminoácidos , Animais , Sequência de Bases , Células COS , Hidrolases de Éster Carboxílico/ultraestrutura , DNA Complementar/biossíntese , DNA Complementar/isolamento & purificação , Epitopos/genética , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Citometria por Imagem , Imuno-Histoquímica , Dados de Sequência Molecular , Coelhos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestrutura , Frações Subcelulares/enzimologia , Frações Subcelulares/ultraestrutura , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA