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 Cancer ; 92(3): 480-7, 2005 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-15668713

RESUMO

The methotrexate (MTX) rescue agent carboxypeptidase G2 (CPDG2) rapidly hydrolyses MTX to the inactive metabolite DAMPA (4-[[2,4-diamino-6-(pteridinyl)methyl]-methylamino]-benzoic acid) and glutamate in patients with MTX-induced renal failure and delayed MTX excretion. DAMPA is thought to be an inactive metabolite of MTX because it is not an effective inhibitor of the MTX target enzyme dihydrofolate reductase. DAMPA is eliminated more rapidly than MTX in these patients, which suggests a nonrenal route of elimination. In a phase II study (May 1997-March 2002), CPDG2 was administered intravenously to 82 patients at a median dose of 50 U kg(-1) (range 33-60 U kg(-1)). Eligible patients for this study had serum MTX concentrations of >10 microM at 36 h or >5 microM at 42 h after start of MTX infusion and documented renal failure (serum creatinine > or =1.5 times the upper limit of normal). Immediately before CPDG2 administration, a median MTX serum level of 11.93 microM (range 0.52-901 microM) was documented. Carboxypeptidase G2 was given at a median of 52 h (range 25-178 h) following the start of an MTX infusion of 1-12 g m(-2) 4-36 h(-1) and resulted in a rapid 97% (range 73-99%) reduction of the MTX serum level. Toxicity related to CPDG2 was not observed. Toxicity related to MTX was documented in about half the patients; four patients died despite CPDG2 administration due to severe myelosuppression and septic complications. In conclusion, administration of CPDG2 is a well-tolerated, safe and a very effective way of MTX elimination in delayed excretion due to renal failure.


Assuntos
Injúria Renal Aguda/induzido quimicamente , Antimetabólitos Antineoplásicos/efeitos adversos , Metotrexato/efeitos adversos , gama-Glutamil Hidrolase/uso terapêutico , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Doenças Hematológicas/induzido quimicamente , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Sepse/induzido quimicamente , Sepse/mortalidade , gama-Glutamil Hidrolase/efeitos adversos
2.
Cancer Res ; 60(15): 4179-86, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10945627

RESUMO

A novel prodrug activation system, endogenous in human tumor cells, is described. A latent enzyme-prodrug system is switched on by a simple synthetic, small molecule co-substrate. This ternary system is inactive if any one of the components is absent. CB 1954 [5-(aziridin-1-yl)-2,4-dinitrobenzamide] is an antitumor prodrug that is activated in certain rat tumors via its 4-hydroxylamine derivative to a potent bifunctional alkylating agent. However, human tumor cells are resistant to CB 1954 because they are unable to catalyze this bioactivation efficiently. A human enzyme has been discovered that can activate CB 1954, and it has been shown to be commonly present in human tumor cells. The enzyme is NQO2 [NAD(P)H quinone oxidoreductase 2], but its activity is normally latent, and a nonbiogenic co-substrate such as NRH [nicotinamide riboside (reduced)] is required for enzymatic activity. There is a very large (100-3000-fold) increase in CB 1954 cytotoxicity toward either NQO2-transfected rodent or nontransfected human tumor cell lines in the presence of NRH. Other reduced pyridinium compounds can also act as co-substrates for NQO2. Thus, the simplest quaternary salt of nicotinamide, 1-methyl-3-carboxamidopyridinium iodide, was a co-substrate for NQO2 when reduced to the corresponding 1,4-dihydropyridine derivative. Increased chain length and/or alkyl load at the 1-position of the dihydropyridine ring improved specific activity, and compounds more active than NRH were found. However, little activity was seen with either the 1-benzyl or 1-(2-phenylethyl) derivatives. A negatively charged substituent at the 3-position of the reduced pyridine ring also negated the ability of these compounds to act as cosubstrates for NQO2. In particular, 1-carbamoylmethyl-3-carbamoyl-1,4dihydropyridine was shown to be a co-substrate for NQO2 with greater stability than NRH, with the ability to enter cells and potentiate the cytotoxicity of CB 1954. Furthermore, this agent is synthetically accessible and suitable for further pharmaceutical development. NQO2 activity appears to be related to expression of NQO1 (DT-diaphorase), an enzyme that is known to have a favorable distribution toward certain human cancers. NQO2 is a novel target for prodrug therapy and has a unique activation mechanism that relies on a synthetic co-substrate to activate an apparently latent enzyme. Our findings may reopen the use of CB 1954 for the direct therapy of human malignant disease.


Assuntos
Antineoplásicos/farmacocinética , Aziridinas/farmacocinética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Pró-Fármacos/farmacocinética , Animais , Antineoplásicos/toxicidade , Aziridinas/toxicidade , Biotransformação , Linhagem Celular , Cricetinae , Cricetulus , Ensaios de Seleção de Medicamentos Antitumorais , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , NAD(P)H Desidrogenase (Quinona)/genética , Niacinamida/análogos & derivados , Niacinamida/metabolismo , Niacinamida/farmacologia , Compostos de Piridínio/metabolismo , Compostos de Piridínio/farmacologia , Especificidade por Substrato , Transfecção , Células Tumorais Cultivadas
3.
Curr Opin Immunol ; 11(5): 579-83, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10508703

RESUMO

Cancer therapy based on the delivery of enzymes to tumour sites has advanced in several directions since antibody-directed enzyme/prodrug therapy was first described. It has been shown that methoxypolyethylene glycol (MPEG) can be used to deliver enzyme to a variety of solid tumours. MPEG-enzyme conjugates show reduced immunogenicity and may allow repeated treatment with enzymes of bacterial origin. Enzyme delivery to tumours by polymers can be used to convert a low toxicity prodrug to a potent cytotoxic agent. An example of such a prodrug is CB1954, which can be activated by a human enzyme in the presence of a cosubstrate. Tumour-located enzymes can also be used in conjunction with a combination of antimetabolites and rescue agents. The rescue agent protects normal tissue but is degraded at cancer sites by the enzyme, thus deprotecting the tumour and allowing prolonged antimetabolite action.


Assuntos
Antineoplásicos/uso terapêutico , Terapia Enzimática , Neoplasias/tratamento farmacológico , Pró-Fármacos/uso terapêutico , Aziridinas/uso terapêutico , Portadores de Fármacos/uso terapêutico , Humanos , Imunoconjugados/uso terapêutico , Polietilenoglicóis/uso terapêutico , Pró-Fármacos/metabolismo , gama-Glutamil Hidrolase/uso terapêutico
4.
Expert Opin Investig Drugs ; 8(2): 161-72, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15992070

RESUMO

Two methods of using tumour located enzymes have been described. These are antibody directed enzyme prodrug therapy (ADEPT) and macromolecule directed enzyme prodrug therapy (MDEPT), where the tumour located enzyme converts a non-toxic prodrug into a cytotoxic drug at tumour sites. The alternative use of tumour located enzymes is to inactivate rescue agents that protect cells from antimetabolite action, and is described as 'Antimetabolite with inactivation of rescue agent at cancer sites' (AMIRACS). The leakiness of tumour blood vessels and poor lymphatic drainage allows enzymes to be targeted to many cancers by attachment to polymeric macromolecules (MDEPT), as well as to antibodies and antibody fragments (ADEPT). To avoid systemic toxicity, enzyme activity in blood and normal tissues must be very low before giving a prodrug or rescue agent. Antibodies directed against the enzyme component of macromolecular conjugates have proved to be very efficient at clearing normal tissues. Human enzymes which are over expressed by cancer cells can be exploited particularly if they require co-factors or co-substrates, either in situ or targeted to extracellular sites. Bacterial enzymes have advantages in specificity but require some form of immunological control in view of their immunogenicity. Prodrugs which generate drugs with very short half lives are desirable, and have been developed, including one which has a differential toxicity between prodrug and the active drug of 1000 to 10,000 fold. The range of antimetabolites available for AMIRACS was initially restricted to inhibitors of dihydrofolate reductase but has been greatly extended by the introduction of inhibitors of other enzymes. The limitations of these systems are discussed.

5.
Cancer Gene Ther ; 4(4): 229-38, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9253508

RESUMO

Expression of genes encoding prodrug-activating enzymes can increase the susceptibility of tumor cells to prodrugs, and may ultimately achieve a better therapeutic index than conventional chemotherapy. CB1954 is a weak, monofunctional alkylating agent which can be activated by Escherichia coli nitroreductase to a potent dysfunctional alkylating agent which crosslinks DNA. We have inserted the nitroreductase gene into an LNCX-based retroviral vector, to allow efficient gene transfer and expression in colorectal (LS174T) and pancreatic (SUIT2, BxPC3, and AsPC1) cancer cell lines. A clone of LS174T cells expressing nitroreductase showed > 50-fold increased sensitivity to CB1954, and nitroreductase-expressing clones of pancreatic tumor lines were up to approximately 500-fold (SUIT2) more sensitive than parental cells. Concentrations of CB1954 minimally toxic to nontransduced cells achieved 100% cell death in a 50:50 mix of parental cells with SUIT2 cells expressing nitroreductase; and marked "bystander" cell killing was seen with just 10% of cells expressing nitroreductase. Significant bystander cell killing was dependent on a high cell density. In conjunction with regional delivery of vectors and tumor selectivity of cell entry and/or gene expression, nitroreductase and CB1954 may be an attractive combination for prodrug-activating enzyme gene therapy of colorectal and pancreatic cancer.


Assuntos
Antineoplásicos/uso terapêutico , Aziridinas/uso terapêutico , Neoplasias Colorretais/terapia , Terapia Genética , Nitrorredutases/genética , Neoplasias Pancreáticas/terapia , Pró-Fármacos/uso terapêutico , Antineoplásicos/farmacologia , Aziridinas/farmacologia , Relação Dose-Resposta a Droga , Escherichia coli/genética , Vetores Genéticos , Humanos , Pró-Fármacos/farmacologia , Retroviridae , Células Tumorais Cultivadas
6.
Structure ; 5(3): 337-47, 1997 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9083113

RESUMO

BACKGROUND: Carboxypeptidase G enzymes hydrolyze the C-terminal glutamate moiety from folic acid and its analogues, such as methotrexate. The enzyme studied here, carboxypeptidase G2 (CPG2), is a dimeric zinc-dependent exopeptidase produced by Pseudomonas sp. strain RS-16. CPG2 has applications in cancer therapy: following its administration as an immunoconjugate, in which CPG2 is linked to an antibody to a tumour-specific antigen, it can enzymatically convert subsequently administered inactive prodrugs to cytotoxic drugs selectively at the tumour site. CPG2 has no significant amino acid sequence homology with proteins of known structure. Hence, structure determination of CPG2 was undertaken to identify active-site residues, which may in turn provide ideas for protein and/or substrate modification with a view to improving its therapeutic usefulness. RESULTS: We have determined the crystal structure of CPG2 at 2.5 A resolution using multiple isomorphous replacement methods and non-crystallographic symmetry averaging. Each subunit of the molecular dimer consists of a larger catalytic domain containing two zinc ions at the active site, and a separate smaller domain that forms the dimer interface. The two active sites in the dimer are more than 60 A apart and are presumed to be independent; each contains a symmetric distribution of carboxylate and histidine ligands around two zinc ions which are 3.3 A apart. This distance is bridged by two shared zinc ligands, an aspartic acid residue and a hydroxyl ion. CONCLUSIONS: We find that the CPG2 catalytic domain has structural homology with other zinc-dependent exopeptidases, both those with a single zinc ion and those with a pair of zinc ions in the active site. The closest structural homology is with the aminopeptidase from Aeromonas proteolytica, where the similarity includes superposable zinc ligands but does not extend to the rest of the active-site residues, consistent with the different substrate specificities. The mechanism of peptide cleavage is likely to be very similar in these two enzymes and may involve the bridging hydroxyl ion ligand acting as a primary nucleophile.


Assuntos
Antineoplásicos/química , Proteínas de Bactérias/química , Conformação Proteica , gama-Glutamil Hidrolase/química , Aminopeptidases/química , Antineoplásicos/uso terapêutico , Proteínas de Bactérias/uso terapêutico , Sítios de Ligação , Catálise , Cristalografia por Raios X , Dimerização , Evolução Molecular , Exopeptidases , Leucil Aminopeptidase/química , Modelos Moleculares , Dados de Sequência Molecular , Peptídeo Hidrolases/química , Peptídeo Hidrolases/classificação , Pseudomonas/enzimologia , Relação Estrutura-Atividade , Zinco/química , gama-Glutamil Hidrolase/uso terapêutico
8.
Gene Ther ; 3(12): 1143-50, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8986441

RESUMO

The most commonly employed 'suicide' gene/prodrug system used in cancer gene therapy is the herpes simplex virus thymidine kinase (HSVtk)/ganciclovir system. We have examined the efficacy of an alternative approach utilising the E. coli nitroreductase B enzyme with CB1954 and a variety of other prodrugs. V79 cells transfected with a nitroreductase expression vector were up to 770-fold more sensitive to CB1954 than control non-expressing cells. In general other prodrugs which were found by HPLC to act as substrates for purified E. coli nitroreductase also exhibited increased cytotoxicity against the nitroreductase-expressing cells, although this correlation was not absolute. In particular nitrofurazone (97-fold) and additional aromatic nitro-compounds (nine- to 50-fold) showed a large differential whereas the quinones and the antimetabolite, B-FU, were less effective (< three-fold). The results support the possibility of using nitroreductase and CB1954 for 'suicide gene' therapy and in addition suggest that alternative prodrugs, such as nitrofurazone, warrant further investigation in this novel approach.


Assuntos
Antineoplásicos/farmacologia , Aziridinas/farmacologia , Escherichia coli/enzimologia , Terapia Genética , Neoplasias/terapia , Nitrorredutases/genética , Pró-Fármacos/farmacologia , Animais , Antineoplásicos/metabolismo , Aziridinas/metabolismo , Biotransformação , Catálise , Linhagem Celular , Cricetinae , Cricetulus , Neoplasias/tratamento farmacológico , Nitrorredutases/metabolismo , Pró-Fármacos/metabolismo , Transfecção
9.
Acta Crystallogr D Biol Crystallogr ; 52(Pt 4): 890-2, 1996 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-15299661

RESUMO

Carboxypeptidase G(2) is a zinc-dependent exopeptidase which has applications in cancer therapy. Crystallization of carboxypeptidase G(2), first achieved more than a decade ago, yields large crystals; however, problems with non-isomorphism between native crystals as well as failure to obtain any useful heavy-atom derivatives have precluded structure solution. A modification of the crystallization protocol leading to a promising new crystal form which diffracts beyond 3.0 A resolution on a rotating-anode source is now reported. These crystals are readily indexed on an apparent C-centred orthorhombic lattice with a = 81.35, b = 230.9 and c = 105.5 A, but the correct crystal system is monoclinic. The crystals have space group P2(1), with a = 81.35, b = 105.5, c = 122.4 A and beta = 109.3 degrees. There are two possible non-equivalent monoclinic indexings with these lattice constants. A partial native data set collected at the SRS, Daresbury, indicates that 1.9 A diffraction is attainable. Structure determination using MIR methods is in progress.

10.
Cancer Res ; 56(14): 3287-92, 1996 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8764123

RESUMO

ZD2767 represents an improved version of antibody-directed enzyme prodrug therapy. It consists of a conjugate of the F(ab')2 A5B7 antibody fragment and carboxypeptidase G2 (CPG2) and a prodrug, 4-[N,N-bis(2-iodoethyl)amino]phenoxycarbonyl L-glutamic acid. The IC50 of the prodrug against LoVo colorectal tumor cells was 47 microM, and cleavage by CPG2 released the potent bis-iodo phenol mustard drug (IC50 = 0.34 microM). The drug killed both proliferating and quiescent LoVo cells. Administration of the ZD2767 conjugate to nude mice bearing LoVo colorectal xenografts resulted in approximately 1% of injected ZD2767 conjugate localizing/g of tumor after 72 h, and blood and normal tissue levels of the conjugate were 10-50-fold lower. A single round of therapy involving the administration of the prodrug 72 h after the conjugate to athymic mice bearing established LoVo xenografts resulted in approximately 50% of the tumors undergoing complete regressions, tumor growth delays greater than 30 days, and little toxicity (as judged by body-weight loss). Similar studies using a control antibody-CPG2 conjugate that does not bind to LoVo tumor cells resulted in a growth delay of less than 5 days, confirming the tumor specificity of this approach. These studies demonstrate the potential of ZD2767 for the treatment of colorectal cancer.


Assuntos
Antineoplásicos Alquilantes/administração & dosagem , Neoplasias Colorretais/tratamento farmacológico , Compostos de Mostarda Nitrogenada/administração & dosagem , Pró-Fármacos/administração & dosagem , Animais , Anticorpos Monoclonais/uso terapêutico , Anticorpos Antineoplásicos/uso terapêutico , Feminino , Humanos , Imunotoxinas/administração & dosagem , Camundongos , Camundongos Nus , Transplante de Neoplasias , Transplante Heterólogo , gama-Glutamil Hidrolase/metabolismo
11.
J Med Chem ; 39(5): 1100-5, 1996 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-8676345

RESUMO

Antibody-directed enzyme prodrug therapy (ADEPT) is a two-step approach for the treatment of cancer which seeks to generate a potent cytotoxic agent selectively at a tumor site. In this work described the cytotoxic agent is generated by the action of an enzyme CPG2 on a relatively nontoxic prodrug. The prodrug 1 currently on clinical trial is a benzamide and is cleaved by CPG2 to a benzoic acid mustard drug 1a. We have synthesized a series of new prodrugs 3-8 where the benzamide link has been replaced by, for example, carbamate or ureido. Some of these alternative links have been shown to be good substrates for CPG2 and therefore new candidates for ADEPT. The active drugs 3a and 4a derived from the best of these prodrugs are potent cytotoxic agents (1-2 microM) some 100 times more than 1a. The prodrugs 3 and 4 are some 100-200-fold less cytotoxic, in a proliferating cell assay, than their corresponding active drugs 3a and 4a.


Assuntos
Mostarda de Anilina/análogos & derivados , Antineoplásicos/síntese química , Imunotoxinas , Pró-Fármacos/síntese química , gama-Glutamil Hidrolase/metabolismo , Mostarda de Anilina/síntese química , Mostarda de Anilina/metabolismo , Mostarda de Anilina/farmacologia , Morte Celular , Divisão Celular/efeitos dos fármacos , Neoplasias Colorretais/patologia , Humanos , Estrutura Molecular , Células Tumorais Cultivadas
12.
Br J Cancer ; 73(5): 565-72, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8605088

RESUMO

Effective radioimmunotherapy is limited by slow antibody clearance from the circulation, which results in low tumour to blood ratios and restricts the dose of radiolabelled anti-tumour antibody that can be safely administrated. Avidin and streptavidin clearing agents have been shown to effectively complex and clear radioactive biotinylated antibodies from the circulation, but their immunogenicity may limit their repeated use. We have investigated whether polyethylene glycol (PEG) modification can reduce the immunogenicity of our galactosylated streptavidin (gal-streptavidin) clearing agent without altering its effectiveness as a clearing agent. The immune response evoked in mice after intraperitoneal infection of 30 micrograms of gal-streptavidin was decreased after PEG modification, as shown by lower antibody titres and a reduction in the number of mice that elicited an anti-gal-streptavidin response. The effect of PEG-modified gal-streptavidin on the blood clearance and tumour localisation of a 125I-labelled biotinylated anti-CEA was investigated in the LS174T human colon carcinoma xenograft in nude mice. Although PEG modified gal-streptavidin bound the [125I]biotinylated antibody in vivo, effective clearance from the circulation was inhibited, resulting in very little reduction in the levels of circulation radioactivity, together with a decrease in the antibody localised to the tumour.


Assuntos
Anticorpos Monoclonais/metabolismo , Anticorpos Antineoplásicos/metabolismo , Proteínas de Bactérias/farmacologia , Polietilenoglicóis/farmacologia , Animais , Anticorpos Monoclonais/farmacocinética , Proteínas de Bactérias/imunologia , Biotina , Cromatografia em Gel , Humanos , Taxa de Depuração Metabólica , Camundongos , Peso Molecular , Radioimunoterapia , Estreptavidina
13.
Immunotechnology ; 2(1): 47-57, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9373327

RESUMO

BACKGROUND: There is considerable interest in the specific targeting of therapeutic agents to cancer cells. Of particular promise is a technique known as Antibody-Directed Enzyme Prodrug Therapy (ADEPT). In this approach an enzyme is targeted to the tumour by its conjugation to a tumour specific-antibody tumour. After allowing sufficient time for the conjugate to localise at the tumour and clear from the circulatory system, a relatively non-toxic prodrug is administered. This prodrug is converted to a highly cytotoxic drug by the action of the targeted enzyme localised at the tumour site. OBJECTIVES: To construct gene fusions between the pseudomonad carboxypeptidase G2 (CPG2) gene and DNA encoding MFE-23 (an anti-carcinoembryonic antigen (CEA) single-chain Fv (scFv) molecule), derived from a phage display library. To overexpress the resultant gene fusions in Escherichia coli, and assess the in vitro and in vivo properties of the purified fusion proteins. STUDY DESIGN: To introduce unique cloning restriction sites into the 5'-end of the CPG2 gene by site-directed mutagenesis to facilitate fusion to the 3'-end of the gene encoding MFE-23 (constructs with or without a flexible (Gly4Ser)3 linker-encoding sequence were designed). To overexpress the resultant gene fusions under transcriptional control of the lac promoter and to direct the fusion proteins produced to the periplasmic space of E. coli through translational coupling to the pelB signal peptide. RESULTS: Biologically active recombinant CPG2::MFE-23 scFv fusion proteins were produced in E. coli and shown to possess enzyme and anti-CEA activity. Affinity chromatography followed by size exclusion gel filtration yielded approximately 0.7-1.4 mg/l from shake flask culture. The fusion protein in which the enzyme and antibody moieties were joined by a linker peptide was shown to be effectively localised in nude mice bearing human colon tumour xenografts, giving favourable tumour to blood ratios. CONCLUSION: MFE-23 scFv serves as an ideal candidate for the antibody arm of a bacterially expressed fusion protein with CPG2. The biological properties of this recombinant protein suggest that it may be employed for tumour specific prodrug activation. However, further assessment of its stability and pharmokinetics is required if genetic fusion is to be considered as an alternative to chemical conjugation.


Assuntos
Antígeno Carcinoembrionário/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , gama-Glutamil Hidrolase/genética , Animais , Fusão Gênica Artificial , Western Blotting , Antígeno Carcinoembrionário/imunologia , Cromatografia de Afinidade , Mapeamento Cromossômico , Clonagem Molecular , Colífagos/genética , Neoplasias Colorretais/metabolismo , Escherichia coli/genética , Expressão Gênica , Biblioteca Gênica , Glicina/genética , Humanos , Óperon Lac , Camundongos , Camundongos Nus , Mutagênese Sítio-Dirigida , Transplante de Neoplasias , Polissacarídeo-Liases/metabolismo , Regiões Promotoras Genéticas , Proteínas Recombinantes de Fusão/imunologia , Serina/genética , Transplante Heterólogo , gama-Glutamil Hidrolase/metabolismo
14.
J Natl Cancer Inst ; 88(3-4): 153-65, 1996 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-8632489

RESUMO

The use of antibody-enzyme conjugates directed at tumor-associated antigens to achieve site-specific activation of prodrugs to potent cytotoxic species, termed "antibody-directed enzyme prodrug therapy" (ADEPT), has attracted considerable interest since the concept was first described in 1987. Prodrug forms of both clinically used anticancer agents and novel cytotoxic compounds have been developed to take advantage of potential prodrug-generating technology employing a variety of enzymes with widely differing substrate specificities. A particular advantage of the ADEPT approach is that it may allow the use of extremely potent agents such as nitrogen mustards and palytoxin, which are too toxic to be readily used in conventional chemotherapy. Preliminary studies using an antibody-enzyme conjugate constructed with a bacterial enzyme and a murine monoclonal antibody not only have established the value of the ADEPT technique, but also have highlighted the potential problem of immunogenicity of proteins of nonhuman origin. This problem has been tackled in the first instance by the use of immunosuppressive agents, but long-term solutions are being investigated in the development of second-generation ADEPT systems, including the development of human antibody-human enzyme fusion proteins and catalytic antibodies. Such improvements, coupled with further refinement of the prodrug-drug element of the system and the wide variety of antibody-enzyme-drug combinations available, should mean that ADEPT-based approaches will form an important element of the search for the anticancer drugs of the future.


Assuntos
Imunoconjugados/uso terapêutico , Pró-Fármacos/administração & dosagem , Fosfatase Alcalina/administração & dosagem , Amidoidrolases/administração & dosagem , Aminopeptidases/administração & dosagem , Carboxipeptidases/administração & dosagem , Glicosídeo Hidrolases/administração & dosagem , Humanos , Nitrorredutases/administração & dosagem , Nucleosídeo Desaminases/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , beta-Lactamases/administração & dosagem
15.
J Med Chem ; 38(26): 5051-65, 1995 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-8544182

RESUMO

Sixteen novel potential prodrugs derived from phenol or aniline mustards and their 16 corresponding drugs with ring substitution and/or different alkylating functionalities were designed. The [[[4-]bis(2-bromoethyl)-(1a), [[[4-[bis(2-iodoethyl)-(1b), and [[[4-[(2-chloroethyl)-[2-(mesyloxy)ethyl]amino]phenyl]oxy] carbonyl]-L-glutamic acids (1c), their [[[2- and 3-substituted-4-[bis(2-chloroethyl)amino]phenyl]oxy]carbonyl]-L- glutamic acids (1e-1), and the [[3-substituted-4-[bis(2-chloroethyl)amino]phenyl]carbamoyl]-L- glutamic acids (1o-r) were synthesized. They are bifunctional alkylating agents in which the activating effect of the phenolic hydroxyl or amino function is masked through an oxycarbonyl or a carbamoyl bond to a glutamic acid. These prodrugs were designed to be activated to their corresponding phenol and aniline nitrogen mustard drugs at a tumor site by prior administration of a monoclonal antibody conjugated to the bacterial enzyme carboxypeptidase G2 (CPG2) in antibody-directed enzyme prodrug therapy (ADEPT). The synthesis of the analogous novel parent drugs (2a-r) is also described. The viability of a colorectal cell line (LoVo) was monitored with the potential prodrugs and the parent drugs. The differential in the cytotoxicity between the potential prodrugs and their corresponding active drugs ranged between 12 and > 195 fold. Compounds 1b-d,f,o exhibited substantial prodrug activity, since a cytotoxicity differential of > 100 was achieved compared to 2b-d,f,o respectively. The ability of the potential prodrugs to act as substrates for CPG2 was determined (kinetic parameters KM and kcat), and the chemical stability was measured for all the compounds. The unsubstituted phenols with different alkylating functionalities (1a-c) proved to have the highest ratio of the substrates kcat:KM. From these studies [[[4-[bis(2-iodoethyl)amino]phenyl]oxy]carbonyl]-L-glutamic acid (1b) emerges as a new ADEPT clinical trial candidate due to its physicochemical and biological characteristics.


Assuntos
Antineoplásicos Alquilantes/síntese química , Compostos de Mostarda Nitrogenada/síntese química , Pró-Fármacos/síntese química , Anticorpos Monoclonais , Antineoplásicos Alquilantes/química , Antineoplásicos Alquilantes/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Humanos , Cinética , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Compostos de Mostarda Nitrogenada/química , Compostos de Mostarda Nitrogenada/farmacologia , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Células Tumorais Cultivadas , gama-Glutamil Hidrolase
16.
Br J Cancer ; 72(5): 1083-8, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7577451

RESUMO

ADEPT is an antibody-based targeting strategy for the treatment of cancer. We have developed two new prodrugs, 4-[N,N-bis(2-chloroethyl)amino]-phenoxycarbonyl-L- glutamic acid (PGP) and (S)-2-[N-[4-[N,N-bis(2-chloroethyl)amino]- phenoxycarbonyl]amino]-4-(5-tetrazoyl)butyric acid (PTP), which are cleaved by the bacterial enzyme CPG2 to release the 4-[N,N-bis(2-chloroethyl)amino] phenol drug. In vitro, both prodrugs are approximately 100- to 200-fold less potent than the parent drug (1 h IC50 = 1.4 microM) in LoVo colorectal tumour cells. These prodrugs have been evaluated for utility in ADEPT when used in combination with a conjugate of CPG2 and the F(ab')2 fragment of the anti-CEA monoclonal antibody, A5B7. The conjugate was shown to localise specifically to established LoVo tumour xenografts growing in nude mice and optimal tumour-normal tissue ratios were achieved after 72 h. Administration of either prodrug, at doses which cause 6-8% body weight loss, 72 h after administration of the A5B7-CPG2 conjugate to the LoVo tumour-bearing mice resulted in tumour regressions and growth delays of 14-28 days. The PTP prodrug in combination with a high dose of conjugate (10 mg kg-1) gave the best anti-tumour activity despite being a 10-fold worse substrate for CPG2 than PGP. Prodrug alone, active drug alone or prodrug in combination with a non-specific conjugate had minimal anti-tumour activity in this tumour model.


Assuntos
Mostarda de Anilina/análogos & derivados , Anticorpos Monoclonais/administração & dosagem , Anticorpos Antineoplásicos/administração & dosagem , Antineoplásicos Alquilantes/uso terapêutico , Proteínas de Bactérias/administração & dosagem , Antígeno Carcinoembrionário/imunologia , Neoplasias Colorretais/tratamento farmacológico , Imunoconjugados/uso terapêutico , Pró-Fármacos/uso terapêutico , gama-Glutamil Hidrolase/administração & dosagem , Mostarda de Anilina/farmacocinética , Mostarda de Anilina/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antineoplásicos/imunologia , Antineoplásicos Alquilantes/farmacocinética , Proteínas de Bactérias/metabolismo , Biotransformação , Neoplasias Colorretais/patologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Fragmentos Fab das Imunoglobulinas/administração & dosagem , Fragmentos Fab das Imunoglobulinas/imunologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Pró-Fármacos/farmacocinética , Pseudomonas/enzimologia , Especificidade por Substrato , Células Tumorais Cultivadas , gama-Glutamil Hidrolase/metabolismo
18.
Biochem Pharmacol ; 50(5): 609-18, 1995 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-7669063

RESUMO

A nitroreductase isolated and purified from Escherichia coli B has been demonstrated to have potential applications in ADEPT (antibody-directed enzyme prodrug therapy) by its ability in vitro to reduce dinitrobenzamides (e.g. 5-aziridinyl 2,4-dinitrobenzamide, CB 1954 and its bischloroethylamino analogue, SN 23862) to form cytotoxic derivatives. In contrast to CB 1954, in which either nitro group is reducible to the corresponding hydroxylamine, SN 23862 is reduced by the nitroreductase to form only the 2-hydroxylamine. This hydroxylamine can react with S-acetylthiocholine to form a species capable of producing interstrand crosslinks in naked DNA. In terms of ADEPT, SN 23862 has a potential advantage over CB 1954 in that it is not reduced by mammalian DT diaphorases. Therefore, a series of compounds related to SN 23862 has been synthesized, and evaluated as potential prodrugs both by determination of kinetic parameters and by ratio of IC50 against UV4 cells when incubated in the presence of prodrug, with and without the E. coli enzyme and cofactor (NADH). Results from the two studies were generally in good agreement in that compounds showing no increase in cytotoxicity in presence of enzyme and cofactor were not substrates for the enzyme. None of the analogues were activated by DT diaphorase isolated from Walker 256 carcinoma cells. For those compounds which were substrates for the E. coli nitroreductase, there was a positive correlation between kcat and IC50 ratio. Two compounds showed advantageous properties: SN 25261 (with a dihydroxypropylcarboxamide ring substituent) which has a more than 10-fold greater aqueous solubility than SN 23862 whilst retaining similar kinetic characteristics and cytotoxic potency; and SN 25084, where a change in the position of the carboxamide group relative to the mustard resulted in an increased cytotoxicity ratio and kcat compared with SN 23862 (IC50 ratios 214 and 135; kcat values of 75 and 26.4 sec-1, respectively). An analogue (SN 25507) incorporating both these structural changes had an enhanced kcat of 576 sec-1. This study elucidates some of the structural requirements of the enzyme and aids identification of further directions in the search for suitable prodrugs for an ADEPT nitroreductase system.


Assuntos
Mostarda de Anilina/análogos & derivados , Antineoplásicos/metabolismo , Aziridinas/metabolismo , Escherichia coli/enzimologia , Nitrorredutases/metabolismo , Pró-Fármacos/metabolismo , Mostarda de Anilina/metabolismo , Mostarda de Anilina/farmacologia , Animais , Antineoplásicos/farmacologia , Aziridinas/farmacologia , Biotransformação , Linhagem Celular , Cricetinae , Cinética , Oxirredução , Pró-Fármacos/farmacologia , Especificidade por Substrato
19.
Biochem Pharmacol ; 49(11): 1641-7, 1995 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-7786305

RESUMO

A nitroreductase enzyme has been isolated from Escherichia coli that has the unusual property of being equally capable of using either NADH or NADPH as a cofactor for the reduction of its substrates which include menadione as well as 5-(aziridin-1-yl)-2,4-dinitrobenzamide (CB 1954). This property is shared with the mammalian enzyme, DT diaphorase. The nitroreductase can, like DT diaphorase, also use simple reduced pyridinium compounds as virtual cofactors. The intact NAD(P)H molecule is not required and the simplest quaternary (and therefore reducible) derivative of nicotinamide, 1-methylnicotinamide (reduced), is as effective as NAD(P)H in its ability to act as an electron donor for the nitroreductase. The structure-activity relationship is not identical to that of DT diaphorase and nicotinic acid riboside (reduced) is selective, being active only for the nitroreductase. Irrespective of the virtual cofactor used, the nitroreductase formed the same reduction products of CB 1954 (the 2- and 4-hydroxylamino derivatives in equal proportions). Nicotinic acid riboside (reduced), unlike NADH, was stable to metabolism by serum enzymes and had a plasma half-life of seven minutes in the mouse after an i.v. bolus administration. NADH had an unmeasurably short half-life. Nicotinic acid riboside (reduced) could also be produced in vivo by administration of nicotinic acid 5'-O-benzoyl riboside (reduced). These results demonstrate that the requirement for a cofactor need not be a limitation in the use of reductive enzymes in antibody directed enzyme prodrug therapy (ADEPT). It is proposed that the E. coli nitroreductase would be a suitable enzyme for ADEPT in combination with CB 1954 and a synthetic, enzyme-selective, virtual cofactor such as nicotinic acid riboside (reduced).


Assuntos
Anticorpos , Aziridinas/metabolismo , Nitrorredutases/metabolismo , Pró-Fármacos , Compostos de Piridínio/farmacocinética , Animais , Escherichia coli/enzimologia , Camundongos , NAD/farmacocinética , Nitrorredutases/isolamento & purificação , Oxirredução , Compostos de Piridínio/síntese química , Ribonucleosídeos/farmacocinética , Vitamina K/metabolismo
20.
Artigo em Inglês | MEDLINE | ID: mdl-9575348

RESUMO

Two antibody fragment-enzyme conjugates, one obtained by random coupling of the two protein component, the other by site-specific ligation of the same component, were compared in vitro and in vivo for their usefulness in antibody directed enzyme prodrug therapy (ADEPT). The in vitro studies have shown that the site-specific conjugate has a higher antigen binding capacity, while both conjugates had similar specific enzymic activities. In vivo, the site-specific conjugate was cleared more rapidly. When correction was made for this faster clearance, both conjugates showed similar antitumor efficacy in a mouse xenograft system upon administration of a prodrug.


Assuntos
Imunoconjugados/química , Fragmentos de Imunoglobulinas/química , gama-Glutamil Hidrolase/química , Animais , Feminino , Humanos , Imunoconjugados/farmacocinética , Imunoconjugados/uso terapêutico , Fragmentos de Imunoglobulinas/metabolismo , Fragmentos de Imunoglobulinas/uso terapêutico , Técnicas In Vitro , Taxa de Depuração Metabólica , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Pró-Fármacos/uso terapêutico , Transplante Heterólogo , Células Tumorais Cultivadas , Redução de Peso/efeitos dos fármacos , gama-Glutamil Hidrolase/farmacocinética , gama-Glutamil Hidrolase/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA